Building Synthetic Cells─From the Technology Infrastructure to Cellular EntitiesClick to copy article linkArticle link copied!
- Lynn J. Rothschild*Lynn J. Rothschild*Email: [email protected]; [email protected]Space Science & Astrobiology Division, NASA Ames Research Center, Moffett Field, California 94035-1000, United StatesDepartment of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island 02912, United StatesMore by Lynn J. Rothschild
- Nils J. H. AvereschNils J. H. AvereschDepartment of Civil and Environmental Engineering, Stanford University, Stanford, California 94305, United StatesMore by Nils J. H. Averesch
- Elizabeth A. StrychalskiElizabeth A. StrychalskiNational Institute of Standards and Technology, Gaithersburg, Maryland 20899, United StatesMore by Elizabeth A. Strychalski
- Felix MoserFelix MoserSynlife, One Kendall Square, Cambridge, Massachusetts 02139-1661, United StatesMore by Felix Moser
- John I. GlassJohn I. GlassJ. Craig Venter Institute, La Jolla, California 92037, United StatesMore by John I. Glass
- Rolando Cruz PerezRolando Cruz PerezDepartment of Bioengineering, Stanford University, Stanford, California 94305, United StatesBlue Marble Space Institute of Science at NASA Ames Research Center, Moffett Field, California 94035-1000, United StatesMore by Rolando Cruz Perez
- Ibrahim O. YekinniIbrahim O. YekinniDepartment of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota 55455, United StatesMore by Ibrahim O. Yekinni
- Brooke Rothschild-MancinelliBrooke Rothschild-MancinelliSchool of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, Georgia 30332-0150, United StatesMore by Brooke Rothschild-Mancinelli
- Garrett A. Roberts KingmanGarrett A. Roberts KingmanNASA Postdoctoral Program, Ames Research Center, Moffett Field, California 94035-1000, United StatesMore by Garrett A. Roberts Kingman
- Feilun Wu
- Jorik WaeterschootJorik WaeterschootMechatronics, Biostatistics and Sensors (MeBioS), KU Leuven, 3000 Leuven BelgiumMore by Jorik Waeterschoot
- Ion A. IoannouIon A. IoannouDepartment of Chemistry, MSRH, Imperial College London, London W12 0BZ, U.K.More by Ion A. Ioannou
- Michael C. JewettMichael C. JewettDepartment of Bioengineering, Stanford University, Stanford, California 94305, United StatesMore by Michael C. Jewett
- Allen P. LiuAllen P. LiuMechanical Engineering & Biomedical Engineering, Cellular and Molecular Biology, Biophysics, Applied Physics, University of Michigan, Ann Arbor, Michigan 48109, United StatesMore by Allen P. Liu
- Vincent NoireauxVincent NoireauxPhysics and Nanotechnology, University of Minnesota, Minneapolis, Minnesota 55455, United StatesMore by Vincent Noireaux
- Carlise SorensonCarlise SorensonDepartment of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota 55455, United StatesMore by Carlise Sorenson
- Katarzyna P. AdamalaKatarzyna P. AdamalaDepartment of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, Minnesota 55455, United StatesMore by Katarzyna P. Adamala
Abstract
The de novo construction of a living organism is a compelling vision. Despite the astonishing technologies developed to modify living cells, building a functioning cell “from scratch” has yet to be accomplished. The pursuit of this goal alone has─and will─yield scientific insights affecting fields as diverse as cell biology, biotechnology, medicine, and astrobiology. Multiple approaches have aimed to create biochemical systems manifesting common characteristics of life, such as compartmentalization, metabolism, and replication and the derived features, evolution, responsiveness to stimuli, and directed movement. Significant achievements in synthesizing each of these criteria have been made, individually and in limited combinations. Here, we review these efforts, distinguish different approaches, and highlight bottlenecks in the current research. We look ahead at what work remains to be accomplished and propose a “roadmap” with key milestones to achieve the vision of building cells from molecular parts.
This publication is licensed under
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
Non-Commercial (NC): Only non-commercial uses of the work are permitted.
No Derivatives (ND): Derivative works may be created for non-commercial purposes, but sharing is prohibited.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
Introduction: What Is a Cell and Why Build One?
What Is a Cell?
Figure 1
Figure 1. Our working definition of living systems, based on Tibor Gánti’s Chemoton model of life. Compartmentalization, replication, and metabolism together make up minimal criteria for a living system. From these other features emerge, including evolution, responsiveness to stimuli, and directed movement.
Why Build a Cell?
Advantages of Synthetic Cells over Cell-Free or Liposome Systems
Figure 2
Figure 2. Four different molecular platforms for studying life. Outgoing from externally provided template DNA (1), a cell-free transcription/translation (Tx/Tl) system uses complex substrates and a chemical energy-donor (2) to synthesize RNA, proteins and potentially other biocatalytically (enzymatically) formed products (3). An encapsulated cell-free system is often a liposome enclosing a cell-free Tx/Tl system. A synthetic cell would function more like a natural cell in that it could be a self-sustaining system that is capable of replication.
cell-free | encapsulated cell-free | synthetic cell | naturally evolved cell | |
---|---|---|---|---|
Defining life/Origin-of-life | Helps understand certain processes | Helps understand a broader range of processes | Ideal to understand the transition from nonliving to living | Noncontroversial example of life |
Alternative cellular systems | Most easily modified for noncanonical components | Noncanonical components functioning in a unit | No constraints of prior evolution, genetics and development | Changes are constrained by evolution, genetics and development |
Studying genes of unknown function | Testing of genes against a defined background | Testing of genes against a defined background | Testing of genes against a defined background | Too many to study; throughput of traditional means too low and slow; reached limit of previous approaches, better to use synthetic biology tools |
Prototyping metabolic pathways | Ideal for simple pathways; no concern about lethality to a cell | Enables compartmentalization for more sophisticated pathways | Most orthogonal while sufficiently capable for more complex pathways | Least orthogonal but most capable for complex pathways |
Molecular biosynthesis development | Testing of metabolite transformation | Testing expansion of metabolite transformation | Integrated multigene pathway testing in controlled/known environment | Analysis of alternative/competing molecular pathways |
Production of toxic compounds | Ideal/unrestricted | Biocontained delivery of biotech | Ideal if not toxic to synthetic cell; potential to study mechanism of toxicity | Prohibited by definition |
Preservation of biodiversity | Understand key metabolic pathways with ecological importance, (e.g., plant–microbe interactions) | Study and engineer key evolutionary and ecological processes (e.g., symbiogenesis) | Study and engineer symbiogenesis, preserve extant and extinct life (e.g., boot-up endangered or extinct genomes), generate new biodiversity | Preserve organism with intrinsic value and natural producer of valuable compound |
Designing environmental stress responses | Protein stability testing | Basic functional tests (e.g., protection of DNA from radiation), subcellular localization and compartmentalization, transport phenomena | Response testing in controlled/known environment | Interaction of stress response with other cellular mechanisms |
Therapeutics production/delivery/testing | Distributed production, logic-gated control | Compartmentalization and targeted delivery, logic-gated control | Allow testing against a known genetic background; could provide orthogonal system for drug delivery | May have superior compatibility with life |
Studying cellular function | Dependent on system used for Tx/Tl | “Toy” system amenable to modeling; reintroducing confinement/organelles/scaffolding in controlled way | Provides modular, defined system | Intact systems for realistic studies |
Evolution studies | Component evolution (e.g., genes) | Component evolution (e.g., transport proteins) | Studies with well-defined cellular systems | Natural evolutionary processes |
Workflows for engineered biological “parts” (modules) | Testing parts in a simplified system toward use in living cells | Testing parts in a simplified system toward use in living cells, with more realistic spatial confinement/biophysics than bulk cell-free | Testing of parts against a defined background | Difficult/impossible/unmet challenge to predict a priori how engineered part will function in vivo |
Is There an Ideal Synthetic Cell?
Strategies for Building a Cell
Figure 3
Figure 3. Major approaches to building a synthetic cell: bottom-up (a), top-down (b), and middle-out (c) are distinguished as the basic concepts of currently ongoing research. Top-down approaches generally seek to find a “minimal genome” of an existing organism, while the bottom-up approach aspires to de novo create a cell “from scratch” or based on macromolecules. Middle-out approaches utilize modules of known function (e.g., organelles, extracts) to assemble a new cell or major elements/mechanisms thereof.
Bottom-up Engineering
Top-down Engineering
Middle-out Engineering
Applications of Synthetic Cells
Understanding the Origin, Evolution, and Diversity of Life
Characterization of Cellular Parts and Elucidation of their Functions
Development of Engineering and Design Tools
Metabolic Engineering and Biomanufacturing
Food Production and Bioremediation
Diagnostics and Therapeutics
Bioprinting
Space Exploration
Research Advancements toward Fulfilling the Criteria of Life
Compartmentalization
Metabolism
Replication
Assembly of Artificial Genomes Based on Natural or Synthetic DNA
Emergent Features of Life
Evolution
Responsiveness
Movement
title | DOI | year | lab | approach | genetic replication | division | compartmentalization | metabolism | movement | sense and respond | evolution |
---|---|---|---|---|---|---|---|---|---|---|---|
Oparin’s Reactions Revisited: Enzymatic Synthesis of Poly(adenylic acid) in Micelles and Self-Reproducing Vesicles | 10.1021/ja00096a010 | 1994 | Luisi | Bottom-up | Yes | Yes | Yes | No | No | No | No |
Man-made cell-like compartments for molecule evolution | 10.1038/nbt0798-652 | 1998 | Griffith | Middle-out | No | No | Yes | No | No | No | No |
A vesicle bioreactor as a step toward an artificial cell assembly | 10.1073/pnas.040823610 | 2004 | Libchaber | Bottom-up | No | No | Yes | Yes | No | No | No |
Design of artificial cell–cell communication using gene and metabolic networks | 10.1073/pnas.0306484101 | 2004 | Liao | Top-down | No | No | Yes | Yes | No | Yes | No |
Self-maintained Movements of Droplets with Convection Flow | 10.1007/978-3-540-76931-6_16 | 2007 | Ikegami | Bottom-up | No | No | No | No | Yes | Yes | No |
Multilevel Selection in Models of Prebiotic Evolution II: A Direct Comparison of Compartmentalization and Spatial Self-Organization | 10.1371/journal.pcbi.1000542 | 2009 | Hogeweg | Top-down | Yes | No | Yes | No | No | No | Yes |
Creation of a bacterial cell controlled by a chemically synthesized genome | 10.1126/science.1190719 | 2010 | Gibson | Top-down | Yes | Yes | No | Yes | No | Yes | Yes |
Self-reproduction of supramolecular giant vesicles combined with the amplification of encapsulated DNA | 10.1038/nchem.1127 | 2011 | Sugawara | Bottom-up | No | Yes | Yes | Yes* | No | No | No |
An Open Question on the Origin of Life: The First Forms of Metabolism | 10.1002/cbdv.201200281 | 2012 | Luisi | Bottom-up | No | No | Yes | Yes | No | No | No |
Spontaneous network formation among cooperative RNA replicators | 10.1038/nature11549 | 2012 | Lehman | Top-down | No | No | No | No | No | Yes | Yes |
Liposome division by a simple bacterial division machinery | 10.1073/pnas.1222254110 | 2013 | Erickson | Bottom-up | No | Yes | Yes | No | No | No | No |
Darwinian evolution in a translation-coupled RNA replication system within a cell-like compartment | 10.1038/ncomms3494 | 2013 | Yomo | Bottom-up | Yes | No | Yes | No | No | No | Yes |
Protein synthesis in artificial cells: using compartmentalization for spatial organization in vesicle bioreactors | 10.1039/C4CP05933F | 2015 | Ces | Bottom-up | No | No | Yes | Yes | No | No | No |
Self Guided Supramolecular Cargo Loaded Nanomotors with Chemotactic Behavior toward Cells | 10.1002/anie.201504186 | 2015 | van Hest and Wilson | Bottom-up | No | No | Yes | No | Yes | No | No |
Design and synthesis of a minimal bacterial genome | 10.1126/science.aad6253 | 2016 | Hutchison | Top-down | Yes | Yes | No | Yes | Yes | Yes | Yes |
Growth and division of active droplets provides a model for protocells | 10.1038/nphys3984 | 2016 | Jülicher | Bottom-up | Yes | Yes | Yes | No | No | No | Yes |
Engineering genetic circuit interactions within and between synthetic minimal cells | 10.1038/nchem.2644 | 2017 | Adamala | Middle-out | No | No | Yes | No | No | Yes | No |
The origin of heredity in protocells | 10.1098/rstb.2016.0419 | 2017 | Lane | Bottom-up | No | Yes | Yes | No | No | No | Yes |
Light-Guided Motility of a Minimal Synthetic Cell | 10.1021/acs.nanolett.8b03469 | 2018 | Wegner | Bottom-up | No | No | Yes | No | Yes | Yes* | No |
Enzyme-powered motility in buoyant organoclay/DNA protocells | 10.1038/s41557-018-0119-3 | 2018 | Mann | Middle-out | No | No | Yes | No | Yes | Yes | No |
Sustainable replication and coevolution of cooperative RNAs in an artificial cell-like system | 10.1038/s41559-018-0650-z | 2018 | Mizuuchi and Ichihashi | Top-down | Yes | Yes | Yes | No | No | No | Yes |
Freeze–thaw cycles induce content exchange between cell-sized lipid vesicles | 10.1088/1367-2630/aabb96 | 2018 | Schwille | Bottom-up | No | No | Yes | No | No | No | No |
Self-replication of DNA by its encoded proteins in liposome-based synthetic cells | 10.1073/pnas.1914656117 | 2018 | Danelon | Bottom-up | Yes | No | Yes | No | No | No | No |
Artificial photosynthetic cell producing energy for protein synthesis | 10.1038/s41467-019-09147-4 | 2019 | Ueda, Kuruma | Bottom-up | No | No | Yes | Yes | No | Yes | No |
Bottom-up Creation of an Artificial Cell Covered with the Adhesive Bacterionanofiber Protein AtaA | 10.1021/jacs.9b09340 | 2019 | Matsuura, Hori | Bottom-up | No | No | Yes | No | No | No | No |
An Adaptive Synthetic Cell Based on Mechanosensing, Biosensing, and Inducible Gene Circuits | 10.1021/acssynbio.9b00204 | 2019 | Noireaux | Middle-out | No | No | Yes | No | No | Yes | No |
Motility of Enzyme-Powered Vesicles | 10.1021/acs.nanolett.9b01830 | 2019 | Sen | Bottom-up | No | No | Yes | No | Yes | No | No |
Mimicking Chemotactic Cell Migration with DNA Programmable Synthetic Vesicles | 10.1021/acs.nanolett.9b04428 | 2019 | Choi | Middle-out? | No | No | Yes | No | Yes | Yes | No |
Chemical Signal Communication between Two Protoorganelles in a Lipid-Based Artificial Cell | 10.1021/acs.analchem.9b01128 | 2019 | Han | Middle-out? | No | No | Yes | No | No | Yes | No |
Bottom-Up Construction of a Minimal System for Cellular Respiration and Energy Regeneration | 10.1021/acssynbio.0c00110 | 2020 | Biner and Hirst | Bottom-up | No | No | Yes | Yes | No | No | No |
Self-division of giant vesicles driven by an internal enzymatic reaction | 10.1039/C9SC05195C | 2020 | Lagzi, Rossi | Bottom-up | No | Yes | Yes | No | No | Yes | No |
Self Propelled PLGA Micromotor with Chemotactic Response to Inflammation | 10.1002/adhm.201901710 | 2020 | Wilson | Bottom-up | No | No | Yes | No | Yes | No | No |
A Step toward Molecular Evolution of RNA: Ribose Binds to Prebiotic Fatty Acid Membranes, and Nucleosides Bind Better than Individual Bases Do | 10.1002/cbic.202000260 | 2020 | Keller | Bottom-up | No | No | Yes | No | No | No | No |
Hydrodynamic accumulation of small molecules and ions into cell-sized liposomes against a concentration gradient | 10.1038/s42004-020-0277-2 | 2020 | Toyota | Bottom-up | No | No | Yes | No | No | No | No |
Dissipative self-assembly, competition and inhibition in a self-reproducing protocell model | 10.1039/D0SC02768E | 2020 | Fletcher | Bottom-up | Yes | No | Yes | No | No | No | No |
Catalytic processing in ruthenium-based polyoxometalate coacervate protocells | 10.1038/s41467-019-13759-1 | 2020 | Mann | Middle-out? | No | No | Yes | No | No | Yes | No |
Membrane molecular crowding enhances MreB polymerization to shape synthetic cells from spheres to rods | 10.1073/pnas.1914656117 | 2020 | Noireaux | Bottom-up | No | No | Yes | Yes | No | No | No |
Signaling and differentiation in emulsion-based multi-compartmentalized in vitro gene circuits | 10.1038/s41557-018-0174-9 | 2020 | Simmel | Bottom-up | No | No | Yes | No | No | Yes | No |
Engineering motile aqueous phase-separated droplets via liposome stabilization | 10.1038/s41467-021-21832-x | 2021 | Ces | Bottom-up | No | No | Yes | No | Yes | No | Nod |
Light-Powered Reactivation of Flagella and Contraction of Microtubule Networks: Toward Building an Artificial Cell | 10.1021/acssynbio.1c00071 | 2021 | Gholami | Middle-out | No | No | Yes | Yes | Yes | Yes | No |
Reconstitution of contractile actomyosin rings in vesicles | 10.1038/s41467-021-22422-7 | 2021 | Schwille | Bottom-up | No | Yes | Yes | No | No | No | No |
Programmable Aggregation of Artificial Cells with DNA Signals | 10.1021/acssynbio.0c00550 | 2021 | Choi | Bottom-up | No | No | Yes | No | No | Yes | No |
Phase Separation and Protein Partitioning in Compartmentalized Cell-Free Expression Reactions | 10.1021/acs.biomac.1c00546 | 2021 | Maeda | Middle-out | No | No | Yes | No | No | No | No |
Light-Triggered Cargo Loading and Division of DNA-Containing Giant Unilamellar Lipid Vesicles | 10.1021/acs.nanolett.1c00822 | 2021 | Göpfrich | Bottom-up | No | Yes | Yes | No | No | No | No |
Chromatophores efficiently promote light-driven ATP synthesis and DNA transcription inside hybrid multicompartment artificial cells | 10.1073/pnas.2012170118 | 2021 | Mavelli | Middle-out | No | No | Yes | Yes | No | Yes | No |
Programmable Fusion and Differentiation of Synthetic Minimal Cells | 10.1021/acssynbio.1c00519 | 2022 | Adamala | Bottom-up | No | No | Yes | Yes | No | No | Yes |
Signal-processing and adaptive prototissue formation in metabolic DNA protocells | 10.1038/s41467-022-31632-6 | 2022 | Walther | Bottom-up | No | No | Yes | No | No | Yes | No |
Living material assembly of bacteriogenic protocells | 10.1038/s41586-022-05223-w | 2022 | Mann | Middle-out | No | No | Yes | Yes | No | Yes | No |
In vitro assembly, positioning and contraction of a division ring in minimal cells | 10.1038/s41467-022-33679-x | 2022 | Schwille | Bottom-up | No | Yes | Yes | No | No | No | No |
Gene silencing and transfection in synthetic cells | 10.1002/bit.28422 | 2023 | Adamala | Bottom-up | No | No | Yes | Yes | No | No | Yes |
Synthesizing a minimal cell with artificial metabolic pathways | 10.1038/s42004-023-00856-y | 2023 | Imai | Middle-out | No | No | Yes | Yes | No | No | No |
Clonal Amplification-Enhanced Gene Expression in Synthetic Vesicles | 10.1021/acssynbio.2c00668 | 2023 | Danelon | Bottom-up | Yes | No | Yes | Yes | No | No | No |
Engineering cellular communication between light-activated synthetic cells and bacteria | 10.1038/s41589-023-01374-7 | 2023 | Booth | Middle-out | No | No | Yes | No | No | Yes | No |
Signal transduction across synthetic cell membranes, with nucleic acid and peptide signals | 10.1016/j.cels.2023.12.008 | 2024 | Adamala | Bottom-up | No | No | Yes | Yes | No | Yes | Yes |
Classification of approach may be ambiguous in certain cases.
Technical Challenges
Steps toward Rational Bottom-up Design and Construction of Living Cells
1. | Accomplish DNA replication in an encapsulated cell-free system | ||||||||||||||||
2. | Develop synthetic translation modules (synthetic ribosome; i.e., ribosomes making ribosomes, and the rest of the translation apparatus) | ||||||||||||||||
3. | Implement cycling/self-replicating Tx/Tl systems | ||||||||||||||||
4. | Achieve autonomous division of
| ||||||||||||||||
5. | Show adaptation/sense response response (adaptive homeostasis) of synthetic biochemical systems | ||||||||||||||||
6. | Revisit the definition of biological life to include synthetic biochemistry |
Figure 4
Figure 4. Milestones toward the de novo design and construction of a synthetic cell. Many of these steps can be performed in parallel with a mixing and matching approach to integration.
Compartmentalization
Metabolism
Transcription and Translation Systems
Replication
Emergent Features
Evolution
Responsiveness
Movement
Concurrent Challenges
Integration with Natural Cells
Integration and Scalability
Cross-Lab and Larger-Scale Coordination
Modular Roadmapping for Individual Components
Biosafety and Regulatory Guidelines
Technology Transfer
Outreach and Education
Accessibility and Equity
Conclusions and Steps Ahead
When and How to Declare Success
Acknowledgments
We are grateful to every current or previous member of the “Build-a-Cell Roadmap Working Group” who drafted or edited this paper but chose not to be coauthors. NSF Research Coordination Network (RCN) funding (award number 1901145) supported this endeavor. In addition, LJR was supported by NASA’s Planetary Science Division Research Program, through the ISFM work package “Evolutionary Processes that Drove the Emergence and Early Distribution of Life” at NASA Ames Research Center. MCJ acknowledges funding from NSF Division of Chemical, Bioengineering, Environmental, and Transport Systems (award number 1936789.). APL acknowledges funding from NSF Division of Emerging Frontiers (award number 1935265).
BSL | BioSafety Levels |
DBTL | Design-Build-Test-Learn |
DIY | do-it-yourself |
GRAA | generally recognized as alive |
iSAT | integrated synthesis, assembly, and translation |
NSAA | nonstandard amino acid |
PCR | polymerase chain reaction |
PURE | protein synthesis using recombinant elements |
RCN | Research Coordination Network |
SOP | standard operating procedure |
STEM | science, technology, engineering and mathematics |
Tx/Tl | transcription/translation |
UBP | unnatural base pair |
XNA | xenonucleic acid. |
References
This article references 222 other publications.
- 1van Stevendaal, M. H. M. E.; Vasiukas, L.; Yewdall, N. A.; Mason, A. F.; van Hest, J. C. M. Engineering of Biocompatible Coacervate-Based Synthetic Cells. ACS Appl. Mater. Interfaces 2021, 13 (7), 7879– 7889, DOI: 10.1021/acsami.0c19052Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXjvFentr8%253D&md5=ee4e6e04fd446421302e5e3d773e6f8cEngineering of Biocompatible Coacervate-Based Synthetic Cellsvan Stevendaal, Marleen H. M. E.; Vasiukas, Laurynas; Yewdall, N. Amy; Mason, Alexander F.; van Hest, Jan C. M.ACS Applied Materials & Interfaces (2021), 13 (7), 7879-7889CODEN: AAMICK; ISSN:1944-8244. (American Chemical Society)Polymer-stabilized complex coacervate microdroplets have emerged as a robust platform for synthetic cell research. Their unique core-shell properties enable the sequestration of high concns. of biol. relevant macromols. and their subsequent release through the semipermeable membrane. These unique properties render the synthetic cell platform highly suitable for a range of biomedical applications, as long as its biocompatibility upon interaction with biol. cells is ensured. The purpose of this study is to investigate how the structure and formulation of these coacervate-based synthetic cells impact the viability of several different cell lines. Through careful examn. of the individual synthetic cell components, it became evident that the presence of free polycation and membrane-forming polymer had to be prevented to ensure cell viability. After closely examg. the structure-toxicity relationship, a set of conditions could be found whereby no detrimental effects were obsd., when the artificial cells were cocultured with RAW264.7 cells. This opens up a range of possibilities to use this modular system for biomedical applications and creates design rules for the next generation of coacervate-based, biomedically relevant particles.
- 2Tsokolov, S. A. Why Is the Definition of Life so Elusive? Epistemological Considerations. Astrobiology 2009, 9 (4), 401– 412, DOI: 10.1089/ast.2007.0201Google ScholarThere is no corresponding record for this reference.
- 3Deplazes-Zemp, A. The Conception of Life in Synthetic Biology. Sci. Eng. Ethics 2012, 18 (4), 757– 774, DOI: 10.1007/s11948-011-9269-zGoogle ScholarThere is no corresponding record for this reference.
- 4Machery, E. Why I Stopped Worrying about the Definition of Life··· and Why You Should as Well. Synthese 2012, 185 (1), 145– 164, DOI: 10.1007/s11229-011-9880-1Google ScholarThere is no corresponding record for this reference.
- 5Bird, A.; Tobin, E. Natural Kinds. In The Stanford Encyclopedia of Philosophy; Zalta, E. N., Nodelman, U., Eds.; Metaphysics Research Lab, Stanford University, 2023.Google ScholarThere is no corresponding record for this reference.
- 6Cleland, C. E.; Chyba, C. F. Defining “Life.. Orig. Life Evol. Biosph. 2002, 32 (4), 387– 393, DOI: 10.1023/A:1020503324273Google ScholarThere is no corresponding record for this reference.
- 7Ganti, T. The Principles of Life; OUP Oxford, 2003.Google ScholarThere is no corresponding record for this reference.
- 8Vibhute, M. A.; Mutschler, H. A Primer on Building Life like Systems. ChemSystemsChem 2023, DOI: 10.1002/syst.202200033Google ScholarThere is no corresponding record for this reference.
- 9Benner, S. A. Defining Life. Astrobiology 2010, 10 (10), 1021– 1030, DOI: 10.1089/ast.2010.0524Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3M%252FkslGktg%253D%253D&md5=e392b1246f1c562063170009c5e89c0eDefining lifeBenner Steven AAstrobiology (2010), 10 (10), 1021-30 ISSN:.Any definition is intricately connected to a theory that gives it meaning. Accordingly, this article discusses various definitions of life held in the astrobiology community by considering their connected "theories of life." These include certain "list" definitions and a popular definition that holds that life is a "self-sustaining chemical system capable of Darwinian evolution." We then act as "anthropologists," studying what scientists do to determine which definition-theories of life they constructively hold as they design missions to seek non-terran life. We also look at how constructive beliefs about biosignatures change as observational data accumulate. And we consider how a definition centered on Darwinian evolution might itself be forced to change as supra-Darwinian species emerge, including in our descendents, and consider the chances of our encountering supra-Darwinian species in our exploration of the Cosmos. Last, we ask what chemical structures might support Darwinian evolution universally; these structures might be universal biosignatures.
- 10Cleland, C. E. The Quest for a Universal Theory of Life: Searching for Life As We Don’t Know It; Cambridge University Press, 2019.Google ScholarThere is no corresponding record for this reference.
- 11Otto, S. An Approach to the De Novo Synthesis of Life. Acc. Chem. Res. 2022, 55 (2), 145– 155, DOI: 10.1021/acs.accounts.1c00534Google ScholarThere is no corresponding record for this reference.
- 12Joyce, G. F. In Vitro Evolution of Nucleic Acids. Curr. Opin. Struct. Biol. 1994, 4, 331– 336, DOI: 10.1016/S0959-440X(94)90100-7Google Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2MXis1SntA%253D%253D&md5=d6ebdd652b2725a3e2e047f84cb5ed54In vitro evolution of nucleic acidsJoyce, Gerald F.Current Opinion in Structural Biology (1994), 4 (3), 331-6CODEN: COSBEF; ISSN:0959-440X.A review with 37 refs. An increasing no. of labs. are applying the principles of Darwinian evolution to large populations of RNA or DNA mols. to obtain compds. that bind a target ligand or catalyze a particular chem. reaction. These mols. may be useful as enzyme inhibitors or diagnostic agents.
- 13Agapakis, C. M. Designing Synthetic Biology. ACS Synth. Biol. 2014, 3 (3), 121– 128, DOI: 10.1021/sb4001068Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhs1KktbrM&md5=40ea81294cd017b2ff94389d5491d8b4Designing Synthetic BiologyAgapakis, Christina M.ACS Synthetic Biology (2014), 3 (3), 121-128CODEN: ASBCD6; ISSN:2161-5063. (American Chemical Society)A review. Synthetic biol. is frequently defined as the application of engineering design principles to biol. Such principles are intended to streamline the practice of biol. engineering, to shorten the time required to design, build, and test synthetic gene networks. This streamlining of iterative design cycles can facilitate the future construction of biol. systems for a range of applications in the prodn. of fuels, foods, materials, and medicines. The promise of these potential applications as well as the emphasis on design has prompted crit. reflection on synthetic biol. from design theorists and practicing designers from many fields, who can bring valuable perspectives to the discipline. While interdisciplinary connections between biologists and engineers have built synthetic biol. via the science and the technol. of biol., interdisciplinary collaboration with artists, designers, and social theorists can provide insight on the connections between technol. and society. Such collaborations can open up new avenues and new principles for research and design, as well as shed new light on the challenging context-dependence-both biol. and social-that face living technologies at many scales. This review is inspired by the session titled Design and Synthetic Biol.: Connecting People and Technol.at Synthetic Biol. 6.0 and covers a range of literature on design practice in synthetic biol. and beyond. Crit. engagement with how design is used to shape the discipline opens up new possibilities for how we might design the future of synthetic biol.
- 14Malaterre, C. Can Synthetic Biology Shed Light on the Origins of Life?. Biol. Theory 2009, 4 (4), 357– 367, DOI: 10.1162/BIOT_a_00002Google ScholarThere is no corresponding record for this reference.
- 15Mukherji, S.; van Oudenaarden, A. Synthetic Biology: Understanding Biological Design from Synthetic Circuits. Nat. Rev. Genet. 2009, 10 (12), 859– 871, DOI: 10.1038/nrg2697Google Scholar15https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhsVaksL%252FO&md5=27ea5f286f69609af97759d5e3effb1bSynthetic biology: understanding biological design from synthetic circuitsMukherji, Shankar; van Oudenaarden, AlexanderNature Reviews Genetics (2009), 10 (12), 859-871CODEN: NRGAAM; ISSN:1471-0056. (Nature Publishing Group)The article highlights how the process of engineering biol. systems has contributed to our understanding of how endogenous systems are put together and function - from a quant. description of gene expression and signal transduction to controlling spatial organization and cell-cell interactions. An important aim of synthetic biol. is to uncover the design principles of natural biol. systems through the rational design of gene and protein circuits. Here, we highlight how the process of engineering biol. systems - from synthetic promoters to the control of cell-cell interactions - has contributed to our understanding of how endogenous systems are put together and function. Synthetic biol. devices allow us to grasp intuitively the ranges of behavior generated by simple biol. circuits, such as linear cascades and interlocking feedback loops, as well as to exert control over natural processes, such as gene expression and population dynamics.
- 16Rothschild, L. J. The Role of Emergence in Biology; Oxford University Press, 2008.Google ScholarThere is no corresponding record for this reference.
- 17Adamala, K.; Szostak, J. W. Nonenzymatic Template-Directed RNA Synthesis inside Model Protocells. Science 2013, 342 (6162), 1098– 1100, DOI: 10.1126/science.1241888Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhvVartr3K&md5=47b93286cfc8db8306f03be866470a70Nonenzymatic Template-Directed RNA Synthesis Inside Model ProtocellsAdamala, Katarzyna; Szostak, Jack W.Science (Washington, DC, United States) (2013), 342 (6162), 1098-1100CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Efforts to recreate a prebiotically plausible protocell, in which RNA replication occurs within a fatty acid vesicle, have been stalled by the destabilizing effect of Mg2+ on fatty acid membranes. Here we report that the presence of citrate protects fatty acid membranes from the disruptive effects of high Mg2+ ion concns. while allowing RNA copying to proceed, while also protecting single-stranded RNA from Mg2+-catalyzed degrdn. This combination of properties has allowed us to demonstrate the chem. copying of RNA templates inside fatty acid vesicles, which in turn allows for an increase in copying efficiency by bathing the vesicles in a continuously refreshed soln. of activated nucleotides.
- 18Deamer, D.; Dworkin, J. P.; Sandford, S. A.; Bernstein, M. P.; Allamandola, L. J. The First Cell Membranes. Astrobiology 2002, 2 (4), 371– 381, DOI: 10.1089/153110702762470482Google Scholar18https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXhtVamur4%253D&md5=263bd36bf3d352bb062f2d04c0406f6aThe first cell membranesDeamer, David; Dworkin, Jason P.; Sandford, Scott A.; Bernstein, Max P.; Allamandola, Louis J.Astrobiology (2002), 2 (4), 371-381CODEN: ASTRC4; ISSN:1531-1074. (Mary Ann Liebert, Inc.)A review. Org. compds. are synthesized in the interstellar medium and can be delivered to planetary surfaces such as the early Earth, where they mix with endogenous species. Some of these compds. are amphiphilic, having polar and nonpolar groups on the same mol. Amphiphilic compds. spontaneously self-assemble into more complex structures such as bimol. layers, which in turn form closed membranous vesicles. The 1st forms of cellular life required self-assembled membranes that were likely to have been produced from amphiphilic compds. on the prebiotic Earth. Lab. simulations show that such vesicles readily encapsulate functional macromols., including nucleic acids and polymerases. The goal of future investigations will be to fabricate artificial cells as models of the origin of life.
- 19Deamer, D. The Role of Lipid Membranes in Life’s Origin. Life 2017, 7 (1), 5, DOI: 10.3390/life7010005Google ScholarThere is no corresponding record for this reference.
- 20Discher, B. M.; Bermudez, H.; Hammer, D. A.; Discher, D. E.; Won, Y.-Y.; Bates, F. S. Cross-Linked Polymersome Membranes: Vesicles with Broadly Adjustable Properties. J. Phys. Chem. B 2002, 106 (11), 2848– 2854, DOI: 10.1021/jp011958zGoogle Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XhsVGjt7s%253D&md5=66052831fe2660d1457252d266f0d7ffCross-linked Polymersome Membranes: Vesicles with Broadly Adjustable PropertiesDischer, Bohdana M.; Bermudez, Harry; Hammer, Daniel A.; Discher, Dennis E.; Won, You-Yeon; Bates, Frank S.Journal of Physical Chemistry B (2002), 106 (11), 2848-2854CODEN: JPCBFK; ISSN:1089-5647. (American Chemical Society)Massively cross-linked and property-tunable membranes have been fabricated by free radical polymn. of self-assembled, block copolymer vesicles - polymersomes. Similar efforts with cross-linkable lipids would appear frustrated in the past due to at least two factors: limited reactivity and membrane fragility under local stresses of nano-confined crosslinking. We describe here a diblock copolymer of poly(ethylene oxide)-polybutadiene that has a hydrophilic wt. fraction like that of lipids and forms robust fluid phase membranes in water. The polymersomes sustain free radical polymn. of the hydrophobic butadiene, thereby generating a semipermeable nano-shell. Cross-linked giant vesicles prove stable in chloroform and can also be dehydrated and re-hydrated without rendering the ∼9 nm thick membrane core; the results imply defect-free membranes many microns-squared in area. Surface elastic moduli as well as sustainable wall stresses up to 103 Atm, orders of magnitude greater than any natural lipid membrane, appear consistent with strong tethering between close-packed neighbors. The enormous stability of the giant vesicles can be tuned down for application: blending in the hydrogenated analog poly(ethylene oxide)-polyethylethylene modulates the effective elastic consts. as well as the rupture strength by orders of magnitude. The results appear consistent with rigidity percolation through a finite-layer stack of two-dimensional lattices. Moreover, below the percolation limit, a regime of hyper-instability emerges, reflecting perhaps nanoscale demixing and suggestive of the limitations encountered with low reactivity lipids. The results provide general insights into covalent crosslinking within self-assembled nanostructures.
- 21Kamat, N. P.; Katz, J. S.; Hammer, D. A. Engineering Polymersome Protocells. J. Phys. Chem. Lett. 2011, 2 (13), 1612– 1623, DOI: 10.1021/jz200640xGoogle Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXnsFWjtLs%253D&md5=3c4972f113a5618037a2c216667ba743Engineering Polymersome ProtocellsKamat, Neha P.; Katz, Joshua S.; Hammer, Daniel A.Journal of Physical Chemistry Letters (2011), 2 (13), 1612-1623CODEN: JPCLCD; ISSN:1948-7185. (American Chemical Society)A review. The field of biomimicry is embracing the construction of complex assemblies that imitate both biol. structure and function. Advancements in the design of these mimetics have generated a growing vision for creating an artificial cell or protocell. Polymersomes are vesicles that can be made from synthetic, biol., or hybrid polymers and can be used as a model template to build cell-like structures. In this perspective, we discuss various areas where polymersomes have been used to mimic cell functions as well as areas in which the synthetic flexibility of polymersomes would make them ideal candidates for a biomembrane mimetic. Designing a polymersome that comprehensively displays the behaviors discussed herein has the potential to lead to the development of an autonomous, responsive particle that resembles the intelligence of a biol. cell.
- 22Vickers, C. E. The Minimal Genome Comes of Age. Nat. Biotechnol. 2016, 34 (6), 623– 624, DOI: 10.1038/nbt.3593Google ScholarThere is no corresponding record for this reference.
- 23Xu, X.; Meier, F.; Blount, B. A.; Pretorius, I. S.; Ellis, T.; Paulsen, I. T.; Williams, T. C. Trimming the Genomic Fat: Minimising and Re-Functionalising Genomes Using Synthetic Biology. Nat. Commun. 2023, 14 (1), 1984, DOI: 10.1038/s41467-023-37748-7Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXnslWksb0%253D&md5=44b6c482b42a43dbcf6a77510da09105Trimming the genomic fat: minimising and re-functionalising genomes using synthetic biologyXu, Xin; Meier, Felix; Blount, Benjamin A.; Pretorius, Isak S.; Ellis, Tom; Paulsen, Ian T.; Williams, Thomas C.Nature Communications (2023), 14 (1), 1984CODEN: NCAOBW; ISSN:2041-1723. (Nature Portfolio)A review. Naturally evolved organisms typically have large genomes that enable their survival and growth under various conditions. However, the complexity of genomes often precludes our complete understanding of them, and limits the success of biotechnol. designs. In contrast, minimal genomes have reduced complexity and therefore improved engineerability, increased biosynthetic capacity through the removal of unnecessary genetic elements, and less recalcitrance to complete characterization. Here, we review the past and current genome minimisation and re-functionalisation efforts, with an emphasis on the latest advances facilitated by synthetic genomics, and provide a crit. appraisal of their potential for industrial applications.
- 24Hutchison, C. A.; Peterson, S. N.; Gill, S. R.; Cline, R. T.; White, O.; Fraser, C. M.; Smith, H. O.; Venter, J. C. Global Transposon Mutagenesis and a Minimal Mycoplasma Genome. Science 1999, 286 (5447), 2165– 2169, DOI: 10.1126/science.286.5447.2165Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1MXotFGru70%253D&md5=863f5f2e7a6dcefb45cd5765e0f892e8Global transposon mutagenesis and a minimal mycoplasma genomeHutchison, Clyde A., III; Peterson, Scott N.; Gill, Steven R.; Cline, Robin T.; White, Owen; Fraser, Claire M.; Smith, Hamilton O.; Venter, J. CraigScience (Washington, D. C.) (1999), 286 (5447), 2165-2169CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Mycoplasma genitalium with 517 genes has the smallest gene complement of any independently replicating cell so far identified. Global transposon mutagenesis was used to identify nonessential genes in an effort to learn whether the naturally occurring gene complement is a true minimal genome under lab. growth conditions. The positions of 2209 transposon insertions in the completely sequenced genomes of M. genitalium and its close relative M. pneumoniae were detd. by sequencing across the junction of the transposon and the genomic DNA. These junctions defined 1354 distinct sites of insertion that were not lethal. The anal. suggests that 265 to 350 of the 480 protein-coding genes of M. genitalium are essential under lab. growth conditions, including about 100 genes of unknown function.
- 25Glass, J. I.; Assad-Garcia, N.; Alperovich, N.; Yooseph, S.; Lewis, M. R.; Maruf, M.; Hutchison, C. A., 3rd; Smith, H. O.; Venter, J. C. Essential Genes of a Minimal Bacterium. Proc. Natl. Acad. Sci. U. S. A. 2006, 103 (2), 425– 430, DOI: 10.1073/pnas.0510013103Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XpsVSltA%253D%253D&md5=a4eabb14b59f1b81454494b19cf287a4Essential genes of a minimal bacteriumGlass, John I.; Assad-Garcia, Nacyra; Alperovich, Nina; Yooseph, Shibu; Lewis, Matthew R.; Maruf, Mahir; Hutchison, Clyde A., III; Smith, Hamilton O.; Venter, J. CraigProceedings of the National Academy of Sciences of the United States of America (2006), 103 (2), 425-430CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Mycoplasma genitalium has the smallest genome of any organism that can be grown in pure culture. It has a minimal metab. and little genomic redundancy. Consequently, its genome is expected to be a close approxn. to the minimal set of genes needed to sustain bacterial life. Using global transposon mutagenesis, gene disruption mutants were isolated and characterized for 100 different nonessential protein-coding genes. None of the 43 RNA-coding genes were disrupted. Herein, 382 of the 482 M. genitalium protein-coding genes were identified as essential, plus 5 sets of disrupted genes that encode proteins with potentially redundant essential functions, such as phosphate transport. Genes encoding proteins of unknown function constitute 28% of the essential protein-coding genes set. Disruption of some genes accelerated M. genitalium growth. The genome of M. genitalium G37 (ATCC 33530) was resequenced and found to differ from the previously sequenced version at 34 sites; the new sequences replaces the original M. genitalium genome sequence in GenBank/EMBL/DDBJ under accession no. L43967.
- 26Hutchison, C. A., 3rd; Chuang, R.-Y.; Noskov, V. N.; Assad-Garcia, N.; Deerinck, T. J.; Ellisman, M. H.; Gill, J.; Kannan, K.; Karas, B. J.; Ma, L.; Pelletier, J. F.; Qi, Z.-Q.; Richter, R. A.; Strychalski, E. A.; Sun, L.; Suzuki, Y.; Tsvetanova, B.; Wise, K. S.; Smith, H. O.; Glass, J. I.; Merryman, C.; Gibson, D. G.; Venter, J. C. Design and Synthesis of a Minimal Bacterial Genome. Science 2016, 351 (6280), aad6253, DOI: 10.1126/science.aad6253Google ScholarThere is no corresponding record for this reference.
- 27Hashimoto, M.; Ichimura, T.; Mizoguchi, H.; Tanaka, K.; Fujimitsu, K.; Keyamura, K.; Ote, T.; Yamakawa, T.; Yamazaki, Y.; Mori, H.; Katayama, T.; Kato, J.-I. Cell Size and Nucleoid Organization of Engineered Escherichia Coli Cells with a Reduced Genome. Mol. Microbiol. 2005, 55 (1), 137– 149, DOI: 10.1111/j.1365-2958.2004.04386.xGoogle Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXotlSrtw%253D%253D&md5=484e3e2ccc942985f12b165903bf23f2Cell size and nucleoid organization of engineered Escherichia coli cells with a reduced genomeHashimoto, Masayuki; Ichimura, Toshiharu; Mizoguchi, Hiroshi; Tanaka, Kimie; Fujimitsu, Kazuyuki; Keyamura, Kenji; Ote, Tomotake; Yamakawa, Takehiro; Yamazaki, Yukiko; Mori, Hideo; Katayama, Tsutomu; Kato, Jun-ichiMolecular Microbiology (2005), 55 (1), 137-149CODEN: MOMIEE; ISSN:0950-382X. (Blackwell Publishing Ltd.)The minimization of a genome is necessary to identify exptl. the minimal gene set that contains only those genes that are essential and sufficient to sustain a functioning cell. Recent developments in genetic techniques have made it possible to generate bacteria with a markedly reduced genome. We developed a simple system for formation of markerless chromosomal deletions, and constructed and characterized a series of large-scale chromosomal deletion mutants of Escherichia coli that lack between 2.4 and 29.7% of the parental chromosome. Combining deletion mutations changes cell length and width, and the mutant cells with larger deletions were even longer and wider than the parental cells. The nucleoid organization of the mutants is also changed: the nucleoids occur as multiple small nucleoids and are localized peripherally near the envelope. Inhibition of translation causes them to condense into one or two packed nucleoids, suggesting that the coupling of transcription and translation of membrane proteins peripherally localizes chromosomes. Because these phenotypes are similar to those of spherical cells, those may be a consequence of the morphol. change. Based on the nucleoid localization obsd. with these mutants, we discuss the cellular nucleoid dynamics.
- 28Pósfai, G.; Plunkett, G., 3rd; Fehér, T.; Frisch, D.; Keil, G. M.; Umenhoffer, K.; Kolisnychenko, V.; Stahl, B.; Sharma, S. S.; de Arruda, M.; Burland, V.; Harcum, S. W.; Blattner, F. R. Emergent Properties of Reduced-Genome Escherichia Coli. Science 2006, 312 (5776), 1044– 1046, DOI: 10.1126/science.1126439Google ScholarThere is no corresponding record for this reference.
- 29Lartigue, C.; Glass, J. I.; Alperovich, N.; Pieper, R.; Parmar, P. P.; Hutchison, C. A., 3rd; Smith, H. O.; Venter, J. C. Genome Transplantation in Bacteria: Changing One Species to Another. Science 2007, 317 (5838), 632– 638, DOI: 10.1126/science.1144622Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXptVCgu7Y%253D&md5=3acd99db81d8698752ffad0636948da2Genome transplantation in bacteria: changing one species to anotherLartigue, Carole; Glass, John I.; Alperovich, Nina; Pieper, Rembert; Parmar, Prashanth P.; Hutchison, Clyde A., III; Smith, Hamilton O.; Venter, J. CraigScience (Washington, DC, United States) (2007), 317 (5838), 632-638CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)As a step toward propagation of synthetic genomes, the authors completely replaced the genome of a bacterial cell with one from another species by transplanting a whole genome as naked DNA. Intact genomic DNA from Mycoplasma mycoides large colony (LC), virtually free of protein, was transplanted into Mycoplasma capricolum cells by polyethylene glycol-mediated transformation. Cells selected for tetracycline resistance, carried by the M. mycoides LC chromosome, contain the complete donor genome and are free of detectable recipient genomic sequences. These cells that result from genome transplantation are phenotypically identical to the M. mycoides LC donor strain as judged by several criteria.
- 30Gibson, D. G.; Benders, G. A.; Andrews-Pfannkoch, C.; Denisova, E. A.; Baden-Tillson, H.; Zaveri, J.; Stockwell, T. B.; Brownley, A.; Thomas, D. W.; Algire, M. A.; Merryman, C.; Young, L.; Noskov, V. N.; Glass, J. I.; Venter, J. C.; Hutchison, C. A., 3rd; Smith, H. O. Complete Chemical Synthesis, Assembly, and Cloning of a Mycoplasma Genitalium Genome. Science 2008, 319 (5867), 1215– 1220, DOI: 10.1126/science.1151721Google Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXisVSksbs%253D&md5=c17e288749853e61fa00ffa9048e27ceComplete Chemical Synthesis, Assembly, and Cloning of a Mycoplasma genitalium GenomeGibson, Daniel G.; Benders, Gwynedd A.; Andrews-Pfannkoch, Cynthia; Denisova, Evgeniya A.; Baden-Tillson, Holly; Zaveri, Jayshree; Stockwell, Timothy B.; Brownley, Anushka; Thomas, David W.; Algire, Mikkel A.; Merryman, Chuck; Young, Lei; Noskov, Vladimir N.; Glass, John I.; Venter, J. Craig; Hutchison, Clyde A., III; Smith, Hamilton O.Science (Washington, DC, United States) (2008), 319 (5867), 1215-1220CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)We have synthesized a 582,970-base pair Mycoplasma genitalium genome. This synthetic genome, named M. genitalium JCVI-1.0, contains all the genes of wild-type M. genitalium G37 except MG408, which was disrupted by an antibiotic marker to block pathogenicity and to allow for selection. To identify the genome as synthetic, we inserted "watermarks" at intergenic sites known to tolerate transposon insertions. Overlapping "cassettes" of 5 to 7 kilobases (kb), assembled from chem. synthesized oligonucleotides, were joined by in vitro recombination to produce intermediate assemblies of approx. 24 kb, 72 kb ("1/8 genome"), and 144 kb ("1/4 genome"), which were all cloned as bacterial artificial chromosomes in Escherichia coli. Most of these intermediate clones were sequenced, and clones of all four 1/4 genomes with the correct sequence were identified. The complete synthetic genome was assembled by transformation-assocd. recombination cloning in the yeast Saccharomyces cerevisiae, then isolated and sequenced. A clone with the correct sequence was identified. The methods described here will be generally useful for constructing large DNA mols. from chem. synthesized pieces and also from combinations of natural and synthetic DNA segments.
- 31Lartigue, C.; Vashee, S.; Algire, M. A.; Chuang, R.-Y.; Benders, G. A.; Ma, L.; Noskov, V. N.; Denisova, E. A.; Gibson, D. G.; Assad-Garcia, N.; Alperovich, N.; Thomas, D. W.; Merryman, C.; Hutchison, C. A., 3rd; Smith, H. O.; Venter, J. C.; Glass, J. I. Creating Bacterial Strains from Genomes That Have Been Cloned and Engineered in Yeast. Science 2009, 325 (5948), 1693– 1696, DOI: 10.1126/science.1173759Google Scholar31https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtFGqt7%252FK&md5=bfc12b44f02c66cdc491d57eee6e46fdCreating Bacterial Strains from Genomes That Have Been Cloned and Engineered in YeastLartigue, Carole; Vashee, Sanjay; Algire, Mikkel A.; Chuang, Ray-Yuan; Benders, Gwynedd A.; Ma, Li; Noskov, Vladimir N.; Denisova, Evgeniya A.; Gibson, Daniel G.; Assad-Garcia, Nacyra; Alperovich, Nina; Thomas, David W.; Merryman, Chuck; Hutchison, Clyde A., III; Smith, Hamilton O.; Venter, J. Craig; Glass, John I.Science (Washington, DC, United States) (2009), 325 (5948), 1693-1696CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)We recently reported the chem. synthesis, assembly, and cloning of a bacterial genome in yeast. To produce a synthetic cell, the genome must be transferred from yeast to a receptive cytoplasm. Here we describe methods to accomplish this. We cloned a Mycoplasma mycoides genome as a yeast centromeric plasmid and then transplanted it into Mycoplasma capricolum to produce a viable M. mycoides cell. While in yeast, the genome was altered by using yeast genetic systems and then transplanted to produce a new strain of M. mycoides. These methods allow the construction of strains that could not be produced with genetic tools available for this bacterium. The complete, annotated sequence of the transplanted M. mycoides capri genome is deposited in GenBank/EMBL/DDBJ with accession no. CP001668.
- 32Gibson, D. G.; Glass, J. I.; Lartigue, C.; Noskov, V. N.; Chuang, R.-Y.; Algire, M. A.; Benders, G. A.; Montague, M. G.; Ma, L.; Moodie, M. M.; Merryman, C.; Vashee, S.; Krishnakumar, R.; Assad-Garcia, N.; Andrews-Pfannkoch, C.; Denisova, E. A.; Young, L.; Qi, Z.-Q.; Segall-Shapiro, T. H.; Calvey, C. H.; Parmar, P. P.; Hutchison, C. A., 3rd; Smith, H. O.; Venter, J. C. Creation of a Bacterial Cell Controlled by a Chemically Synthesized Genome. Science 2010, 329 (5987), 52– 56, DOI: 10.1126/science.1190719Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXotVeqsLg%253D&md5=f978c98f3e5c3c9b14bd4c7d8a1eecc7Creation of a bacterial cell controlled by a chemically synthesized genomeGibson, Daniel G.; Glass, John I.; Lartigue, Carole; Noskov, Vladimir N.; Chuang, Ray-Yuan; Algire, Mikkel A.; Benders, Gwynedd A.; Montague, Michael G.; Ma, Li; Moodie, Monzia M.; Merryman, Chuck; Vashee, Sanjay; Krishnakumar, Radha; Assad-Garcia, Nacyra; Andrews-Pfannkoch, Cynthia; Denisova, Evgeniya A.; Young, Lei; Qi, Zhi-Qing; Segall-Shapiro, Thomas H.; Calvey, Christopher H.; Parmar, Prashanth P.; Hutchison, Clyde A., III; Smith, Hamilton O.; Venter, J. CraigScience (Washington, DC, United States) (2010), 329 (5987), 52-56CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)We report the design, synthesis, and assembly of the 1.08-mega-base pair Mycoplasma mycoides JCVI-syn1.0 genome starting from digitized genome sequence information and its transplantation into a M. capricolum recipient cell to create new M. mycoides cells that are controlled only by the synthetic chromosome. The only DNA in the cells is the designed synthetic DNA sequence, including "watermark" sequences and other designed gene deletions and polymorphisms, and mutations acquired during the building process. The new cells have expected phenotypic properties and are capable of continuous self-replication. The complete, annotated synthetic genome sequence is deposited in GenBank/EMBL/DDBJ with accession no. CP002027.
- 33Stano, P.; Luisi, P. L. Semi-Synthetic Minimal Cells: Origin and Recent Developments. Curr. Opin. Biotechnol. 2013, 24 (4), 633– 638, DOI: 10.1016/j.copbio.2013.01.002Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhslCnu7w%253D&md5=0bb71b3497381842f77cfd6bc763c81cSemi-synthetic minimal cells: origin and recent developmentsStano, Pasquale; Luisi, Pier LuigiCurrent Opinion in Biotechnology (2013), 24 (4), 633-638CODEN: CUOBE3; ISSN:0958-1669. (Elsevier B.V.)A review. The notion of minimal cells refers to cellular structures that contain the minimal and sufficient complexity to still be defined as living, or at least capable to display the most important features of biol. cells. Here the authors briefly describe the lab. construction of minimal cells, a project within the broader field of synthetic biol. In particular the authors discuss the advancements in the prepn. of semi-synthetic cells based on the encapsulation of biochems. inside liposomes, illustrating from the one hand the origin of this research and the most recent developments; and from the other the difficulties and limits of the approach. The role of physicochem. understandings is greatly emphasized.
- 34Olivi, L.; Berger, M.; Creyghton, R. N. P.; De Franceschi, N.; Dekker, C.; Mulder, B. M.; Claassens, N. J.; Ten Wolde, P. R.; van der Oost, J. Towards a Synthetic Cell Cycle. Nat. Commun. 2021, 12 (1), 4531, DOI: 10.1038/s41467-021-24772-8Google Scholar34https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhsleqs7jE&md5=dcc559e4d649037affd64b1951698d1fTowards a synthetic cell cycleOlivi, Lorenzo; Berger, Mareike; Creyghton, Ramon N. P.; De Franceschi, Nicola; Dekker, Cees; Mulder, Bela M.; Claassens, Nico J.; ten Wolde, Pieter Rein; van der Oost, JohnNature Communications (2021), 12 (1), 4531CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)Abstr.: Recent developments in synthetic biol. may bring the bottom-up generation of a synthetic cell within reach. A key feature of a living synthetic cell is a functional cell cycle, in which DNA replication and segregation as well as cell growth and division are well integrated. Here, we describe different approaches to recreate these processes in a synthetic cell, based on natural systems and/or synthetic alternatives. Although some individual machineries have recently been established, their integration and control in a synthetic cell cycle remain to be addressed. In this Perspective, we discuss potential paths towards an integrated synthetic cell cycle.
- 35Adamala, K. P.; Martin-Alarcon, D. A.; Guthrie-Honea, K. R.; Boyden, E. S. Engineering Genetic Circuit Interactions within and between Synthetic Minimal Cells. Nat. Chem. 2017, 9 (5), 431– 439, DOI: 10.1038/nchem.2644Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhvVGiur3E&md5=b362d8195003a28512978349366fa02fEngineering genetic circuit interactions within and between synthetic minimal cellsAdamala, Katarzyna P.; Martin-Alarcon, Daniel A.; Guthrie-Honea, Katriona R.; Boyden, Edward S.Nature Chemistry (2017), 9 (5), 431-439CODEN: NCAHBB; ISSN:1755-4330. (Nature Publishing Group)Genetic circuits and reaction cascades are of great importance for synthetic biol., biochem. and bioengineering. An open question is how to maximize the modularity of their design to enable the integration of different reaction networks and to optimize their scalability and flexibility. One option is encapsulation within liposomes, which enables chem. reactions to proceed in well-isolated environments. Here we adapt liposome encapsulation to enable the modular, controlled compartmentalization of genetic circuits and cascades. We demonstrate that it is possible to engineer genetic circuit-contg. synthetic minimal cells (synells) to contain multiple-part genetic cascades, and that these cascades can be controlled by external signals as well as inter-liposomal communication without crosstalk. We also show that liposomes that contain different cascades can be fused in a controlled way so that the products of incompatible reactions can be brought together. Synells thus enable a more modular creation of synthetic biol. cascades, an essential step towards their ultimate programmability.
- 36Fenz, S. F.; Sachse, R.; Schmidt, T.; Kubick, S. Cell-Free Synthesis of Membrane Proteins: Tailored Cell Models out of Microsomes. Biochim. Biophys. Acta 2014, 1838 (5), 1382– 1388, DOI: 10.1016/j.bbamem.2013.12.009Google ScholarThere is no corresponding record for this reference.
- 37Kroll, A. V.; Fang, R. H.; Zhang, L. Biointerfacing and Applications of Cell Membrane-Coated Nanoparticles. Bioconjugate Chem. 2017, 28 (1), 23– 32, DOI: 10.1021/acs.bioconjchem.6b00569Google Scholar37https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhslKgtrbK&md5=aae744045651438cbf397dfce59bb3d7Biointerfacing and Applications of Cell Membrane-Coated NanoparticlesKroll, Ashley V.; Fang, Ronnie H.; Zhang, LiangfangBioconjugate Chemistry (2017), 28 (1), 23-32CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)The cell membrane-coated nanoparticle is a biomimetic platform consisting of a nanoparticulate core coated with membrane derived from a cell, such as a red blood cell, platelet, or cancer cell. The cell membrane "disguise" allows the particles to be perceived by the body as the source cell by interacting with its surroundings using the translocated surface membrane components. The newly bestowed characteristics of the membrane-coated nanoparticle can be utilized for biol. interfacing in the body, providing natural solns. to many biomedical issues. This Review will cover the interactions of these cell membrane-coated nanoparticles and their applications within three biomedical areas of interest, including (i) drug delivery, (ii) detoxification, and (iii) immune modulation.
- 38Gurramkonda, C.; Rao, A.; Borhani, S.; Pilli, M.; Deldari, S.; Ge, X.; Pezeshk, N.; Han, T.-C.; Tolosa, M.; Kostov, Y.; Tolosa, L.; Wood, D. W.; Vattem, K.; Frey, D. D.; Rao, G. Improving the Recombinant Human Erythropoietin Glycosylation Using Microsome Supplementation in CHO Cell-Free System. Biotechnol. Bioeng. 2018, 115 (5), 1253– 1264, DOI: 10.1002/bit.26554Google ScholarThere is no corresponding record for this reference.
- 39Su’etsugu, M.; Takada, H.; Katayama, T.; Tsujimoto, H. Exponential Propagation of Large Circular DNA by Reconstitution of a Chromosome-Replication Cycle. Nucleic Acids Res. 2017, 45 (20), 11525– 11534, DOI: 10.1093/nar/gkx822Google Scholar39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtVyisbvM&md5=97d99e9a4ac7d2baaa3820e6a1d474c8Exponential propagation of large circular DNA by reconstitution of a chromosome-replication cycleSu'etsugu, Masayuki; Takada, Hiraku; Katayama, Tsutomu; Tsujimoto, HirokoNucleic Acids Research (2017), 45 (20), 11525-11534CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)Propagation of genetic information is a fundamental property of living organisms. Escherichia coli has a 4.6 Mb circular chromosome with a replication origin, oriC. While the oriC replication has been reconstituted in vitro more than 30 years ago, continuous repetition of the replication cycle has not yet been achieved. Here, we reconstituted the entire replication cycle with 14 purified enzymes (25 polypeptides) that catalyze initiation at oriC, bidirectional fork progression, Okazaki-fragment maturation and decatenation of the replicated circular products. Because decatenation provides covalently closed supercoiled monomers that are competent for the next round of replication initiation, the replication cycle repeats autonomously and continuously in an isothermal condition. This replication-cycle reaction (RCR) propagates ∼10 kb circular DNA exponentially as intact covalently closed mols., even from a single DNA mol., with a doubling time of ∼8 min and extremely high fidelity. Very large DNA up to 0.2 Mb is successfully propagated within 3 h. We further demonstrate a cell-free cloning in which RCR selectively propagates circular mols. constructed by a multi-fragment assembly reaction. Our results define the min. element necessary for the repetition of the chromosome-replication cycle, and also provide a powerful in vitro tool to generate large circular DNA mols. without relying on conventional biol. cloning.
- 40Shimizu, Y.; Inoue, A.; Tomari, Y.; Suzuki, T.; Yokogawa, T.; Nishikawa, K.; Ueda, T. Cell-Free Translation Reconstituted with Purified Components. Nat. Biotechnol. 2001, 19 (8), 751– 755, DOI: 10.1038/90802Google Scholar40https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXlslekt7g%253D&md5=8560f1b7319ea88b4784a4f02bafcbafCell-free translation reconstituted with purified componentsShimizu, Yoshihiro; Inoue, Akio; Tomari, Yukihide; Suzuki, Tsutomu; Yokogawa, Takashi; Nishikawa, Kazuya; Ueda, TakuyaNature Biotechnology (2001), 19 (8), 751-755CODEN: NABIF9; ISSN:1087-0156. (Nature America Inc.)We have developed a protein-synthesizing system reconstituted from recombinant tagged protein factors purified to homogeneity. The system was able to produce protein at a rate of about 160 μg/mL/h in a batch mode without the need for any supplementary app. The protein products were easily purified within 1 h using affinity chromatog. to remove the tagged protein factors. Moreover, omission of a release factor allowed efficient incorporation of an unnatural amino acid using suppressor tRNA.
- 41Drienovská, I.; Roelfes, G. Expanding the Enzyme Universe with Genetically Encoded Unnatural Amino Acids. Nature Catalysis 2020, 3 (3), 193– 202, DOI: 10.1038/s41929-019-0410-8Google ScholarThere is no corresponding record for this reference.
- 42Medina, E.; Yik, E. J.; Herdewijn, P.; Chaput, J. C. Functional Comparison of Laboratory-Evolved XNA Polymerases for Synthetic Biology. ACS Synth. Biol. 2021, 10 (6), 1429– 1437, DOI: 10.1021/acssynbio.1c00048Google ScholarThere is no corresponding record for this reference.
- 43Theobald, D. L. A Formal Test of the Theory of Universal Common Ancestry. Nature 2010, 465 (7295), 219– 222, DOI: 10.1038/nature09014Google ScholarThere is no corresponding record for this reference.
- 44Yang, Z.; Chen, F.; Alvarado, J. B.; Benner, S. A. Amplification, Mutation, and Sequencing of a Six-Letter Synthetic Genetic System. J. Am. Chem. Soc. 2011, 133 (38), 15105– 15112, DOI: 10.1021/ja204910nGoogle Scholar44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtFaks7zL&md5=cc4561e9527ca359c82494deb9e0be2dAmplification, mutation, and sequencing of a six-letter synthetic genetic systemYang, Zun-Yi; Chen, Fei; Alvarado, J. Brian; Benner, Steven A.Journal of the American Chemical Society (2011), 133 (38), 15105-15112CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)The next goals in the development of a synthetic biol. that uses artificial genetic systems will require chem.-biol. combinations that allow the amplification of DNA contg. any no. of sequential and nonsequential nonstandard nucleotides. This amplification must ensure that the nonstandard nucleotides are not unidirectionally lost during PCR amplification (unidirectional loss would cause the artificial system to revert to an all-natural genetic system). Further, technol. is needed to sequence artificial genetic DNA mols. The work reported here meets all three of these goals for a six-letter artificially expanded genetic information system (AEGIS) that comprises four std. nucleotides (G, A, C, and T) and two addnl. nonstandard nucleotides (Z and P). We report polymerases and PCR conditions that amplify a wide range of GACTZP DNA sequences having multiple consecutive unnatural synthetic genetic components with low (0.2% per theor. cycle) levels of mutation. We demonstrate that residual mutation processes both introduce and remove unnatural nucleotides, allowing the artificial genetic system to evolve as such, rather than revert to a wholly natural system. We then show that mechanisms for these residual mutation processes can be exploited in a strategy to sequence "six-letter" GACTZP DNA. These are all not yet reported for any other synthetic genetic system.
- 45Malyshev, D. A.; Dhami, K.; Lavergne, T.; Chen, T.; Dai, N.; Foster, J. M.; Corrêa, I. R., Jr; Romesberg, F. E. A Semi-Synthetic Organism with an Expanded Genetic Alphabet. Nature 2014, 509 (7500), 385– 388, DOI: 10.1038/nature13314Google Scholar45https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXotVyqtb8%253D&md5=97b4b184cda52cc809b1705e5e88ad8eA semi-synthetic organism with an expanded genetic alphabetMalyshev, Denis A.; Dhami, Kirandeep; Lavergne, Thomas; Chen, Tingjian; Dai, Nan; Foster, Jeremy M.; Correa, Ivan R.; Romesberg, Floyd E.Nature (London, United Kingdom) (2014), 509 (7500), 385-388CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Organisms are defined by the information encoded in their genomes, and since the origin of life this information has been encoded using a two-base-pair genetic alphabet (A-T and G-C). In vitro, the alphabet has been expanded to include several unnatural base pairs (UBPs). We have developed a class of UBPs formed between nucleotides bearing hydrophobic nucleobases, exemplified by the pair formed between d5SICS and dNaM (d5SICS-dNaM), which is efficiently PCR-amplified and transcribed in vitro, and whose unique mechanism of replication has been characterized. However, expansion of an organism's genetic alphabet presents new and unprecedented challenges: the unnatural nucleoside triphosphates must be available inside the cell; endogenous polymerases must be able to use the unnatural triphosphates to faithfully replicate DNA contg. the UBP within the complex cellular milieu; and finally, the UBP must be stable in the presence of pathways that maintain the integrity of DNA. Here we show that an exogenously expressed algal nucleotide triphosphate transporter efficiently imports the triphosphates of both d5SICS and dNaM (d5SICSTP and dNaMTP) into Escherichia coli, and that the endogenous replication machinery uses them to accurately replicate a plasmid contg. d5SICS-dNaM. Neither the presence of the unnatural triphosphates nor the replication of the UBP introduces a notable growth burden. Lastly, we find that the UBP is not efficiently excised by DNA repair pathways. Thus, the resulting bacterium is the first organism to propagate stably an expanded genetic alphabet.
- 46Rech, E. L.; Arber, W. Biodiversity as a Source for Synthetic Domestication of Useful Specific Traits. Ann. Appl. Biol. 2013, 162 (2), 141– 144, DOI: 10.1111/aab.12013Google ScholarThere is no corresponding record for this reference.
- 47Rech, E. Genomics and Synthetic Biology as a Viable Option to Intensify Sustainable Use of Biodiversity. Nat. Precedings 2011, 1– 1, DOI: 10.1038/npre.2011.5759.1Google ScholarThere is no corresponding record for this reference.
- 48Kiyama, H.; Kakizawa, S.; Sasajima, Y.; Tahara, Y. O.; Miyata, M. Reconstitution of a Minimal Motility System Based on Spiroplasma Swimming by Two Bacterial Actins in a Synthetic Minimal Bacterium. Sci. Adv. 2022, 8 (48), eabo7490, DOI: 10.1126/sciadv.abo7490Google ScholarThere is no corresponding record for this reference.
- 49Geiger, O.; Sanchez-Flores, A.; Padilla-Gomez, J.; Degli Esposti, M. Multiple Approaches of Cellular Metabolism Define the Bacterial Ancestry of Mitochondria. Sci. Adv. 2023, 9 (32), eadh0066, DOI: 10.1126/sciadv.adh0066Google ScholarThere is no corresponding record for this reference.
- 50Pinheiro, V. B.; Taylor, A. I.; Cozens, C.; Abramov, M.; Renders, M.; Zhang, S.; Chaput, J. C.; Wengel, J.; Peak-Chew, S.-Y.; McLaughlin, S. H.; Herdewijn, P.; Holliger, P. Synthetic Genetic Polymers Capable of Heredity and Evolution. Science 2012, 336 (6079), 341– 344, DOI: 10.1126/science.1217622Google Scholar50https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XlslOqtL0%253D&md5=4402af6a172599d03c6b7f8a65b7cea0Synthetic Genetic Polymers Capable of Heredity and EvolutionPinheiro, Vitor B.; Taylor, Alexander I.; Cozens, Christopher; Abramov, Mikhail; Renders, Marleen; Zhang, Su; Chaput, John C.; Wengel, Jesper; Peak-Chew, Sew-Yeu; McLaughlin, Stephen H.; Herdewijn, Piet; Holliger, PhilippScience (Washington, DC, United States) (2012), 336 (6079), 341-344CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Genetic information storage and processing rely on just two polymers, DNA and RNA, yet whether their role reflects evolutionary history or fundamental functional constraints is currently unknown. With the use of polymerase evolution and design, we show that genetic information can be stored in and recovered from six alternative genetic polymers based on simple nucleic acid architectures not found in nature [xeno-nucleic acids (XNAs)]. We also select XNA aptamers, which bind their targets with high affinity and specificity, demonstrating that beyond heredity, specific XNAs have the capacity for Darwinian evolution and folding into defined structures. Thus, heredity and evolution, two hallmarks of life, are not limited to DNA and RNA but are likely to be emergent properties of polymers capable of information storage.
- 51Hoshika, S.; Leal, N. A.; Kim, M.-J.; Kim, M.-S.; Karalkar, N. B.; Kim, H.-J.; Bates, A. M.; Watkins, N. E., Jr; SantaLucia, H. A.; Meyer, A. J.; DasGupta, S.; Piccirilli, J. A.; Ellington, A. D.; SantaLucia, J., Jr; Georgiadis, M. M.; Benner, S. A. Hachimoji DNA and RNA: A Genetic System with Eight Building Blocks. Science 2019, 363 (6429), 884– 887, DOI: 10.1126/science.aat0971Google Scholar51https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXjt1elu7c%253D&md5=1ae87a691cfdfbe36a819d4782c305b9Hachimoji DNA and RNA: A genetic system with eight building blocksHoshika, Shuichi; Leal, Nicole A.; Kim, Myong-Jung; Kim, Myong-Sang; Karalkar, Nilesh B.; Kim, Hyo-Joong; Bates, Alison M.; Watkins, Norman E., Jr.; SantaLucia, Holly A.; Meyer, Adam J.; DasGupta, Saurja; Piccirilli, Joseph A.; Ellington, Andrew D.; SantaLucia, John, Jr.; Georgiadis, Millie M.; Benner, Steven A.Science (Washington, DC, United States) (2019), 363 (6429), 884-887CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)We report DNA- and RNA-like systems built from eight nucleotide "letters" (hence the name "hachimoji") that form four orthogonal pairs. These synthetic systems meet the structural requirements needed to support Darwinian evolution, including a polyelectrolyte backbone, predictable thermodn. stability, and stereoregular building blocks that fit a Schroedinger aperiodic crystal. Measured thermodn. parameters predict the stability of hachimoji duplexes, allowing hachimoji DNA to increase the information d. of natural terran DNA. Three crystal structures show that the synthetic building blocks do not perturb the aperiodic crystal seen in the DNA double helix. Hachimoji DNA was then transcribed to give hachimoji RNA in the form of a functioning fluorescent hachimoji aptamer. These results expand the scope of mol. structures that might support life, including life throughout the cosmos.
- 52Jumper, J.; Evans, R.; Pritzel, A.; Green, T.; Figurnov, M.; Ronneberger, O.; Tunyasuvunakool, K.; Bates, R.; Žídek, A.; Potapenko, A.; Bridgland, A.; Meyer, C.; Kohl, S. A. A.; Ballard, A. J.; Cowie, A.; Romera-Paredes, B.; Nikolov, S.; Jain, R.; Adler, J.; Back, T.; Petersen, S.; Reiman, D.; Clancy, E.; Zielinski, M.; Steinegger, M.; Pacholska, M.; Berghammer, T.; Bodenstein, S.; Silver, D.; Vinyals, O.; Senior, A. W.; Kavukcuoglu, K.; Kohli, P.; Hassabis, D. Highly Accurate Protein Structure Prediction with AlphaFold. Nature 2021, 596 (7873), 583– 589, DOI: 10.1038/s41586-021-03819-2Google Scholar52https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvVaktrrL&md5=25964ab1157cd5b74a437333dd86650dHighly accurate protein structure prediction with AlphaFoldJumper, John; Evans, Richard; Pritzel, Alexander; Green, Tim; Figurnov, Michael; Ronneberger, Olaf; Tunyasuvunakool, Kathryn; Bates, Russ; Zidek, Augustin; Potapenko, Anna; Bridgland, Alex; Meyer, Clemens; Kohl, Simon A. A.; Ballard, Andrew J.; Cowie, Andrew; Romera-Paredes, Bernardino; Nikolov, Stanislav; Jain, Rishub; Adler, Jonas; Back, Trevor; Petersen, Stig; Reiman, David; Clancy, Ellen; Zielinski, Michal; Steinegger, Martin; Pacholska, Michalina; Berghammer, Tamas; Bodenstein, Sebastian; Silver, David; Vinyals, Oriol; Senior, Andrew W.; Kavukcuoglu, Koray; Kohli, Pushmeet; Hassabis, DemisNature (London, United Kingdom) (2021), 596 (7873), 583-589CODEN: NATUAS; ISSN:0028-0836. (Nature Portfolio)Proteins are essential to life, and understanding their structure can facilitate a mechanistic understanding of their function. Through an enormous exptl. effort, the structures of around 100,000 unique proteins have been detd., but this represents a small fraction of the billions of known protein sequences. Structural coverage is bottlenecked by the months to years of painstaking effort required to det. a single protein structure. Accurate computational approaches are needed to address this gap and to enable large-scale structural bioinformatics. Predicting the three-dimensional structure that a protein will adopt based solely on its amino acid sequence-the structure prediction component of the 'protein folding problem'-has been an important open research problem for more than 50 years. Despite recent progress, existing methods fall far short of at. accuracy, esp. when no homologous structure is available. Here we provide the first computational method that can regularly predict protein structures with at. accuracy even in cases in which no similar structure is known. We validated an entirely redesigned version of our neural network-based model, AlphaFold, in the challenging 14th Crit. Assessment of protein Structure Prediction (CASP14), demonstrating accuracy competitive with exptl. structures in a majority of cases and greatly outperforming other methods. Underpinning the latest version of AlphaFold is a novel machine learning approach that incorporates phys. and biol. knowledge about protein structure, leveraging multi-sequence alignments, into the design of the deep learning algorithm.
- 53Lee, J. W.; Na, D.; Park, J. M.; Lee, J.; Choi, S.; Lee, S. Y. Systems Metabolic Engineering of Microorganisms for Natural and Non-Natural Chemicals. Nat. Chem. Biol. 2012, 8 (6), 536– 546, DOI: 10.1038/nchembio.970Google Scholar53https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XntF2nt70%253D&md5=c9ca7c6d604a94b7539387f3ed463e6fSystems metabolic engineering of microorganisms for natural and non-natural chemicalsLee, Jeong Wook; Na, Dokyun; Park, Jong Myoung; Lee, Joungmin; Choi, Sol; Lee, Sang YupNature Chemical Biology (2012), 8 (6), 536-546CODEN: NCBABT; ISSN:1552-4450. (Nature Publishing Group)A review. Growing concerns over limited fossil resources and assocd. environmental problems are motivating the development of sustainable processes for the prodn. of chems., fuels and materials from renewable resources. Metabolic engineering is a key enabling technol. for transforming microorganisms into efficient cell factories for these compds. Systems metabolic engineering, which incorporates the concepts and techniques of systems biol., synthetic biol. and evolutionary engineering at the systems level, offers a conceptual and technol. framework to speed the creation of new metabolic enzymes and pathways or the modification of existing pathways for the optimal prodn. of desired products. Here we discuss the general strategies of systems metabolic engineering and examples of its application and offer insights as to when and how each of the different strategies should be used. Finally, we highlight the limitations and challenges to be overcome for the systems metabolic engineering of microorganisms at more advanced levels.
- 54Nielsen, J. Systems Biology of Metabolism. Annu. Rev. Biochem. 2017, 86, 245– 275, DOI: 10.1146/annurev-biochem-061516-044757Google Scholar54https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXksVCqsLc%253D&md5=7bcd6c37b4b2a4f31a09469c652baa0dSystems Biology of MetabolismNielsen, JensAnnual Review of Biochemistry (2017), 86 (), 245-275CODEN: ARBOAW; ISSN:0066-4154. (Annual Reviews)Metab. is highly complex and involves thousands of different connected reactions; it is therefore necessary to use math. models for holistic studies. The use of math. models in biol. is referred to as systems biol. In this review, the principles of systems biol. are described, and two different types of math. models used for studying metab. are discussed: kinetic models and genome-scale metabolic models. The use of different omics technologies, including transcriptomics, proteomics, metabolomics, and fluxomics, for studying metab. is presented. Finally, the application of systems biol. for analyzing global regulatory structures, engineering the metab. of cell factories, and analyzing human diseases is discussed.
- 55Choi, K. R.; Jang, W. D.; Yang, D.; Cho, J. S.; Park, D.; Lee, S. Y. Systems Metabolic Engineering Strategies: Integrating Systems and Synthetic Biology with Metabolic Engineering. Trends Biotechnol. 2019, 37 (8), 817– 837, DOI: 10.1016/j.tibtech.2019.01.003Google Scholar55https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhvF2ntLg%253D&md5=958b9cb4e7d60ebda46f6b086dcd7e97Systems Metabolic Engineering Strategies: Integrating Systems and Synthetic Biology with Metabolic EngineeringChoi, Kyeong Rok; Jang, Woo Dae; Yang, Dongsoo; Cho, Jae Sung; Park, Dahyeon; Lee, Sang YupTrends in Biotechnology (2019), 37 (8), 817-837CODEN: TRBIDM; ISSN:0167-7799. (Elsevier Ltd.)A review. Metabolic engineering allows development of microbial strains efficiently producing chems. and materials, but it requires much time, effort, and cost to make the strains industrially competitive. Systems metabolic engineering, which integrates tools and strategies of systems biol., synthetic biol., and evolutionary engineering with traditional metabolic engineering, has recently been used to facilitate development of high-performance strains. The past decade has witnessed this interdisciplinary strategy continuously being improved toward the development of industrially competitive overproducer strains. In this article, current trends in systems metabolic engineering including tools and strategies are reviewed, focusing on recent developments in selection of host strains, metabolic pathway reconstruction, tolerance enhancement, and metabolic flux optimization. Also, future challenges and prospects are discussed.
- 56Karim, A. S.; Dudley, Q. M.; Juminaga, A.; Yuan, Y.; Crowe, S. A.; Heggestad, J. T.; Garg, S.; Abdalla, T.; Grubbe, W. S.; Rasor, B. J.; Coar, D. N.; Torculas, M.; Krein, M.; Liew, F. E.; Quattlebaum, A.; Jensen, R. O.; Stuart, J. A.; Simpson, S. D.; Köpke, M.; Jewett, M. C. In Vitro Prototyping and Rapid Optimization of Biosynthetic Enzymes for Cell Design. Nat. Chem. Biol. 2020, 16 (8), 912– 919, DOI: 10.1038/s41589-020-0559-0Google Scholar56https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtFKgtL3K&md5=aac0bdcd46766b221480e35b41f9639cIn vitro prototyping and rapid optimization of biosynthetic enzymes for cell designKarim, Ashty S.; Dudley, Quentin M.; Juminaga, Alex; Yuan, Yongbo; Crowe, Samantha A.; Heggestad, Jacob T.; Garg, Shivani; Abdalla, Tanus; Grubbe, William S.; Rasor, Blake J.; Coar, David N.; Torculas, Maria; Krein, Michael; Liew, FungMin; Quattlebaum, Amy; Jensen, Rasmus O.; Stuart, Jeffrey A.; Simpson, Sean D.; Kopke, Michael; Jewett, Michael C.Nature Chemical Biology (2020), 16 (8), 912-919CODEN: NCBABT; ISSN:1552-4450. (Nature Research)The design and optimization of biosynthetic pathways for industrially relevant, non-model organisms is challenging due to transformation idiosyncrasies, reduced nos. of validated genetic parts and a lack of high-throughput workflows. Here the authors describe a platform for in vitro prototyping and rapid optimization of biosynthetic enzymes (iPROBE) to accelerate this process. In iPROBE, cell lysates are enriched with biosynthetic enzymes by cell-free protein synthesis and then metabolic pathways are assembled in a mix-and-match fashion to assess pathway performance. The authors demonstrate iPROBE by screening 54 different cell-free pathways for 3-hydroxybutyrate prodn. and optimizing a six-step butanol pathway across 205 permutations using data-driven design. Observing a strong correlation (r = 0.79) between cell-free and cellular performance, the authors then scaled up the authors' highest-performing pathway, which improved in vivo 3-HB prodn. in Clostridium by 20-fold to 14.63 ± 0.48 g L-1. The authors expect iPROBE to accelerate design-build-test cycles for industrial biotechnol.
- 57Dudley, Q. M.; Karim, A. S.; Nash, C. J.; Jewett, M. C. In Vitro Prototyping of Limonene Biosynthesis Using Cell-Free Protein Synthesis. Metab. Eng. 2020, 61, 251– 260, DOI: 10.1016/j.ymben.2020.05.006Google Scholar57https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtl2itrzI&md5=bc5f4261c3a62805f1c904e797ec2134In vitro prototyping of limonene biosynthesis using cell-free protein synthesisDudley, Quentin M.; Karim, Ashty S.; Nash, Connor J.; Jewett, Michael C.Metabolic Engineering (2020), 61 (), 251-260CODEN: MEENFM; ISSN:1096-7176. (Elsevier B.V.)Metabolic engineering of microorganisms to produce sustainable chems. has emerged as an important part of the global bioeconomy. Unfortunately, efforts to design and engineer microbial cell factories are challenging because design-build-test cycles, iterations of re-engineering organisms to test and optimize new sets of enzymes, are slow. To alleviate this challenge, we demonstrate a cell-free approach termed in vitro Prototyping and Rapid Optimization of Biosynthetic Enzymes (or iPROBE). In iPROBE, a large no. of pathway combinations can be rapidly built and optimized. The key idea is to use cell-free protein synthesis (CFPS) to manuf. pathway enzymes in sep. reactions that are then mixed to modularly assemble multiple, distinct biosynthetic pathways. As a model, we apply our approach to the 9-step heterologous enzyme pathway to limonene in exts. from Escherichia coli. In iterative cycles of design, we studied the impact of 54 enzyme homologs, multiple enzyme levels, and cofactor concns. on pathway performance. In total, we screened over 150 unique sets of enzymes in 580 unique pathway conditions to increase limonene prodn. in 24 h from 0.2 to 4.5 mM (23-610 mg/L). Finally, to demonstrate the modularity of this pathway, we also synthesized the biofuel precursors pinene and bisabolene. We anticipate that iPROBE will accelerate design-build-test cycles for metabolic engineering, enabling data-driven multiplexed cell-free methods for testing large combinations of biosynthetic enzymes to inform cellular design.
- 58Averesch, N. J. H.; Krömer, J. O. Metabolic Engineering of the Shikimate Pathway for Production of Aromatics and Derived Compounds-Present and Future Strain Construction Strategies. Front. Bioeng. Biotechnol. 2018, 6, 32, DOI: 10.3389/fbioe.2018.00032Google Scholar58https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1MjgsVGqtw%253D%253D&md5=413681743faac53a82a74216557d0b12Metabolic Engineering of the Shikimate Pathway for Production of Aromatics and Derived Compounds-Present and Future Strain Construction StrategiesAveresch Nils J H; Kromer Jens OFrontiers in bioengineering and biotechnology (2018), 6 (), 32 ISSN:2296-4185.The aromatic nature of shikimate pathway intermediates gives rise to a wealth of potential bio-replacements for commonly fossil fuel-derived aromatics, as well as naturally produced secondary metabolites. Through metabolic engineering, the abundance of certain intermediates may be increased, while draining flux from other branches off the pathway. Often targets for genetic engineering lie beyond the shikimate pathway, altering flux deep in central metabolism. This has been extensively used to develop microbial production systems for a variety of compounds valuable in chemical industry, including aromatic and non-aromatic acids like muconic acid, para-hydroxybenzoic acid, and para-coumaric acid, as well as aminobenzoic acids and aromatic α-amino acids. Further, many natural products and secondary metabolites that are valuable in food- and pharma-industry are formed outgoing from shikimate pathway intermediates. (Re)construction of such routes has been shown by de novo production of resveratrol, reticuline, opioids, and vanillin. In this review, strain construction strategies are compared across organisms and put into perspective with requirements by industry for commercial viability. Focus is put on enhancing flux to and through shikimate pathway, and engineering strategies are assessed in order to provide a guideline for future optimizations.
- 59Jang, W. D.; Kim, G. B.; Lee, S. Y. An Interactive Metabolic Map of Bio-Based Chemicals. Trends Biotechnol. 2023, 41 (1), 10– 14, DOI: 10.1016/j.tibtech.2022.07.013Google ScholarThere is no corresponding record for this reference.
- 60Lai, H.-E.; Obled, A. M. C.; Chee, S. M.; Morgan, R. M.; Lynch, R.; Sharma, S. V.; Moore, S. J.; Polizzi, K. M.; Goss, R. J. M.; Freemont, P. S. GenoChemetic Strategy for Derivatization of the Violacein Natural Product Scaffold. ACS Chem. Biol. 2021, 16 (11), 2116– 2123, DOI: 10.1021/acschembio.1c00483Google Scholar60https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXit1ehtr3I&md5=c0abd319d896030a21f20c0660a2b8a1GenoChemetic Strategy for Derivatization of the Violacein Natural Product ScaffoldLai, Hung-En; Obled, Alan M. C.; Chee, Soo Mei; Morgan, Rhodri M.; Lynch, Rosemary; Sharma, Sunil V.; Moore, Simon J.; Polizzi, Karen M.; Goss, Rebecca J. M.; Freemont, Paul S.ACS Chemical Biology (2021), 16 (11), 2116-2123CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)Natural products and their analogs are often challenging to synthesize due to their complex scaffolds and embedded functional groups. Solely relying on engineering the biosynthesis of natural products may lead to limited compd. diversity. Integrating synthetic biol. with synthetic chem. allows rapid access to much more diverse portfolios of xenobiotic compds., which may accelerate the discovery of new therapeutics. As a proof-of-concept, by supplementing an Escherichia coli strain expressing the violacein biosynthesis pathway with 5-bromo-tryptophan in vitro or tryptophan 7-halogenase RebH in vivo, six halogenated analogs of violacein or deoxyviolacein were generated, demonstrating the promiscuity of the violacein biosynthesis pathway. Furthermore, 20 new derivs. were generated from 5-brominated violacein analogs via the Suzuki-Miyaura cross-coupling reaction directly using the crude ext. without prior purifn. Herein we demonstrate a flexible and rapid approach to access a diverse chem. space that can be applied to a wide range of natural product scaffolds.
- 61Galanie, S.; Thodey, K.; Trenchard, I. J.; Filsinger Interrante, M.; Smolke, C. D. Complete Biosynthesis of Opioids in Yeast. Science 2015, 349 (6252), 1095– 1100, DOI: 10.1126/science.aac9373Google Scholar61https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsVyntbbK&md5=7304186260192653b8c7c78b9119fde3Complete biosynthesis of opioids in yeastGalanie, Stephanie; Thodey, Kate; Trenchard, Isis J.; Filsinger Interrante, Maria; Smolke, Christina D.Science (Washington, DC, United States) (2015), 349 (6252), 1095-1100CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Opioids are the primary drugs used in Western medicine for pain management and palliative care. Farming of opium poppies remains the sole source of these essential medicines, despite diverse market demands and uncertainty in crop yields due to weather, climate change, and pests. We engineered yeast to produce the selected opioid compds. thebaine and hydrocodone starting from sugar. All work was conducted in a lab. that is permitted and secured for work with controlled substances. We combined enzyme discovery, enzyme engineering, and pathway and strain optimization to realize full opiate biosynthesis in yeast. The resulting opioid biosynthesis strains required the expression of 21 (thebaine) and 23 (hydrocodone) enzyme activities from plants, mammals, bacteria, and yeast itself. This is a proof of principle, and major hurdles remain before optimization and scale-up could be achieved. Open discussions of options for governing this technol. are also needed in order to responsibly realize alternative supplies for these medically relevant compds.
- 62Trenchard, I. J.; Siddiqui, M. S.; Thodey, K.; Smolke, C. D. De Novo Production of the Key Branch Point Benzylisoquinoline Alkaloid Reticuline in Yeast. Metab. Eng. 2015, 31, 74– 83, DOI: 10.1016/j.ymben.2015.06.010Google Scholar62https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtFOktL%252FE&md5=e07d3d284b8d8cb60631dd1045b2d68aDe novo production of the key branch point benzylisoquinoline alkaloid reticuline in yeastTrenchard, Isis J.; Siddiqui, Michael S.; Thodey, Kate; Smolke, Christina D.Metabolic Engineering (2015), 31 (), 74-83CODEN: MEENFM; ISSN:1096-7176. (Elsevier B. V.)Microbial biosynthesis for plant-based natural products, such as the benzylisoquinoline alkaloids (BIAs), has the potential to address limitations in plant-based supply of established drugs and make new mols. available for drug discovery. While yeast strains have been engineered to produce a variety of downstream BIAs including the opioids, these strains have relied on feeding an early BIA substrate. We describe the de novo synthesis of the major BIA branch point intermediate reticuline via norcoclaurine in Saccharomyces cerevisiae. Modifications were introduced into yeast central metab. to increase supply of the BIA precursor tyrosine, allowing us to achieve a 60-fold increase in prodn. of the early benzylisoquinoline scaffold from fed dopamine with no supply of exogenous tyrosine. Yeast strains further engineered to express a mammalian tyrosine hydroxylase, 4 mammalian tetrahydrobiopterin biosynthesis and recycling enzymes, and a bacterial DOPA decarboxylase produced norcoclaurine de novo. We further increased prodn. of early benzylisoquinoline scaffolds by 160-fold through introducing mutant tyrosine hydroxylase enzymes, an optimized plant norcoclaurine synthase variant, and optimizing culture conditions. Finally, we incorporated 5 addnl. plant enzymes (3 methyltransferases, a cytochrome P 450, and its reductase partner) to achieve de novo prodn. of the key branch point mol. reticuline with a titer of 19.2 μg/L. These strains and reconstructed pathways will serve as a platform for the biosynthesis of diverse natural and novel BIAs.
- 63Chan, C. T. Y.; Lee, J. W.; Cameron, D. E.; Bashor, C. J.; Collins, J. J. Deadman” and “Passcode” Microbial Kill Switches for Bacterial Containment. Nat. Chem. Biol. 2016, 12 (2), 82– 86, DOI: 10.1038/nchembio.1979Google Scholar63https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhvFemsbrE&md5=c1cd6452791fc321b2432e61ad682557'Deadman' and 'Passcode' microbial kill switches for bacterial containmentChan, Clement T. Y.; Lee, Jeong Wook; Cameron, D. Ewen; Bashor, Caleb J.; Collins, James J.Nature Chemical Biology (2016), 12 (2), 82-86CODEN: NCBABT; ISSN:1552-4450. (Nature Publishing Group)Biocontainment systems that couple environmental sensing with circuit-based control of cell viability could be used to prevent escape of genetically modified microbes into the environment. Here we present two engineered safeguard systems known as the 'Deadman' and 'Passcode' kill switches. The Deadman kill switch uses unbalanced reciprocal transcriptional repression to couple a specific input signal with cell survival. The Passcode kill switch uses a similar two-layered transcription design and incorporates hybrid LacI-GalR family transcription factors to provide diverse and complex environmental inputs to control circuit function. These synthetic gene circuits efficiently kill Escherichia coli and can be readily reprogrammed to change their environmental inputs, regulatory architecture and killing mechanism.
- 64Caliando, B. J.; Voigt, C. A. Targeted DNA Degradation Using a CRISPR Device Stably Carried in the Host Genome. Nat. Commun. 2015, 6, 6989, DOI: 10.1038/ncomms7989Google Scholar64https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtF2kt7fJ&md5=a01b28683e14e8f6c185dbdfcc300293Targeted DNA degradation using a CRISPR device stably carried in the host genomeCaliando, Brian J.; Voigt, Christopher A.Nature Communications (2015), 6 (), 6989CODEN: NCAOBW; ISSN:2041-1723. (Nature Publishing Group)Once an engineered organism completes its task, it is useful to degrade the assocd. DNA to reduce environmental release and protect intellectual property. Here we present a genetically encoded device (DNAi) that responds to a transcriptional input and degrades user-defined DNA. This enables engineered regions to be obscured when the cell enters a new environment. DNAi is based on type-IE CRISPR biochem. and a synthetic CRISPR array defines the DNA target(s). When the input is on, plasmid DNA is degraded 108-fold. When the genome is targeted, this causes cell death, reducing viable cells by a factor of 108. Further, the CRISPR nuclease can direct degrdn. to specific genomic regions (for example, engineered or inserted DNA), which could be used to complicate recovery and sequencing efforts. DNAi can be stably carried in an engineered organism, with no impact on cell growth, plasmid stability or DNAi inducibility even after passaging for >2 mo.
- 65Nyerges, A.; Vinke, S.; Flynn, R.; Owen, S. V.; Rand, E. A.; Budnik, B.; Keen, E.; Narasimhan, K.; Marchand, J. A.; Baas-Thomas, M.; Liu, M.; Chen, K.; Chiappino-Pepe, A.; Hu, F.; Baym, M.; Church, G. M. A Swapped Genetic Code Prevents Viral Infections and Gene Transfer. Nature 2023, 615 (7953), 720– 727, DOI: 10.1038/s41586-023-05824-zGoogle ScholarThere is no corresponding record for this reference.
- 66Scrinis, G.; Lyons, K. The Emerging Nano-Corporate Paradigm: Nanotechnology and the Transformation of Nature, Food and Agri-Food Systems. Int. J. Sociol. Agric. Food 2007, 15 (2), 22– 44Google ScholarThere is no corresponding record for this reference.
- 67Nitrogen fixing bacteria - microbial fertilizer. https://www.pivotbio.com/ (accessed on April 30, 2023).Google ScholarThere is no corresponding record for this reference.
- 68Kaul, S.; Choudhary, M.; Gupta, S.; Dhar, M. K. Engineering Host Microbiome for Crop Improvement and Sustainable Agriculture. Front. Microbiol. 2021, 12, 635917, DOI: 10.3389/fmicb.2021.635917Google ScholarThere is no corresponding record for this reference.
- 69Mueller, U. G.; Sachs, J. L. Engineering Microbiomes to Improve Plant and Animal Health. Trends Microbiol. 2015, 23 (10), 606– 617, DOI: 10.1016/j.tim.2015.07.009Google Scholar69https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsFKhtL7L&md5=8743f515980f584435a175d2b74a33bdEngineering Microbiomes to Improve Plant and Animal HealthMueller, U. G.; Sachs, J. L.Trends in Microbiology (2015), 23 (10), 606-617CODEN: TRMIEA; ISSN:0966-842X. (Elsevier Ltd.)A review. Animal and plant microbiomes encompass diverse microbial communities that colonize every accessible host tissue. These microbiomes enhance host functions, contributing to host health and fitness. A novel approach to improve animal and plant fitness is to artificially select upon microbiomes, thus engineering evolved microbiomes with specific effects on host fitness. The authors call this engineering approach host-mediated microbiome selection, because this method selects upon microbial communities indirectly through the host and leverages host traits that evolved to influence microbiomes. In essence, host phenotypes were used as probes to gauge and manipulate those microbiome functions that impact host fitness. To facilitate research on host-mediated microbiome engineering, the authors explain and compare the principal methods to impose artificial selection on microbiomes; discuss advantages and potential challenges of each method; offer a skeptical appraisal of each method in light of these potential challenges; and outline exptl. strategies to optimize microbiome engineering. Finally, the authors develop a predictive framework for microbiome engineering that organizes research around principles of artificial selection, quant. genetics, and microbial community ecol.
- 70Jin Song, S.; Woodhams, D. C.; Martino, C.; Allaband, C.; Mu, A.; Javorschi-Miller-Montgomery, S.; Suchodolski, J. S.; Knight, R. Engineering the Microbiome for Animal Health and Conservation. Exp. Biol. Med. 2019, 244 (6), 494– 504, DOI: 10.1177/1535370219830075Google ScholarThere is no corresponding record for this reference.
- 71Aziz, C. E.; Borden, R. C.; Coates, J. D.; Cox, E. E.; Downey, D. C.; Evans, P. J.; Hatzinger, P. B.; Henry, B. M.; Andrew Jackson, W.; Krug, T. A.; Tony Lieberman, M.; Loehr, R. C.; Norris, R. D.; Nzengung, V. A.; Perlmutter, M. W.; Schaefer, C. E.; Stroo, H. F.; Herb Ward, C.; Winstead, C. J.; Wolfe, C. In Situ Bioremediation of Perchlorate in Groundwater; Springer: New York.Google ScholarThere is no corresponding record for this reference.
- 72Hou, D.; O’Connor, D.; Igalavithana, A. D.; Alessi, D. S.; Luo, J.; Tsang, D. C. W.; Sparks, D. L.; Yamauchi, Y.; Rinklebe, J.; Ok, Y. S. Metal Contamination and Bioremediation of Agricultural Soils for Food Safety and Sustainability. Nature Reviews Earth & Environment 2020, 1 (7), 366– 381, DOI: 10.1038/s43017-020-0061-yGoogle ScholarThere is no corresponding record for this reference.
- 73Maity, W.; Maity, S.; Bera, S.; Roy, A. Emerging Roles of PETase and MHETase in the Biodegradation of Plastic Wastes. Appl. Biochem. Biotechnol. 2021, 193 (8), 2699– 2716, DOI: 10.1007/s12010-021-03562-4Google ScholarThere is no corresponding record for this reference.
- 74de Lorenzo, V.; Prather, K. L.; Chen, G.-Q.; O’Day, E.; von Kameke, C.; Oyarzún, D. A.; Hosta-Rigau, L.; Alsafar, H.; Cao, C.; Ji, W.; Okano, H.; Roberts, R. J.; Ronaghi, M.; Yeung, K.; Zhang, F.; Lee, S. Y. The Power of Synthetic Biology for Bioproduction, Remediation and Pollution Control: The UN’s Sustainable Development Goals Will Inevitably Require the Application of Molecular Biology and Biotechnology on a Global Scale. EMBO Rep 2018, DOI: 10.15252/embr.201745658Google ScholarThere is no corresponding record for this reference.
- 75Thavarajah, W.; Verosloff, M. S.; Jung, J. K.; Alam, K. K.; Miller, J. D.; Jewett, M. C.; Young, S. L.; Lucks, J. B. A Primer on Emerging Field-Deployable Synthetic Biology Tools for Global Water Quality Monitoring. NPJ. Clean Water 2020, DOI: 10.1038/s41545-020-0064-8Google ScholarThere is no corresponding record for this reference.
- 76Gupta, R. M.; Schnitzler, G. R.; Fang, S.; Lee-Kim, V. S.; Barry, A. Multiomic Analysis and CRISPR Perturbation Screens Identify Endothelial Cell Programs and Novel Therapeutic Targets for Coronary Artery Disease. Arterioscler. Thromb. Vasc. Biol. 2023, 43 (5), 600– 608, DOI: 10.1161/ATVBAHA.123.318328Google ScholarThere is no corresponding record for this reference.
- 77Virchow, R. L. K. Disease, Life and Man: Selected Essays; Rather, L. J., Translator; Stanford University Press, 1958.Google ScholarThere is no corresponding record for this reference.
- 78Center for Biologics Evaluation; Research. Approved Cellular and Gene Therapy Products. U.S. Food and Drug Administration. https://www.fda.gov/vaccines-blood-biologics/cellular-gene-therapy-products/approved-cellular-and-gene-therapy-products (accessed on August 21, 2023).Google ScholarThere is no corresponding record for this reference.
- 79Bashor, C. J.; Hilton, I. B.; Bandukwala, H.; Smith, D. M.; Veiseh, O. Engineering the next Generation of Cell-Based Therapeutics. Nat. Rev. Drug Discovery 2022, 21 (9), 655– 675, DOI: 10.1038/s41573-022-00476-6Google ScholarThere is no corresponding record for this reference.
- 80Rosenbaum, L. Tragedy, Perseverance, and Chance - The Story of CAR-T Therapy. N. Engl. J. Med. 2017, 377 (14), 1313– 1315, DOI: 10.1056/NEJMp1711886Google ScholarThere is no corresponding record for this reference.
- 81Hay, A. E.; Cheung, M. C. CAR T-Cells: Costs, Comparisons, and Commentary. J. Med. Econ. 2019, 22 (7), 613– 615, DOI: 10.1080/13696998.2019.1582059Google ScholarThere is no corresponding record for this reference.
- 82Sampson, K.; Sorenson, C.; Adamala, K. FDA needs to get ready to evaluate synthetic cells, the next generation of therapeutics. STAT. https://www.statnews.com/2022/07/26/fda-develop-framework-evaluate-synthetic-cells/ (accessed on January 6, 2024).Google ScholarThere is no corresponding record for this reference.
- 83Lussier, F.; Staufer, O.; Platzman, I.; Spatz, J. P. Can Bottom-Up Synthetic Biology Generate Advanced Drug-Delivery Systems?. Trends Biotechnol. 2021, 39 (5), 445– 459, DOI: 10.1016/j.tibtech.2020.08.002Google Scholar83https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhs1OltLfJ&md5=7fae97c9cc9dc7fcaec9b229371a4c69Can Bottom-Up Synthetic Biology Generate Advanced Drug-Delivery SystemsLussier, Felix; Staufer, Oskar; Platzman, Ilia; Spatz, Joachim P.Trends in Biotechnology (2021), 39 (5), 445-459CODEN: TRBIDM; ISSN:0167-7799. (Elsevier Ltd.)A review. Creating a magic bullet that can selectively kill cancer cells while sparing nearby healthy cells remains one of the most ambitious objectives in pharmacol. Nanomedicine, which relies on the use of nanotechnologies to fight disease, was envisaged to fulfill this coveted goal. Despite substantial progress, the structural complexity of therapeutic vehicles impedes their broad clin. application. Novel modular manufg. approaches for engineering programmable drug carriers may be able to overcome some fundamental limitations of nanomedicine. We discuss how bottom-up synthetic biol. principles, empowered by microfluidics, can palliate current drug carrier assembly limitations, and we demonstrate how such a magic bullet could be engineered from the bottom up to ultimately improve clin. outcomes for patients.
- 84Staufer, O.; Dietrich, F.; Rimal, R.; Schröter, M.; Fabritz, S.; Boehm, H.; Singh, S.; Möller, M.; Platzman, I.; Spatz, J. P. Bottom-up Assembly of Biomedical Relevant Fully Synthetic Extracellular Vesicles. Sci. Adv. 2021, 7 (36), eabg6666, DOI: 10.1126/sciadv.abg6666Google ScholarThere is no corresponding record for this reference.
- 85Ghaemmaghamian, Z.; Zarghami, R.; Walker, G.; O’Reilly, E.; Ziaee, A. Stabilizing Vaccines via Drying: Quality by Design Considerations. Adv. Drug Delivery Rev. 2022, 187, 114313, DOI: 10.1016/j.addr.2022.114313Google Scholar85https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhsFKjsLfM&md5=268c97346f1c1595a47a83b73069aaa1Stabilizing vaccines via drying: Quality by design considerationsGhaemmaghamian, Zahra; Zarghami, Reza; Walker, Gavin; O'Reilly, Emmet; Ziaee, AhmadAdvanced Drug Delivery Reviews (2022), 187 (), 114313CODEN: ADDREP; ISSN:0169-409X. (Elsevier B.V.)A review. Pandemics and epidemics are continually challenging human beings' health and imposing major stresses on the societies particularly over the last few decades, when their frequency has increased significantly. Protecting humans from multiple diseases is best achieved through vaccination. However, vaccines thermal instability has always been a hurdle in their widespread application, esp. in less developed countries. Furthermore, insufficient vaccine processing capacity is also a major challenge for global vaccination programs. Continuous drying of vaccine formulations is one of the potential solns. to these challenges. This review highlights the challenges on implementing the continuous drying techniques for drying vaccines. The conventional drying methods, emerging technologies and their adaptation by biopharmaceutical industry are investigated considering the patented technologies for drying of vaccines. Moreover, the current progress in applying Quality by Design (QbD) in each of the drying techniques considering the crit. quality attributes (CQAs), crit. process parameters (CPPs) are comprehensively reviewed. An expert advice is presented on the required actions to be taken within the biopharmaceutical industry to move towards continuous stabilization of vaccines in the realm of QbD.
- 86Adiga, R.; Al-Adhami, M.; Andar, A.; Borhani, S.; Brown, S.; Burgenson, D.; Cooper, M. A.; Deldari, S.; Frey, D. D.; Ge, X.; Guo, H.; Gurramkonda, C.; Jensen, P.; Kostov, Y.; LaCourse, W.; Liu, Y.; Moreira, A.; Mupparapu, K.; Peñalber-Johnstone, C.; Pilli, M.; Punshon-Smith, B.; Rao, A.; Rao, G.; Rauniyar, P.; Snovida, S.; Taurani, K.; Tilahun, D.; Tolosa, L.; Tolosa, M.; Tran, K.; Vattem, K.; Veeraraghavan, S.; Wagner, B.; Wilhide, J.; Wood, D. W.; Zuber, A. Point-of-Care Production of Therapeutic Proteins of Good-Manufacturing-Practice Quality. Nat. Biomed Eng. 2018, 2 (9), 675– 686, DOI: 10.1038/s41551-018-0259-1Google Scholar86https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtFensr3N&md5=bbe1f9134ed2590f5d0c500997cb50b5Point-of-care production of therapeutic proteins of good-manufacturing-practice qualityAdiga, Rajani; Al-adhami, Mustafa; Andar, Abhay; Borhani, Shayan; Brown, Sheniqua; Burgenson, David; Cooper, Merideth A.; Deldari, Sevda; Frey, Douglas D.; Ge, Xudong; Guo, Hui; Gurramkonda, Chandrasekhar; Jensen, Penny; Kostov, Yordan; LaCourse, William; Liu, Yang; Moreira, Antonio; Mupparapu, KarunaSri; Penalber-Johnstone, Chariz; Pilli, Manohar; Punshon-Smith, Benjamin; Rao, Aniruddha; Rao, Govind; Rauniyar, Priyanka; Snovida, Sergei; Taurani, Kanika; Tilahun, Dagmawi; Tolosa, Leah; Tolosa, Michael; Tran, Kevin; Vattem, Krishna; Veeraraghavan, Sudha; Wagner, Brandon; Wilhide, Joshua; Wood, David W.; Zuber, AdilNature Biomedical Engineering (2018), 2 (9), 675-686CODEN: NBEAB3; ISSN:2157-846X. (Nature Research)Manufg. technologies for biologics rely on large, centralized, good-manufg.-practice (GMP) prodn. facilities and on a cumbersome product-distribution network. Here, we report the development of an automated and portable medicines-on-demand device that enables consistent, small-scale GMP manufg. of therapeutic-grade biologics on a timescale of hours. The device couples the in vitro translation of target proteins from ribosomal DNA, using exts. from reconstituted lyophilized Chinese hamster ovary cells, with the continuous purifn. of the proteins. We used the device to reproducibly manuf. His-tagged granulocyte-colony stimulating factor, erythropoietin, glucose-binding protein and diphtheria toxoid DT5. Medicines-on-demand technol. may enable the rapid manufg. of biologics at the point of care.
- 87Ginsburg, G. S.; Phillips, K. A. Precision Medicine: From Science To Value. Health Aff. 2018, 37 (5), 694– 701, DOI: 10.1377/hlthaff.2017.1624Google ScholarThere is no corresponding record for this reference.
- 88Agarwal, S.; Saha, S.; Balla, V. K.; Pal, A.; Barui, A.; Bodhak, S. Current Developments in 3D Bioprinting for Tissue and Organ Regeneration-A Review. Front. Mech. Eng. Chin 2020, DOI: 10.3389/fmech.2020.589171Google ScholarThere is no corresponding record for this reference.
- 89Ghosh, S.; Yi, H.-G. A Review on Bioinks and Their Application in Plant Bioprinting. Int. J. Bioprint 2022, 8 (4), 612, DOI: 10.18063/ijb.v8i4.612Google ScholarThere is no corresponding record for this reference.
- 90Santomartino, R.; Averesch, N. J. H.; Bhuiyan, M.; Cockell, C. S.; Colangelo, J.; Gumulya, Y.; Lehner, B.; Lopez-Ayala, I.; McMahon, S.; Mohanty, A.; Santa Maria, S. R.; Urbaniak, C.; Volger, R.; Yang, J.; Zea, L. Toward Sustainable Space Exploration: A Roadmap for Harnessing the Power of Microorganisms. Nat. Commun. 2023, 14 (1), 1391, DOI: 10.1038/s41467-023-37070-2Google ScholarThere is no corresponding record for this reference.
- 91Rothschild, L. J. Synthetic Biology Meets Bioprinting: Enabling Technologies for Humans on Mars (and Earth). Biochem. Soc. Trans. 2016, 44 (4), 1158– 1164, DOI: 10.1042/BST20160067Google Scholar91https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhtlCju7jO&md5=e97bf6b90993c7664400df619e087f6cSynthetic biology meets bioprinting: enabling technologies for humans on Mars (and Earth)Rothschild, Lynn J.Biochemical Society Transactions (2016), 44 (4), 1158-1164CODEN: BCSTB5; ISSN:0300-5127. (Portland Press Ltd.)Human exploration off planet is severely limited by the cost of launching materials into space and by re-supply. Thus materials brought from Earth must be light, stable and reliable at destination. Using traditional approaches, a lunar or Mars base would require either transporting a hefty store of metals or heavy manufg. equipment and construction materials for in situ extn.; both would severely limit any other mission objectives. Long-term human space presence requires periodic replenishment, adding a massive cost overhead. Even robotic missions often sacrifice science goals for heavy radiation and thermal protection. Biol. has the potential to solve these problems because life can replicate and repair itself, and perform a wide variety of chem. reactions including making food, fuel and materials. Synthetic biol. enhances and expands life's evolved repertoire. Using organisms as feedstock, additive manufg. through bioprinting will make possible the dream of producing bespoke tools, food, smart fabrics and even replacement organs on demand. This new approach and the resulting novel products will enable human exploration and settlement on Mars, while providing new manufg. approaches for life on Earth.
- 92Averesch, N. J. H.; Berliner, A. J.; Nangle, S. N.; Zezulka, S.; Vengerova, G. L.; Ho, D.; Casale, C. A.; Lehner, B. A. E.; Snyder, J. E.; Clark, K. B.; Dartnell, L. R.; Criddle, C. S.; Arkin, A. P. Microbial Biomanufacturing for Space-Exploration-What to Take and When to Make. Nat. Commun. 2023, 14 (1), 2311, DOI: 10.1038/s41467-023-37910-1Google ScholarThere is no corresponding record for this reference.
- 93Averesch, N. J. H. Choice of Microbial System for in-Situ Resource Utilization on Mars. Front. Astron. Space Sci. 2021, DOI: 10.3389/fspas.2021.700370Google ScholarThere is no corresponding record for this reference.
- 94Cockell, C. S. Bridging the Gap between Microbial Limits and Extremes in Space: Space Microbial Biotechnology in the next 15 Years. Microb. Biotechnol. 2022, 15 (1), 29– 41, DOI: 10.1111/1751-7915.13927Google ScholarThere is no corresponding record for this reference.
- 95Buddingh’, B. C.; van Hest, J. C. M. Artificial Cells: Synthetic Compartments with Life-like Functionality and Adaptivity. Acc. Chem. Res. 2017, 50 (4), 769– 777, DOI: 10.1021/acs.accounts.6b00512Google Scholar95https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXps1SisQ%253D%253D&md5=979537efa2cf22428e5de31f8267fb31Artificial Cells: Synthetic Compartments with Life-like Functionality and AdaptivityBuddingh', Bastiaan C.; van Hest, Jan C. M.Accounts of Chemical Research (2017), 50 (4), 769-777CODEN: ACHRE4; ISSN:0001-4842. (American Chemical Society)A review. Cells are highly advanced microreactors that form the basis of all life. Their fascinating complexity has inspired scientists to create analogs from synthetic and natural components using a bottom-up approach. The ultimate goal here is to assemble a fully man-made cell that displays functionality and adaptivity as advanced as that found in nature, which will not only provide insight into the fundamental processes in natural cells but also pave the way for new applications of such artificial cells. In this Account, the authors highlight their recent work and that of others on the construction of artificial cells. First, the authors will introduce the key features that characterize a living system; next, the authors will discuss how these have been imitated in artificial cells. First, compartmentalization is crucial to sep. the inner chem. milieu from the external environment. Current state-of-the-art artificial cells comprise subcompartments to mimic the hierarchical architecture of eukaryotic cells and tissue. Furthermore, synthetic gene circuits have been used to encode genetic information that creates complex behavior like pulses or feedback. Addnl., artificial cells have to reproduce to maintain a population. Controlled growth and fission of synthetic compartments have been demonstrated, but the extensive regulation of cell division in nature is still unmatched. Here, the authors also point out important challenges the field needs to overcome to realize its full potential. As artificial cells integrate increasing orders of functionality, maintaining a supporting metab. that can regenerate key metabolites becomes crucial. Furthermore, life does not operate in isolation. Natural cells constantly sense their environment, exchange (chem.) signals, and can move toward a chemoattractant. Here, the authors specifically explore recent efforts to reproduce such adaptivity in artificial cells. For instance, synthetic compartments have been produced that can recruit proteins to the membrane upon an external stimulus or modulate their membrane compn. and permeability to control their interaction with the environment. A next step would be the communication of artificial cells with either bacteria or another artificial cell. Indeed, examples of such primitive chem. signaling are presented. Finally, motility is important for many organisms and has, therefore, also been pursued in synthetic systems. Synthetic compartments that were designed to move in a directed, controlled manner have been assembled, and directed movement toward a chem. attractant is among one of the most life-like directions currently under research. Although the bottom-up construction of an artificial cell that can be truly considered "alive" is still an ambitious goal, the recent work discussed in this Account shows that this is an active field with contributions from diverse disciplines like materials chem. and biochem. Notably, research during the past decade has already provided valuable insights into complex synthetic systems with life-like properties. In the future, artificial cells are thought to contribute to an increased understanding of processes in natural cells and provide opportunities to create smart, autonomous, cell-like materials.
- 96Ichihashi, N. What Can We Learn from the Construction of in Vitro Replication Systems?. Ann. N.Y. Acad. Sci. 2019, 1447 (1), 144– 156, DOI: 10.1111/nyas.14042Google Scholar96https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3M%252Fis1yqtw%253D%253D&md5=0cb66816ca21190294e9a09ce2768e10What can we learn from the construction of in vitro replication systems?Ichihashi NorikazuAnnals of the New York Academy of Sciences (2019), 1447 (1), 144-156 ISSN:.Replication is a central function of living organisms. Several types of replication systems have been constructed in vitro from various molecules, including peptides, DNA, RNA, and proteins. In this review, I summarize the progress in the construction of replication systems over the past few decades and discuss what we can learn from their construction. I introduce various types of replication systems, supporting the feasibility of the spontaneous appearance of replication early in Earth's history. In the latter part of the review, I focus on parasitic replicators, one of the largest obstacles for sustainable replication. Compartmentalization is discussed as a possible solution.
- 97Groaz, A.; Moghimianavval, H.; Tavella, F.; Giessen, T. W.; Vecchiarelli, A. G.; Yang, Q.; Liu, A. P. Engineering Spatiotemporal Organization and Dynamics in Synthetic Cells. WIREs Nanomed. Nanobiotechnol. 2021, 13 (3), e1685, DOI: 10.1002/wnan.1685Google ScholarThere is no corresponding record for this reference.
- 98Gaut, N. J.; Adamala, K. P. Reconstituting Natural Cell Elements in Synthetic Cells. Adv. Biol. 2021, 5 (3), e2000188, DOI: 10.1002/adbi.202000188Google ScholarThere is no corresponding record for this reference.
- 99Stal, L. J. Nitrogen Fixation in Cyanobacteria. In eLS; John Wiley & Sons, Ltd: Chichester, UK, 2015; pp 1– 9. DOI: 10.1002/9780470015902.a0021159.pub2 .Google ScholarThere is no corresponding record for this reference.
- 100Poirier, Y.; Antonenkov, V. D.; Glumoff, T.; Hiltunen, J. K. Peroxisomal β-oxidation─A Metabolic Pathway with Multiple Functions. Biochimica et Biophysica Acta (BBA) - Molecular Cell Research 2006, 1763 (12), 1413– 1426, DOI: 10.1016/j.bbamcr.2006.08.034Google Scholar100https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xhtlakur%252FK&md5=6d7a975e32e574ff04ead583cf4ff881Peroxisomal β-oxidation-A metabolic pathway with multiple functionsPoirier, Yves; Antonenkov, Vasily D.; Glumoff, Tuomo; Hiltunen, J. KalervoBiochimica et Biophysica Acta, Molecular Cell Research (2006), 1763 (12), 1413-1426CODEN: BBAMCO; ISSN:0167-4889. (Elsevier Ltd.)A review. Fatty acid degrdn. in most organisms occurs primarily via the β-oxidn. cycle. In mammals, β-oxidn. occurs in both mitochondria and peroxisomes, whereas plants and most fungi harbor the β-oxidn. cycle only in the peroxisomes. Although several of the enzymes participating in this pathway in both organelles are similar, some distinct physiol. roles have been uncovered. Recent advances in the structural elucidation of numerous mammalian and yeast enzymes involved in β-oxidn. have shed light on the basis of the substrate specificity for several of them. Of particular interest is the structural organization and function of the type 1 and 2 multifunctional enzyme (MFE-1 and MFE-2), two enzymes evolutionarily distant yet catalyzing the same overall enzymic reactions but via opposite stereochem. New data on the physiol. roles of the various enzymes participating in β-oxidn. have been gathered through the anal. of knockout mutants in plants, yeast and animals, as well as by the use of polyhydroxyalkanoate synthesis from β-oxidn. intermediates as a tool to study carbon flux through the pathway. In plants, both forward and reverse genetics performed on the model plant Arabidopsis thaliana have revealed novel roles for β-oxidn. in the germination process that is independent of the generation of carbohydrates for growth, as well as in embryo and flower development, and the generation of the phytohormone indole-3-acetic acid and the signal mol. jasmonic acid.
- 101Murat, D.; Byrne, M.; Komeili, A. Cell Biology of Prokaryotic Organelles. Cold Spring Harb. Perspect. Biol. 2010, 2 (10), a000422, DOI: 10.1101/cshperspect.a000422Google ScholarThere is no corresponding record for this reference.
- 102Nott, T. J.; Craggs, T. D.; Baldwin, A. J. Membraneless Organelles Can Melt Nucleic Acid Duplexes and Act as Biomolecular Filters. Nat. Chem. 2016, 8 (6), 569– 575, DOI: 10.1038/nchem.2519Google Scholar102https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XotV2nt70%253D&md5=0224be564579d21e8d4dd9c8bbc42fcdMembraneless organelles can melt nucleic acid duplexes and act as biomolecular filtersNott, Timothy J.; Craggs, Timothy D.; Baldwin, Andrew J.Nature Chemistry (2016), 8 (6), 569-575CODEN: NCAHBB; ISSN:1755-4330. (Nature Publishing Group)Membraneless organelles are cellular compartments made from drops of liq. protein inside a cell. These compartments assemble via the phase sepn. of disordered regions of proteins in response to changes in the cellular environment and the cell cycle. Here we demonstrate that the solvent environment within the interior of these cellular bodies behaves more like an org. solvent than like water. One of the most-stable biol. structures known, the DNA double helix, can be melted once inside the liq. droplet, and simultaneously structures formed from regulatory single-stranded nucleic acids are stabilized. Moreover, proteins are shown to have a wide range of absorption or exclusion from these bodies, and can act as importers for otherwise-excluded nucleic acids, which suggests the existence of a protein-mediated trafficking system. A common strategy in org. chem. is to utilize different solvents to influence the behavior of mols. and reactions. These results reveal that cells have also evolved this capability by exploiting the interiors of membraneless organelles.
- 103Lyon, A. S.; Peeples, W. B.; Rosen, M. K. A Framework for Understanding the Functions of Biomolecular Condensates across Scales. Nat. Rev. Mol. Cell Biol. 2021, 22 (3), 215– 235, DOI: 10.1038/s41580-020-00303-zGoogle Scholar103https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXit1yht7rP&md5=36e48922d2d86afd61d54380ef468ab5A framework for understanding the functions of biomolecular condensates across scalesLyon, Andrew S.; Peeples, William B.; Rosen, Michael K.Nature Reviews Molecular Cell Biology (2021), 22 (3), 215-235CODEN: NRMCBP; ISSN:1471-0072. (Nature Research)Abstr.: Biomol. condensates are found throughout eukaryotic cells, including in the nucleus, in the cytoplasm and on membranes. They are also implicated in a wide range of cellular functions, organizing mols. that act in processes ranging from RNA metab. to signalling to gene regulation. Early work in the field focused on identifying condensates and understanding how their phys. properties and regulation arise from mol. constituents. Recent years have brought a focus on understanding condensate functions. Studies have revealed functions that span different length scales: from mol. (modulating the rates of chem. reactions) to mesoscale (organizing large structures within cells) to cellular (facilitating localization of cellular materials and homeostatic responses). In this Roadmap, we discuss representative examples of biochem. and cellular functions of biomol. condensates from the recent literature and organize these functions into a series of non-exclusive classes across the different length scales. We conclude with a discussion of areas of current interest and challenges in the field, and thoughts about how progress may be made to further our understanding of the widespread roles of condensates in cell biol.
- 104Protter, D. S. W.; Parker, R. Principles and Properties of Stress Granules. Trends Cell Biol. 2016, 26 (9), 668– 679, DOI: 10.1016/j.tcb.2016.05.004Google Scholar104https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XosVSnsLw%253D&md5=799c56059a5fcd164ddb5fb50a9348cbPrinciples and Properties of Stress GranulesProtter, David S. W.; Parker, RoyTrends in Cell Biology (2016), 26 (9), 668-679CODEN: TCBIEK; ISSN:0962-8924. (Elsevier Ltd.)A review. Stress granules are assemblies of untranslating messenger ribonucleoproteins (mRNPs) that form from mRNAs stalled in translation initiation. Stress granules form through interactions between mRNA-binding proteins that link together populations of mRNPs. Interactions promoting stress granule formation include conventional protein-protein interactions as well as interactions involving intrinsically disordered regions (IDRs) of proteins. Assembly and disassembly of stress granules are modulated by various post-translational modifications as well as numerous ATP-dependent RNP or protein remodeling complexes, illustrating that stress granules represent an active liq. wherein energy input maintains their dynamic state. Stress granule formation modulates the stress response, viral infection, and signaling pathways. Persistent or aberrant stress granule formation contributes to neurodegenerative disease and some cancers.
- 105Roger, A. J.; Muñoz-Gómez, S. A.; Kamikawa, R. The Origin and Diversification of Mitochondria. Curr. Biol. 2017, 27 (21), R1177– R1192, DOI: 10.1016/j.cub.2017.09.015Google Scholar105https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhslygsL%252FO&md5=4611fac5e6db78c3eb8bb4bc313ceb32The Origin and Diversification of MitochondriaRoger, Andrew J.; Munoz-Gomez, Sergio A.; Kamikawa, RyomaCurrent Biology (2017), 27 (21), R1177-R1192CODEN: CUBLE2; ISSN:0960-9822. (Cell Press)A review. Mitochondria are best known for their role in the generation of ATP by aerobic respiration. Yet, research in the past half century has shown that they perform a much larger suite of functions and that these functions can vary substantially among diverse eukaryotic lineages. Despite this diversity, all mitochondria derive from a common ancestral organelle that originated from the integration of an endosymbiotic alphaproteobacterium into a host cell related to Asgard Archaea. The transition from endosymbiotic bacterium to permanent organelle entailed a massive no. of evolutionary changes including the origins of hundreds of new genes and a protein import system, insertion of membrane transporters, integration of metab. and reprodn., genome redn., endosymbiotic gene transfer, lateral gene transfer and the retargeting of proteins. These changes occurred incrementally as the endosymbiont and the host became integrated. Although many insights into this transition have been gained, controversy persists regarding the nature of the original endosymbiont, its initial interactions with the host and the timing of its integration relative to the origin of other features of eukaryote cells. Since the establishment of the organelle, proteins have been gained, lost, transferred and retargeted as mitochondria have specialized into the spectrum of functional types seen across the eukaryotic tree of life.
- 106Mehta, A. P.; Supekova, L.; Chen, J.-H.; Pestonjamasp, K.; Webster, P.; Ko, Y.; Henderson, S. C.; McDermott, G.; Supek, F.; Schultz, P. G. Engineering Yeast Endosymbionts as a Step toward the Evolution of Mitochondria. Proc. Natl. Acad. Sci. U. S. A. 2018, 115 (46), 11796– 11801, DOI: 10.1073/pnas.1813143115Google Scholar106https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXit1Sgt7fE&md5=fb74ba83e9dc4402b3d436758bafb90fEngineering yeast endosymbionts as a step toward the evolution of mitochondriaMehta, Angad P.; Supekova, Lubica; Chen, Jian-Hua; Pestonjamasp, Kersi; Webster, Paul; Ko, Yeonjin; Henderson, Scott C.; McDermott, Gerry; Supek, Frantisek; Schultz, Peter G.Proceedings of the National Academy of Sciences of the United States of America (2018), 115 (46), 11796-11801CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)It has been hypothesized that mitochondria evolved from a bacterial ancestor that initially became established in an archaeal host cell as an endosymbiont. Here we model this first stage of mitochondrial evolution by engineering endosymbiosis between Escherichia coli and Saccharomyces cerevisiae. An ADP/ATP translocase-expressing E. coli provided ATP to a respiration-deficient cox2 yeast mutant and enabled growth of a yeast-E. coli chimera on a nonfermentable carbon source. In a reciprocal fashion, yeast provided thiamin to an endosymbiotic E. coli thiamin auxotroph. Expression of several SNARE-like proteins in E. coli was also required, likely to block lysosomal degrdn. of intracellular bacteria. This chimeric system was stable for more than 40 doublings, and GFP-expressing E. coli endosymbionts could be obsd. in the yeast by fluorescence microscopy and X-ray tomog. This readily manipulated system should allow exptl. delineation of host-endosymbiont adaptations that occurred during evolution of the current, highly reduced mitochondrial genome.
- 107Headen, D. M.; Aubry, G.; Lu, H.; García, A. J. Microfluidic-Based Generation of Size-Controlled, Biofunctionalized Synthetic Polymer Microgels for Cell Encapsulation. Adv. Mater. 2014, 26 (19), 3003– 3008, DOI: 10.1002/adma.201304880Google Scholar107https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXktVWmtL8%253D&md5=9f60d129d74f04352d95959a640285f0Microfluidic-based generation of size-controlled, biofunctionalized synthetic polymer microgels for cell encapsulationHeaden, Devon M.; Aubry, Guillaume; Lu, Hang; Garcia, Andres J.Advanced Materials (Weinheim, Germany) (2014), 26 (19), 3003-3008CODEN: ADVMEW; ISSN:0935-9648. (Wiley-VCH Verlag GmbH & Co. KGaA)The high potential of synthetic hydrogel microencapsulation for cell and protein therapeutics has been limited by the lack of synthetic polymer systems with tunable capsule size, cytocompatible crosslinking reactions, rapid crosslinking rates, adequate biomol. permeability, and ease of functionalization with bioactive mols. (e.g., adhesive peptides). Using a synthetic hydrogel system with tunable network and crosslinking characteristics and a microfluidics encapsulation platform, we have created an integrated and robust strategy for microencapsulation of cells in which we can control capsule size and local cellular microenvironment. Addnl., microgel network structure can be tuned to optimize permeability of the capsule to mols. of various sizes. We have demonstrated, proof of concept with two different clin. relevant human cell types, but the versatility of this strategy will allow it to be tailored to fit diverse engineering applications.
- 108Torre, P.; Keating, C. D.; Mansy, S. S. Multiphase Water-in-Oil Emulsion Droplets for Cell-Free Transcription-Translation. Langmuir 2014, 30 (20), 5695– 5699, DOI: 10.1021/la404146gGoogle Scholar108https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXns1Cns7w%253D&md5=d1aa45d707f57c13621a12046cc04eb0Multiphase water-in-oil emulsion droplets for cell-free transcription-translationTorre, Paola; Keating, Christine D.; Mansy, Sheref S.Langmuir (2014), 30 (20), 5695-5699CODEN: LANGD5; ISSN:0743-7463. (American Chemical Society)The construction of genetically encoded cellular mimics in compartments contg. organized synthetic cytosols is desirable for the development of artificial cells. Phase sepd. aq. domains were placed within water-in-oil emulsion droplets in a manner compatible with transcription and translation machinery. Aq. two-phase and three-phase systems (ATPS and A3PS) were assembled with dextran, poly(ethylene glycol), and Ficoll. Aq. two-phase systems were capable of supporting the cell-free expression of protein within water droplets, whereas the aq. three-phase-based system did not give rise to detectable protein synthesis. The expressed protein preferentially partitioned to the dextran-enriched phase. The system could serve as a foundation for building cellular mimics with liq. organelles.
- 109Zhang, Y.; Kojima, T.; Kim, G.-A.; McNerney, M. P.; Takayama, S.; Styczynski, M. P. Protocell Arrays for Simultaneous Detection of Diverse Analytes. Nat. Commun. 2021, 12 (1), 5724, DOI: 10.1038/s41467-021-25989-3Google Scholar109https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXitFCgtLjO&md5=310fc2fe6fcb267fbcf136e7d1951810Protocell arrays for simultaneous detection of diverse analytesZhang, Yan; Kojima, Taisuke; Kim, Ge-Ah; McNerney, Monica P.; Takayama, Shuichi; Styczynski, Mark P.Nature Communications (2021), 12 (1), 5724CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)Simultaneous detection of multiple analytes from a single sample (multiplexing), particularly when done at the point of need, can guide complex decision-making without increasing the required sample vol. or cost per test. Despite recent advances, multiplexed analyte sensing still typically faces the crit. limitation of measuring only one type of mol. (e.g., small mols. or nucleic acids) per assay platform. Here, we address this bottleneck with a customizable platform that integrates cell-free expression (CFE) with a polymer-based aq. two-phase system (ATPS), producing membrane-less protocells contg. transcription and translation machinery used for detection. We show that multiple protocells, each performing a distinct sensing reaction, can be arrayed in the same microwell to detect chem. diverse targets from the same sample. Furthermore, these protocell arrays are compatible with human biofluids, maintain function after lyophilization and rehydration, and can produce visually interpretable readouts, illustrating this platforms potential as a minimal-equipment, field-deployable, multi-analyte detection tool.
- 110Allen, M. E.; Hindley, J. W.; Baxani, D. K.; Ces, O.; Elani, Y. Hydrogels as Functional Components in Artificial Cell Systems. Nat. Rev. Chem. 2022, 6 (8), 562– 578, DOI: 10.1038/s41570-022-00404-7Google Scholar110https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhvFGnsbbK&md5=a18a79ffa095b7ee78a973929cdd92a2Hydrogels as functional components in artificial cell systemsAllen, Matthew E.; Hindley, James W.; Baxani, Divesh K.; Ces, Oscar; Elani, YuvalNature Reviews Chemistry (2022), 6 (8), 562-578CODEN: NRCAF7; ISSN:2397-3358. (Nature Portfolio)A review. Recent years have seen substantial efforts aimed at constructing artificial cells from various mol. components with the aim of mimicking the processes, behaviors and architectures found in biol. systems. Artificial cell development ultimately aims to produce model constructs that progress our understanding of biol., as well as forming the basis for functional bio-inspired devices that can be used in fields such as therapeutic delivery, biosensing, cell therapy and bioremediation. Typically, artificial cells rely on a bilayer membrane chassis and have fluid aq. interiors to mimic biol. cells. However, a desire to more accurately replicate the gel-like properties of intracellular and extracellular biol. environments has driven increasing efforts to build cell mimics based on hydrogels. This has enabled researchers to exploit some of the unique functional properties of hydrogels that have seen them deployed in fields such as tissue engineering, biomaterials and drug delivery. In this Review, we explore how hydrogels can be leveraged in the context of artificial cell development. We also discuss how hydrogels can potentially be incorporated within the next generation of artificial cells to engineer improved biol. mimics and functional microsystems.
- 111Has, C.; Sunthar, P. A Comprehensive Review on Recent Preparation Techniques of Liposomes. J. Liposome Res. 2020, 30 (4), 336– 365, DOI: 10.1080/08982104.2019.1668010Google Scholar111https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhvVGntL3N&md5=8a224f78b2356da1539e1a35bad7b076A comprehensive review on recent preparation techniques of liposomesHas, C.; Sunthar, P.Journal of Liposome Research (2020), 30 (4), 336-365CODEN: JLREE7; ISSN:0898-2104. (Taylor & Francis Ltd.)A review Liposomes (or lipid vesicles) are a versatile platform as carriers for the delivery of the drugs and other macromols. into human and animal bodies. Though the method of using liposomes has been known since 1960s, and major developments and commercialization of liposomal formulations took place in the late nineties (or early part of this century), newer methods of liposome synthesis and drug encapsulation continue to be an active area of research. With the developments in related fields, such as electrohydrodynamics and microfluidics, and a growing understanding of the mechanisms of lipid assembly from colloidal and intermol. forces, liposome prepn. techniques have been enriched and more predictable than before. This has led to better methods that can also scale at an industrial prodn. level. In this review, we present several novel methods that were introduced over the last decade and compare their advantages over conventional methods. Researchers beginning to explore liposomal formulations will find this resource useful to give an overall direction for an appropriate choice of method. Where possible, we have also provided the known mechanisms behind the prepn. methods.
- 112Luisi, P. L.; Allegretti, M.; Pereira de Souza, T.; Steiniger, F.; Fahr, A.; Stano, P. Spontaneous Protein Crowding in Liposomes: A New Vista for the Origin of Cellular Metabolism. Chembiochem 2010, 11 (14), 1989– 1992, DOI: 10.1002/cbic.201000381Google ScholarThere is no corresponding record for this reference.
- 113Michelon, M.; Huang, Y.; de la Torre, L. G.; Weitz, D. A.; Cunha, R. L. Single-Step Microfluidic Production of W/O/W Double Emulsions as Templates for β-Carotene-Loaded Giant Liposomes Formation. Chem. Eng. J. 2019, 366, 27– 32, DOI: 10.1016/j.cej.2019.02.021Google Scholar113https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXjtFOns7Y%253D&md5=392e7b1f109abc07e1a4137388de4360Single-step microfluidic production of W/O/W double emulsions as templates for β-carotene-loaded giant liposomes formationMichelon, Mariano; Huang, Yuting; de la Torre, Lucimara Gaziola; Weitz, David A.; Cunha, Rosiane LopesChemical Engineering Journal (Amsterdam, Netherlands) (2019), 366 (), 27-32CODEN: CMEJAJ; ISSN:1385-8947. (Elsevier B.V.)We demonstrated the microfluidic prodn. of W/O/W double emulsion droplets aiming formation of β-carotene-incorporated giant liposomes for food and/or pharmaceutical applications. For this purpose, glass-capillary microfluidic devices were fabricated to create a truly three-dimensional flow aiming prodn. of giant unilamellar liposomes by solvent evapn. process after W/O/W double emulsion droplet templates formation. A great challenge of microfluidic prodn. of monodisperse and stable W/O/W double emulsion templates for this proposal is the replacement of org. solvents potentially toxic for phospholipids dissoln. Besides, the high cost of several semi-synthetic phospholipids commonly used for giant liposome formation remains as a major technol. challenge to be overcome. Thus, β-carotene-incorporated giant liposomes were generated using biocompatible solvents with low toxic potential (Et acetate and pentane) and non-purified soybean lecithin - a food-grade phospholipid mixt. with low cost - by dewetting and evapn. of the solvents forming the oily intermediate phase of W/O/W double emulsion droplet templates. Our results showed monodisperse β-carotene-loaded giant liposomes with diam. ranging between 100 μm and 180 μm and a stability of approx. 7 days. In this way, a single-step microfluidic process with highly accurate control of size distribution was developed. This microfluidic process proposed is potentially useful for a broad range of applications in protection and delivery of active compds.
- 114Elani, Y.; Trantidou, T.; Wylie, D.; Dekker, L.; Polizzi, K.; Law, R. V.; Ces, O. Constructing Vesicle-Based Artificial Cells with Embedded Living Cells as Organelle-like Modules. Sci. Rep. 2018, 8 (1), 4564, DOI: 10.1038/s41598-018-22263-3Google Scholar114https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1Mnht1yisw%253D%253D&md5=8e62e29719807e28d859d2c4d4edfe5bConstructing vesicle-based artificial cells with embedded living cells as organelle-like modulesElani Yuval; Trantidou Tatiana; Wylie Douglas; Law Robert V; Ces Oscar; Elani Yuval; Wylie Douglas; Ces Oscar; Dekker Linda; Polizzi KarenScientific reports (2018), 8 (1), 4564 ISSN:.There is increasing interest in constructing artificial cells by functionalising lipid vesicles with biological and synthetic machinery. Due to their reduced complexity and lack of evolved biochemical pathways, the capabilities of artificial cells are limited in comparison to their biological counterparts. We show that encapsulating living cells in vesicles provides a means for artificial cells to leverage cellular biochemistry, with the encapsulated cells serving organelle-like functions as living modules inside a larger synthetic cell assembly. Using microfluidic technologies to construct such hybrid cellular bionic systems, we demonstrate that the vesicle host and the encapsulated cell operate in concert. The external architecture of the vesicle shields the cell from toxic surroundings, while the cell acts as a bioreactor module that processes encapsulated feedstock which is further processed by a synthetic enzymatic metabolism co-encapsulated in the vesicle.
- 115Tayeb, H. H.; Sainsbury, F. Nanoemulsions in Drug Delivery: Formulation to Medical Application. Nanomedicine 2018, 13 (19), 2507– 2525, DOI: 10.2217/nnm-2018-0088Google Scholar115https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitV2isL7F&md5=10a78be18038f4a9b8a01afa21fe96feNanoemulsions in drug delivery: formulation to medical applicationTayeb, Hossam H.; Sainsbury, FrankNanomedicine (London, United Kingdom) (2018), 13 (19), 2507-2525CODEN: NLUKAC; ISSN:1748-6963. (Future Medicine Ltd.)Nanoscale oil-in-water emulsions (NEs), heterogeneous systems of two immiscible liqs. stabilized by emulsifiers or surfactants, show great potential in medical applications because of their attractive characteristics for drug delivery. NEs have been explored as therapeutic carriers for hydrophobic compds. via various routes of administration. NEs provide opportunities to improve drug delivery via alternative administration routes. However, deep understanding of the NE manufg. and functionalization fundamentals, and how they relate to the choice of administration route and pharmacol. profile is still needed to ease the clin. translation of NEs. Here, we review the diversity of medical applications for NEs and how that governs their formulation, route of administration, and the emergence of increasing sophistication in NE design for specific application.
- 116Vogele, K.; Frank, T.; Gasser, L.; Goetzfried, M. A.; Hackl, M. W.; Sieber, S. A.; Simmel, F. C.; Pirzer, T. Towards Synthetic Cells Using Peptide-Based Reaction Compartments. Nat. Commun. 2018, 9 (1), 3862, DOI: 10.1038/s41467-018-06379-8Google Scholar116https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3czhsV2jsA%253D%253D&md5=7ee8f4968fc15a261b9d2deb5b7d789aTowards synthetic cells using peptide-based reaction compartmentsVogele Kilian; Frank Thomas; Gasser Lukas; Goetzfried Marisa A; Simmel Friedrich C; Pirzer Tobias; Hackl Mathias W; Sieber Stephan A; Simmel Friedrich CNature communications (2018), 9 (1), 3862 ISSN:.Membrane compartmentalization and growth are central aspects of living cells, and are thus encoded in every cell's genome. For the creation of artificial cellular systems, genetic information and production of membrane building blocks will need to be coupled in a similar manner. However, natural biochemical reaction networks and membrane building blocks are notoriously difficult to implement in vitro. Here, we utilized amphiphilic elastin-like peptides (ELP) to create self-assembled vesicular structures of about 200 nm diameter. In order to genetically encode the growth of these vesicles, we encapsulate a cell-free transcription-translation system together with the DNA template inside the peptide vesicles. We show in vesiculo production of a functioning fluorescent RNA aptamer and a fluorescent protein. Furthermore, we implement in situ expression of the membrane peptide itself and finally demonstrate autonomous vesicle growth due to the incorporation of this ELP into the membrane.
- 117Mushnoori, S.; Lu, C. Y.; Schmidt, K.; Zang, E.; Dutt, M. Peptide-Based Vesicles and Droplets: A Review. J. Phys.: Condens. Matter 2020, 33 (5), 053002, DOI: 10.1088/1361-648X/abb995Google ScholarThere is no corresponding record for this reference.
- 118Sharma, B.; Ma, Y.; Hiraki, H. L.; Baker, B. M.; Ferguson, A. L.; Liu, A. P. Facile Formation of Giant Elastin-like Polypeptide Vesicles as Synthetic Cells. Chem. Commun. 2021, 57 (97), 13202– 13205, DOI: 10.1039/D1CC05579HGoogle Scholar118https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXisFWlu7nI&md5=210d236dd96e6fcbdf19e6c8f2042f95Facile formation of giant elastin-like polypeptide vesicles as synthetic cellsSharma, Bineet; Ma, Yutao; Hiraki, Harrison L.; Baker, Brendon M.; Ferguson, Andrew L.; Liu, Allen P.Chemical Communications (Cambridge, United Kingdom) (2021), 57 (97), 13202-13205CODEN: CHCOFS; ISSN:1359-7345. (Royal Society of Chemistry)We demonstrate the facile and robust generation of giant peptide vesicles by using an emulsion transfer method. These robust vesicles can sustain chem. and phys. stresses. The peptide vesicles can host cell-free expression reactions by encapsulating essential ingredients. We show the incorporation of another cell-free expressed elastin-like polypeptide into the existing membrane of the peptide vesicles.
- 119Vieregg, J. R.; Lueckheide, M.; Marciel, A. B.; Leon, L.; Bologna, A. J.; Rivera, J. R.; Tirrell, M. V. Oligonucleotide-Peptide Complexes: Phase Control by Hybridization. J. Am. Chem. Soc. 2018, 140 (5), 1632– 1638, DOI: 10.1021/jacs.7b03567Google Scholar119https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXlvVChsQ%253D%253D&md5=7c78b1f95043da89c34c10a3d3543529Oligonucleotide-peptide complexes: Phase control by hybridizationVieregg, Jeffrey R.; Lueckheide, Michael; Marciel, Amanda B.; Leon, Lorraine; Bologna, Alex J.; Rivera, Josean Reyes; Tirrell, Matthew V.Journal of the American Chemical Society (2018), 140 (5), 1632-1638CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)When oppositely charged polymers are mixed, counterion release drives phase sepn.; understanding this process is a key unsolved problem in polymer science and biophys. chem., particularly for nucleic acids, polyanions whose biol. functions are intimately related to their high charge d. In the cell, complexation by basic proteins condenses DNA into chromatin, and membraneless organelles formed by liq.-liq. phase sepn. of RNA and proteins perform vital functions and have been linked to disease. Electrostatic interactions are also the primary method used for assembly of nanoparticles to deliver therapeutic nucleic acids into cells. This work describes complexation expts. with oligonucleotides and cationic peptides spanning a wide range of polymer lengths, concns., and structures, including RNA and methylphosphonate backbones. We find that the phase of the complexes is controlled by the hybridization state of the nucleic acid, with double-stranded nucleic acids forming solid ppts. while single-stranded oligonucleotides form liq. coacervates, apparently due to their lower charge d. Adding salt "melts" ppts. into coacervates, and oligonucleotides in coacervates remain competent for sequence-specific hybridization and phase change, suggesting the possibility of environmentally responsive complexes and nanoparticles for therapeutic or sensing applications.
- 120Fraccia, T. P.; Jia, T. Z. Liquid Crystal Coacervates Composed of Short Double-Stranded DNA and Cationic Peptides. ACS Nano 2020, 14 (11), 15071– 15082, DOI: 10.1021/acsnano.0c05083Google Scholar120https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhs12rtr7L&md5=04619533c8c1cacce2a6b7f7dd591b6eLiquid Crystal Coacervates Composed of Short Double-Stranded DNA and Cationic PeptidesFraccia, Tommaso P.; Jia, Tony Z.ACS Nano (2020), 14 (11), 15071-15082CODEN: ANCAC3; ISSN:1936-0851. (American Chemical Society)Phase sepn. of nucleic acids and proteins is a ubiquitous phenomenon regulating subcellular compartment structure and function. While complex coacervation of flexible single-stranded nucleic acids is broadly investigated, coacervation of double-stranded DNA (dsDNA) is less studied because of its propensity to generate solid ppts. Here, we reverse this perspective by showing that short dsDNA and poly-L-lysine coacervates can escape pptn. while displaying a surprisingly complex phase diagram, including the full set of liq. crystal (LC) mesophases obsd. to date in bulk dsDNA. Short dsDNA supramol. aggregation and packing in the dense coacervate phase are the main parameters regulating the global LC-coacervate phase behavior. LC-coacervate structure was characterized upon variations in temp. and monovalent salt, DNA, and peptide concns., which allow continuous reversible transitions between all accessible phases. A deeper understanding of LC-coacervates can gain insights to decipher structures and phase transition mechanisms within biomol. condensates, to design stimuli-responsive multiphase synthetic compartments with different degrees of order and to exploit self-assembly driven cooperative prebiotic evolution of nucleic acids and peptides.
- 121Aumiller, W. M., Jr; Pir Cakmak, F.; Davis, B. W.; Keating, C. D. RNA-Based Coacervates as a Model for Membraneless Organelles: Formation, Properties, and Interfacial Liposome Assembly. Langmuir 2016, 32 (39), 10042– 10053, DOI: 10.1021/acs.langmuir.6b02499Google Scholar121https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsVKrtrnO&md5=1298cfe8fb86980343543917b9831be1RNA-Based Coacervates as a Model for Membraneless Organelles: Formation, Properties, and Interfacial Liposome AssemblyAumiller, William M.; Pir Cakmak, Fatma; Davis, Bradley W.; Keating, Christine D.Langmuir (2016), 32 (39), 10042-10053CODEN: LANGD5; ISSN:0743-7463. (American Chemical Society)Liq.-liq. phase sepn. is responsible for the formation of P granules, nucleoli, and other membraneless subcellular organelles composed of RNA and proteins. Efforts to understand the phys. basis of liq. organelle formation have thus far focused on intrinsically disordered proteins (IDPs) as major components that dictate occurrence and properties. Here, the authors show that complex coacervates composed of low complexity RNA [polyuridylic acid, poly(U)] and short polyamines (spermine and spermidine) share many features of IDP-based coacervates. Poly(U)/polyamine coacervates compartmentalized biomols. (peptides, oligonucleotides) in a sequence- and length- dependent manner. These solutes retained mobility within the coacervate droplets, as demonstrated by rapid recovery from photobleaching. Coacervation was reversible with changes in soln. temp. due to changes in the poly(U) structure that impacted its interactions with polyamines. The authors further demonstrated that lipid vesicles assembled at the droplet interface without impeding RNA entry/egress. These vesicles remained intact at the interface and could be released upon temp.-induced droplet dissoln.
- 122Rimoli, M. G.; Rabaioli, M. R.; Melisi, D.; Curcio, A.; Mondello, S.; Mirabelli, R.; Abignente, E. Synthetic Zeolites as a New Tool for Drug Delivery. J. Biomed. Mater. Res., Part A 2008, 87 (1), 156– 164, DOI: 10.1002/jbm.a.31763Google Scholar122https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhtFCqsL7K&md5=9d159c5f96d5a848b766aa5e118a087aSynthetic zeolites as a new tool for drug deliveryRimoli, Maria G.; Rabaioli, Maria R.; Melisi, Daniela; Curcio, Annalisa; Mondello, Sandro; Mirabelli, Rosella; Abignente, EnricoJournal of Biomedical Materials Research, Part A (2008), 87A (1), 156-164CODEN: JBMRCH; ISSN:1549-3296. (John Wiley & Sons, Inc.)Synthetic zeolites were studied in order to investigate their ability to encapsulate and to release drugs. In particular, a zeolite X and a zeolitic product obtained from a cocrystn. of zeolite X and zeolite A were examd. These materials were characterized by chem. analyses (ICP-AES), x-ray diffraction, nitrogen adsorption isotherm, SEM, laser diffraction, and IR spectroscopy. Since ketoprofen was chosen as a model drug for the formulation of controlled-release dosage forms, it was encapsulated into these two types of synthetic zeolites by a soaking procedure. Drug-loaded matrixes were then characterized for entrapped drug amt. and thermogravimetric behavior. In both types of activated zeolites, the total amt. of ketoprofen (800 mg) was encapsulated in 2 g of matrix. By using HPLC measurements, ketoprofen release studies were done at different pH conditions so as to mimic gastrointestinal fluids. The absence of release in acid conditions and a double phased release, at two different pH values (5 and 6.8), suggest that after activation these materials offer good potential for a modified release delivery system of ketoprofen.
- 123Hyman, A. A.; Weber, C. A.; Jülicher, F. Liquid-Liquid Phase Separation in Biology. Annu. Rev. Cell Dev. Biol. 2014, 30, 39– 58, DOI: 10.1146/annurev-cellbio-100913-013325Google Scholar123https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXitVeit7vL&md5=a4d83a5d473be634e6a4d9bdbcc6e63aLiquid-liquid phase separation in biologyHyman, Anthony A.; Weber, Christoph A.; Juelicher, FrankAnnual Review of Cell and Developmental Biology (2014), 30 (), 39-58CODEN: ARDBF8; ISSN:1081-0706. (Annual Reviews)A review. Cells organize many of their biochem. reactions in non-membrane compartments. Recent evidence showed that many of these compartments are liqs. that form by phase sepn. from the cytoplasm. Here the basic phys. concepts necessary to understand the consequences of liq.-like states for biol. functions are discussed.
- 124Junge, F.; Haberstock, S.; Roos, C.; Stefer, S.; Proverbio, D.; Dötsch, V.; Bernhard, F. Advances in Cell-Free Protein Synthesis for the Functional and Structural Analysis of Membrane Proteins. N. Biotechnol. 2011, 28 (3), 262– 271, DOI: 10.1016/j.nbt.2010.07.002Google ScholarThere is no corresponding record for this reference.
- 125Lee, K. Y.; Park, S.-J.; Lee, K. A.; Kim, S.-H.; Kim, H.; Meroz, Y.; Mahadevan, L.; Jung, K.-H.; Ahn, T. K.; Parker, K. K.; Shin, K. Photosynthetic Artificial Organelles Sustain and Control ATP-Dependent Reactions in a Protocellular System. Nat. Biotechnol. 2018, 36 (6), 530– 535, DOI: 10.1038/nbt.4140Google Scholar125https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtVWktrzM&md5=38cc9b7aa604f285b5d84c793c9e26cfPhotosynthetic artificial organelles sustain and control ATP-dependent reactions in a protocellular systemLee, Keel Yong; Park, Sung-Jin; Lee, Keon Ah; Kim, Se-Hwan; Kim, Heeyeon; Meroz, Yasmine; Mahadevan, L.; Jung, Kwang-Hwan; Ahn, Tae Kyu; Parker, Kevin Kit; Shin, KwanwooNature Biotechnology (2018), 36 (6), 530-535CODEN: NABIF9; ISSN:1087-0156. (Nature Research)Inside cells, complex metabolic reactions are distributed across the modular compartments of organelles. Reactions in organelles have been recapitulated in vitro by reconstituting functional protein machineries into membrane systems. However, maintaining and controlling these reactions is challenging. Here we designed, built, and tested a switchable, light-harvesting organelle that provides both a sustainable energy source and a means of directing intravesicular reactions. An ATP (ATP) synthase and two photoconverters (plant-derived photosystem II and bacteria-derived proteorhodopsin) enable ATP synthesis. Independent optical activation of the two photoconverters allows dynamic control of ATP synthesis: red light facilitates and green light impedes ATP synthesis. We encapsulated the photosynthetic organelles in a giant vesicle to form a protocellular system and demonstrated optical control of two ATP-dependent reactions, carbon fixation and actin polymn., with the latter altering outer vesicle morphol. Switchable photosynthetic organelles may enable the development of biomimetic vesicle systems with regulatory networks that exhibit homeostasis and complex cellular behaviors.
- 126Jewett, M. C.; Swartz, J. R. Mimicking the Escherichia Coli Cytoplasmic Environment Activates Long-Lived and Efficient Cell-Free Protein Synthesis. Biotechnol. Bioeng. 2004, 86 (1), 19– 26, DOI: 10.1002/bit.20026Google Scholar126https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXis1aksrY%253D&md5=e8d45540857fdb7b7b269e2704afb08cMimicking the Escherichia coli cytoplasmic environment activates long-lived and efficient cell-free protein synthesisJewett, Michael C.; Swartz, James R.Biotechnology and Bioengineering (2004), 86 (1), 19-26CODEN: BIBIAU; ISSN:0006-3592. (John Wiley & Sons, Inc.)Cell-free translation systems generally utilize high-energy phosphate compds. to regenerate the ATP necessary to drive protein synthesis. This hampers the widespread use and practical implementation of this technol. in a batch format due to expensive reagent costs; the accumulation of inhibitory byproducts, such as phosphate; and pH change. To address these problems, a cell-free protein synthesis system has been engineered that is capable of using pyruvate as an energy source to produce high yields of protein. The "Cytomim" system, synthesizes chloramphenicol acetyl-transferase (CAT) for up to 6 h in a batch reaction to yield 700 μg/mL of protein. By more closely replicating the physiol. conditions of the cytoplasm of Escherichia coli, the Cytomim system provides a stable energy supply for protein expression without phosphate accumulation, pH change, exogenous enzyme addn., or the need for expensive high-energy phosphate compds.
- 127Caschera, F.; Noireaux, V. Synthesis of 2.3 Mg/mL of Protein with an All Escherichia Coli Cell-Free Transcription-Translation System. Biochimie 2014, 99, 162– 168, DOI: 10.1016/j.biochi.2013.11.025Google Scholar127https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhvFOhtrjM&md5=7d951478b25a8eaaba5a48e3ca59522cSynthesis of 2.3 mg/ml of protein with an all Escherichia coli cell-free transcription-translation systemCaschera, Filippo; Noireaux, VincentBiochimie (2014), 99 (), 162-168CODEN: BICMBE; ISSN:0300-9084. (Elsevier Masson SAS)Cell-free protein synthesis is becoming a useful technique for synthetic biol. As more applications are developed, the demand for novel and more powerful in vitro expression systems is increasing. In this work, an all Escherichia coli cell-free system, that uses the endogenous transcription and translation mol. machineries, is optimized to synthesize up to 2.3 mg/mL of a reporter protein in batch mode reactions. A new metab. based on maltose allows recycling of inorg. phosphate through its incorporation into newly available glucose mols., which are processed through the glycolytic pathway to produce more ATP. As a result, the ATP regeneration is more efficient and cell-free protein synthesis lasts up to 10 h. Using a com. E. coli strain, we show for the first time that more than 2 mg/mL of protein can be synthesized in run-off cell-free transcription-translation reactions by optimizing the energy regeneration and waste products recycling. This work suggests that endogenous enzymes present in the cytoplasmic ext. can be used to implement new metabolic pathways for increasing protein yields. This system is the new basis of a cell-free gene expression platform used to construct and to characterize complex biochem. processes in vitro such as gene circuits.
- 128Garamella, J.; Marshall, R.; Rustad, M.; Noireaux, V. The All E. Coli TX-TL Toolbox 2.0: A Platform for Cell-Free Synthetic Biology. ACS Synth. Biol. 2016, 5 (4), 344– 355, DOI: 10.1021/acssynbio.5b00296Google Scholar128https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsFGqu74%253D&md5=609156affc1417f89485cfc7b001d95cThe All E. coli TX-TL Toolbox 2.0: A Platform for Cell-Free Synthetic BiologyGaramella, Jonathan; Marshall, Ryan; Rustad, Mark; Noireaux, VincentACS Synthetic Biology (2016), 5 (4), 344-355CODEN: ASBCD6; ISSN:2161-5063. (American Chemical Society)We report on and provide a detailed characterization of the performance and properties of a recently developed, all Escherichia coli, cell-free transcription and translation system. Gene expression is entirely based on the endogenous translation components and transcription machinery provided by an E. coli cytoplasmic ext., thus expanding the repertoire of regulatory parts to hundreds of elements. We use a powerful metab. for ATP regeneration to achieve more than 2 mg/mL of protein synthesis in batch mode reactions, and more than 6 mg/mL in semicontinuous mode. While the strength of cell-free expression is increased by a factor of 3 on av., the output signal of simple gene circuits and the synthesis of entire bacteriophages are increased by orders of magnitude compared to previous results. MRNAs and protein degrdn., resp. tuned using E. coli MazF interferase and ClpXP AAA+ proteases, are characterized over a much wider range of rates than the first version of the cell-free toolbox. This system is a highly versatile cell-free platform to construct complex biol. systems through the execution of DNA programs composed of synthetic and natural bacterial regulatory parts.
- 129Kim, D.-M.; Swartz, J. R. Efficient Production of a Bioactive, Multiple Disulfide-Bonded Protein Using Modified Extracts of Escherichia Coli. Biotechnol. Bioeng. 2004, 85 (2), 122– 129, DOI: 10.1002/bit.10865Google ScholarThere is no corresponding record for this reference.
- 130Jeong, S.; Nguyen, H. T.; Kim, C. H.; Ly, M. N.; Shin, K. Toward Artificial Cells: Novel Advances in Energy Conversion and Cellular Motility. Adv. Funct. Mater. 2020, 30 (11), 1907182, DOI: 10.1002/adfm.201907182Google Scholar130https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvFSls70%253D&md5=ff8cbe75b127ecc70c73f49fd1075be0Toward Artificial Cells: Novel Advances in Energy Conversion and Cellular MotilityJeong, Sungwoo; Nguyen, Huong Thanh; Kim, Chang Ho; Ly, Mai Nguyet; Shin, KwanwooAdvanced Functional Materials (2020), 30 (11), 1907182CODEN: AFMDC6; ISSN:1616-301X. (Wiley-VCH Verlag GmbH & Co. KGaA)A review. The demand to discover every single cellular component has been continuously increasing along with the development of biol. techniques. The bottom-up approach to construct a cell-mimicking system from well-defined and tunable compns. is accelerating, with the ultimate goal of comprehending a biol. cell. From among the available techniques, the artificial cell has been increasingly recognized as one of the most powerful tools for building a cell-like system from scratch. This review summarizes the development of artificial cells, from a pure giant unilamellar vesicle (GUV) to a controllable, self-fueled proteoliposome, both of which are highly compartmentalized. The basic components of an artificial cell, as well as the optimal conditions required for successful, reproducible GUV formation and protein reconstitution, are discussed. Most importantly, progress in studying the metabolic reactions in and the motility of a reconstituted artificial cell are the main focus of the review. The ability to perform a complicated reaction cascade in a controllable manner is highlighted as a promising perspective to obtaining an autonomous and movable GUV.
- 131Hanczyc, M. M.; Fujikawa, S. M.; Szostak, J. W. Experimental Models of Primitive Cellular Compartments: Encapsulation, Growth, and Division. Science 2003, 302 (5645), 618– 622, DOI: 10.1126/science.1089904Google Scholar131https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXotlWqt74%253D&md5=647b217542d67ffe265a705e5231f180Experimental Models of Primitive Cellular Compartments: Encapsulation, Growth, and DivisionHanczyc, Martin M.; Fujikawa, Shelly M.; Szostak, Jack W.Science (Washington, DC, United States) (2003), 302 (5645), 618-622CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)The clay montmorillonite is known to catalyze the polymn. of RNA from activated ribonucleotides. Here we report that montmorillonite accelerates the spontaneous conversion of fatty acid micelles into vesicles. Clay particles often become encapsulated in these vesicles, thus providing a pathway for the prebiotic encapsulation of catalytically active surfaces within membrane vesicles. In addn., RNA adsorbed to clay can be encapsulated within vesicles. Once formed, such vesicles can grow by incorporating fatty acid supplied as micelles and can divide without diln. of their contents by extrusion through small pores. These processes mediate vesicle replication through cycles of growth and division. The formation, growth, and division of the earliest cells may have occurred in response to similar interactions with mineral particles and inputs of material and energy.
- 132Kretschmer, S.; Ganzinger, K. A.; Franquelim, H. G.; Schwille, P. Synthetic Cell Division via Membrane-Transforming Molecular Assemblies. BMC Biol. 2019, 17 (1), 43, DOI: 10.1186/s12915-019-0665-1Google Scholar132https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3M7psFSntg%253D%253D&md5=834c319f13f711a883cb26e659512c92Synthetic cell division via membrane-transforming molecular assembliesKretschmer Simon; Ganzinger Kristina A; Franquelim Henri G; Schwille Petra; Kretschmer Simon; Ganzinger Kristina ABMC biology (2019), 17 (1), 43 ISSN:.Reproduction, i.e. the ability to produce new individuals from a parent organism, is a hallmark of living matter. Even the simplest forms of reproduction require cell division: attempts to create a designer cell therefore should include a synthetic cell division machinery. In this review, we will illustrate how nature solves this task, describing membrane remodelling processes in general and focusing on bacterial cell division in particular. We discuss recent progress made in their in vitro reconstitution, identify open challenges, and suggest how purely synthetic building blocks could provide an additional and attractive route to creating artificial cell division machineries.
- 133Anzai, K.; Yoshida, M.; Kirino, Y. Change in Intravesicular Volume of Liposomes by Freeze-Thaw Treatment as Studied by the ESR Stopped-Flow Technique. Biochimica et Biophysica Acta (BBA) - Biomembranes 1990, 1021 (1), 21– 26, DOI: 10.1016/0005-2736(90)90378-2Google ScholarThere is no corresponding record for this reference.
- 134van der Valk, T.; Pečnerová, P.; Díez-Del-Molino, D.; Bergström, A.; Oppenheimer, J.; Hartmann, S.; Xenikoudakis, G.; Thomas, J. A.; Dehasque, M.; Sağlıcan, E.; Fidan, F. R.; Barnes, I.; Liu, S.; Somel, M.; Heintzman, P. D.; Nikolskiy, P.; Shapiro, B.; Skoglund, P.; Hofreiter, M.; Lister, A. M.; Götherström, A.; Dalén, L. Million-Year-Old DNA Sheds Light on the Genomic History of Mammoths. Nature 2021, 591 (7849), 265– 269, DOI: 10.1038/s41586-021-03224-9Google Scholar134https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXktlGhuro%253D&md5=a3cb5a5555d6390029f7aa91ec3671f2Million-year-old DNA sheds light on the genomic history of mammothsvan der Valk, Tom; Pecnerova, Patricia; Diez-del-Molino, David; Bergstroem, Anders; Oppenheimer, Jonas; Hartmann, Stefanie; Xenikoudakis, Georgios; Thomas, Jessica A.; Dehasque, Marianne; Saglican, Ekin; Fidan, Fatma Rabia; Barnes, Ian; Liu, Shanlin; Somel, Mehmet; Heintzman, Peter D.; Nikolskiy, Pavel; Shapiro, Beth; Skoglund, Pontus; Hofreiter, Michael; Lister, Adrian M.; Goetherstroem, Anders; Dalen, LoveNature (London, United Kingdom) (2021), 591 (7849), 265-269CODEN: NATUAS; ISSN:0028-0836. (Nature Research)Temporal genomic data hold great potential for studying evolutionary processes such as speciation. However, sampling across speciation events would, in many cases, require genomic time series that stretch well back into the Early Pleistocene subepoch. Although theor. models suggest that DNA should survive on this timescale1, the oldest genomic data recovered so far are from a horse specimen dated to 780-560 thousand years ago2. Here we report the recovery of genome-wide data from three mammoth specimens dating to the Early and Middle Pleistocene subepochs, two of which are more than one million years old. We find that two distinct mammoth lineages were present in eastern Siberia during the Early Pleistocene. One of these lineages gave rise to the woolly mammoth and the other represents a previously unrecognized lineage that was ancestral to the first mammoths to colonize North America. Our analyses reveal that the Columbian mammoth of North America traces its ancestry to a Middle Pleistocene hybridization between these two lineages, with roughly equal admixt. proportions. Finally, we show that the majority of protein-coding changes assocd. with cold adaptation in woolly mammoths were already present one million years ago. These findings highlight the potential of deep-time palaeogenomics to expand our understanding of speciation and long-term adaptive evolution.
- 135Li, Z.; Deutscher, M. P. Analyzing the Decay of Stable RNAs in E. Coli. Methods Enzymol. 2008, 447, 31– 45, DOI: 10.1016/S0076-6879(08)02202-7Google ScholarThere is no corresponding record for this reference.
- 136Chan, L. Y.; Mugler, C. F.; Heinrich, S.; Vallotton, P.; Weis, K. Non-Invasive Measurement of mRNA Decay Reveals Translation Initiation as the Major Determinant of mRNA Stability. eLife 2018, DOI: 10.7554/eLife.32536Google ScholarThere is no corresponding record for this reference.
- 137Paul, N.; Joyce, G. F. A Self-Replicating Ligase Ribozyme. Proc. Natl. Acad. Sci. U. S. A. 2002, 99 (20), 12733– 12740, DOI: 10.1073/pnas.202471099Google ScholarThere is no corresponding record for this reference.
- 138Robertson, M. P.; Joyce, G. F. Highly Efficient Self-Replicating RNA Enzymes. Chem. Biol. 2014, 21 (2), 238– 245, DOI: 10.1016/j.chembiol.2013.12.004Google Scholar138https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXisV2isg%253D%253D&md5=5a1af26a9543c01dd17ab35366474b4fHighly Efficient Self-Replicating RNA EnzymesRobertson, Michael P.; Joyce, Gerald F.Chemistry & Biology (Oxford, United Kingdom) (2014), 21 (2), 238-245CODEN: CBOLE2; ISSN:1074-5521. (Elsevier Ltd.)An RNA enzyme has been developed that catalyzes the joining of oligonucleotide substrates to form addnl. copies of itself, undergoing self-replication with exponential growth. The enzyme also can cross-replicate with a partner enzyme, resulting in their mutual exponential growth and enabling self-sustained Darwinian evolution. The opportunity for inventive evolution within this synthetic genetic system depends on the diversity of the evolving population, which is limited by the catalytic efficiency of the enzyme. Directed evolution was used to improve the efficiency of the enzyme and increase its exponential growth rate to 0.14 min-1, corresponding to a doubling time of 5 min. This is close to the limit of 0.21 min-1 imposed by the rate of product release, but sufficient to enable more than 80 logs of growth per day.
- 139Chen, I. A.; Salehi-Ashtiani, K.; Szostak, J. W. RNA Catalysis in Model Protocell Vesicles. J. Am. Chem. Soc. 2005, 127 (38), 13213– 13219, DOI: 10.1021/ja051784pGoogle Scholar139https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXpsVGhtr0%253D&md5=02ae4e206b5b8529dd41d7df46c56793RNA Catalysis in Model Protocell VesiclesChen, Irene A.; Salehi-Ashtiani, Kourosh; Szostak, Jack W.Journal of the American Chemical Society (2005), 127 (38), 13213-13219CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)We are engaged in a long-term effort to synthesize chem. systems capable of Darwinian evolution, based on the encapsulation of self-replicating nucleic acids in self-replicating membrane vesicles. Here, we address the issue of the compatibility of these two replicating systems. Fatty acids form vesicles that are able to grow and divide, but vesicles composed solely of fatty acids are incompatible with the folding and activity of most ribozymes, because low concns. of divalent cations (e.g., Mg2+) cause fatty acids to ppt. Furthermore, vesicles that grow and divide must be permeable to the cations and substrates required for internal metab. We used a mixt. of myristoleic acid and its glycerol monoester to construct vesicles that were Mg2+-tolerant and found that Mg2+ cations can permeate the membrane and equilibrate within a few minutes. In vesicles encapsulating a hammerhead ribozyme, the addn. of external Mg2+ led to the activation and self-cleavage of the ribozyme mols. Vesicles composed of these amphiphiles grew spontaneously through osmotically driven competition between vesicles, and further modification of the membrane compn. allowed growth following mixed micelle addn. Our results show that membranes made from simple amphiphiles can form vesicles that are stable enough to retain encapsulated RNAs in the presence of divalent cations, yet dynamic enough to grow spontaneously and allow the passage of Mg2+ and mononucleotides without specific macromol. transporters. This combination of stability and dynamics is crit. for building model protocells in the lab. and may have been important for early cellular evolution.
- 140Gorbalenya, A. E.; Enjuanes, L.; Ziebuhr, J.; Snijder, E. J. Nidovirales: Evolving the Largest RNA Virus Genome. Virus Res. 2006, 117 (1), 17– 37, DOI: 10.1016/j.virusres.2006.01.017Google ScholarThere is no corresponding record for this reference.
- 141Joyce, G. F.; Szostak, J. W. Protocells and RNA Self-Replication. Cold Spring Harb. Perspect. Biol. 2018, 10 (9), a034801, DOI: 10.1101/cshperspect.a034801Google Scholar141https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhslCltrfL&md5=db161c0090d2a46ee85da2e6e93dba23Protocells and RNA self-replicationJoyce, Gerald F.; Szostak, Jack W.Cold Spring Harbor Perspectives in Biology (2018), 10 (9), a034801/1-a034801/20CODEN: CSHPEU; ISSN:1943-0264. (Cold Spring Harbor Laboratory Press)The general notion of an "RNA world" is that, in the early development of life on the Earth, genetic continuity was assured by the replication of RNA, and RNA mols. were the chief agents of catalytic function. Assuming that all of the components of RNA were available in some prebiotic locale, these components could have assembled into activated nucleotides that condensed to form RNA polymers, setting the stage for the chem. replication of polynucleotides through RNA-templated RNA polymn. If a sufficient diversity of RNAs could be copied with reasonable rate and fidelity, then Darwinian evolution would begin with RNAs that facilitated their own reprodn. enjoying a selective advantage. The concept of a "protocell" refers to a compartment where replication of the primitive genetic material took place and where primitive catalysts gave rise to products that accumulated locally for the benefit of the replicating cellular entity. Replication of both the protocell and its encapsulated genetic material would have enabled natural selection to operate based on the differential fitness of competing cellular entities, ultimately giving rise to modern cellular life.
- 142Jahn, M.; Vorpahl, C.; Hübschmann, T.; Harms, H.; Müller, S. Copy Number Variability of Expression Plasmids Determined by Cell Sorting and Droplet Digital PCR. Microb. Cell Fact. 2016, 15 (1), 211, DOI: 10.1186/s12934-016-0610-8Google ScholarThere is no corresponding record for this reference.
- 143Nielsen, A. A. K.; Der, B. S.; Shin, J.; Vaidyanathan, P.; Paralanov, V.; Strychalski, E. A.; Ross, D.; Densmore, D.; Voigt, C. A. Genetic Circuit Design Automation. Science 2016, 352 (6281), aac7341, DOI: 10.1126/science.aac7341Google ScholarThere is no corresponding record for this reference.
- 144Salis, H. M.; Mirsky, E. A.; Voigt, C. A. Automated Design of Synthetic Ribosome Binding Sites to Control Protein Expression. Nat. Biotechnol. 2009, 27 (10), 946– 950, DOI: 10.1038/nbt.1568Google Scholar144https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXht1WgsbzO&md5=2e61e8668c2011f8d0afa0b1378a3f25Automated design of synthetic ribosome binding sites to control protein expressionSalis, Howard M.; Mirsky, Ethan A.; Voigt, Christopher A.Nature Biotechnology (2009), 27 (10), 946-950CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)Microbial engineering often requires fine control over protein expression-for example, to connect genetic circuits or control flux through a metabolic pathway. To circumvent the need for trial and error optimization, we developed a predictive method for designing synthetic ribosome binding sites, enabling a rational control over the protein expression level. Exptl. validation of >100 predictions in Escherichia coli showed that the method is accurate to within a factor of 2.3 over a range of 100,000-fold. The design method also correctly predicted that reusing identical ribosome binding site sequences in different genetic contexts can result in different protein expression levels. We demonstrate the method's utility by rationally optimizing protein expression to connect a genetic sensor to a synthetic circuit. The proposed forward engineering approach should accelerate the construction and systematic optimization of large genetic systems.
- 145Ostrov, N.; Beal, J.; Ellis, T.; Gordon, D. B.; Karas, B. J.; Lee, H. H.; Lenaghan, S. C.; Schloss, J. A.; Stracquadanio, G.; Trefzer, A.; Bader, J. S.; Church, G. M.; Coelho, C. M.; Efcavitch, J. W.; Güell, M.; Mitchell, L. A.; Nielsen, A. A. K.; Peck, B.; Smith, A. C.; Stewart, C. N., Jr; Tekotte, H. Technological Challenges and Milestones for Writing Genomes. Science 2019, 366 (6463), 310– 312, DOI: 10.1126/science.aay0339Google Scholar145https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhvFyntrnK&md5=24eb50b4c0f38df350da15331a3e6838Technological challenges and milestones for writing genomesOstrov, Nili; Beal, Jacob; Ellis, Tom; Gordon, D. Benjamin; Karas, Bogumil J.; Lee, Henry H.; Lenaghan, Scott C.; Schloss, Jeffery A.; Stracquadanio, Giovanni; Trefzer, Axel; Bader, Joel S.; Church, George M.; Coelho, Cintia M.; Efcavitch, J. William; Guell, Marc; Mitchell, Leslie A.; Nielsen, Alec A. K.; Peck, Bill; Smith, Alexander C.; Stewart, C. Neal, Jr.; Tekotte, HilleScience (Washington, DC, United States) (2019), 366 (6463), 310-312CODEN: SCIEAS; ISSN:1095-9203. (American Association for the Advancement of Science)A review. Engineering biol. with recombinant DNA, broadly called synthetic biol., has progressed tremendously in the last decade, owing to continued industrialization of DNA synthesis, discovery and development of mol. tools and organisms, and increasingly sophisticated modeling and analytic tools. However, we have yet to understand the full potential of engineering biol. because of our inability to write and test whole genomes, which we call synthetic genomics. Substantial improvements are needed to reduce the cost and increase the speed and reliability of genetic tools. Here, we identify emerging technologies and improvements to existing methods that will be needed in four major areas to advance synthetic genomics within the next 10 years: genome design, DNA synthesis, genome editing, and chromosome construction (see table). Similar to other large-scale projects for responsible advancement of innovative technologies, such as the Human Genome Project, an international, cross-disciplinary effort consisting of public and private entities will likely yield maximal return on investment and open new avenues of research and biotechnol.
- 146Farasat, I.; Kushwaha, M.; Collens, J.; Easterbrook, M.; Guido, M.; Salis, H. M. Efficient Search, Mapping, and Optimization of Multi-Protein Genetic Systems in Diverse Bacteria. Mol. Syst. Biol. 2014, 10 (6), 731, DOI: 10.15252/msb.20134955Google Scholar146https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2cfkvFGlug%253D%253D&md5=3531fb8c73dfed9285fea7b9e68b307cEfficient search, mapping, and optimization of multi-protein genetic systems in diverse bacteriaFarasat Iman; Easterbrook Michael; Guido Matthew; Kushwaha Manish; Collens Jason; Salis Howard MMolecular systems biology (2014), 10 (), 731 ISSN:.Developing predictive models of multi-protein genetic systems to understand and optimize their behavior remains a combinatorial challenge, particularly when measurement throughput is limited. We developed a computational approach to build predictive models and identify optimal sequences and expression levels, while circumventing combinatorial explosion. Maximally informative genetic system variants were first designed by the RBS Library Calculator, an algorithm to design sequences for efficiently searching a multi-protein expression space across a > 10,000-fold range with tailored search parameters and well-predicted translation rates. We validated the algorithm's predictions by characterizing 646 genetic system variants, encoded in plasmids and genomes, expressed in six gram-positive and gram-negative bacterial hosts. We then combined the search algorithm with system-level kinetic modeling, requiring the construction and characterization of 73 variants to build a sequence-expression-activity map (SEAMAP) for a biosynthesis pathway. Using model predictions, we designed and characterized 47 additional pathway variants to navigate its activity space, find optimal expression regions with desired activity response curves, and relieve rate-limiting steps in metabolism. Creating sequence-expression-activity maps accelerates the optimization of many protein systems and allows previous measurements to quantitatively inform future designs.
- 147Pretorius, I. S.; Boeke, J. D. Yeast 2.0-Connecting the Dots in the Construction of the World’s First Functional Synthetic Eukaryotic Genome. FEMS Yeast Res. 2018, DOI: 10.1093/femsyr/foy032Google ScholarThere is no corresponding record for this reference.
- 148Blight, K. J.; Kolykhalov, A. A.; Rice, C. M. Efficient Initiation of HCV RNA Replication in Cell Culture. Science 2000, 290 (5498), 1972– 1974, DOI: 10.1126/science.290.5498.1972Google Scholar148https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXoslKgsL4%253D&md5=67eb65d15efc36854faf3940db9a5d91Efficient initiation of HCV RNA replication in cell cultureBlight, Keril J.; Kolykhalov, Alexander A.; Rice, Charles M.Science (Washington, D. C.) (2000), 290 (5498), 1972-1974CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Hepatitis C virus (HCV) infection is a global health problem affecting an estd. 170 million individuals worldwide. We report the identification of multiple independent adaptive mutations that cluster in the HCV nonstructural protein NS5A and confer increased replicative ability in vitro. Among these adaptive mutations were a single amino acid substitution that allowed HCV RNA replication in 10% of transfected hepatoma cells and a deletion of 47 amino acids encompassing the interferon (IFN) sensitivity detg. region (ISDR). Independent of the ISDR, IFN-α rapidly inhibited HCV RNA replication in vitro. This work establishes a robust, cell-based system for genetic and functional analyses of HCV replication.
- 149Gibson, D. G.; Young, L.; Chuang, R.-Y.; Venter, J. C.; Hutchison, C. A., 3rd; Smith, H. O. Enzymatic Assembly of DNA Molecules up to Several Hundred Kilobases. Nat. Methods 2009, 6 (5), 343– 345, DOI: 10.1038/nmeth.1318Google Scholar149https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXksVemsbw%253D&md5=46284924c7d73c47cfb490983338e480Enzymatic assembly of DNA molecules up to several hundred kilobasesGibson, Daniel G.; Young, Lei; Chuang, Ray-Yuan; Venter, J. Craig; Hutchison, Clyde A.; Smith, Hamilton O.Nature Methods (2009), 6 (5), 343-345CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)The authors describe an isothermal, single-reaction method for assembling multiple overlapping DNA mols. by the concerted action of a 5' exonuclease, a DNA polymerase and a DNA ligase. First they recessed DNA fragments, yielding single-stranded DNA overhangs that specifically annealed, and then covalently joined them. This assembly method can be used to seamlessly construct synthetic and natural genes, genetic pathways and entire genomes, and could be a useful mol. engineering tool.
- 150Venter, J. C.; Glass, J. I.; Hutchison, C. A., 3rd; Vashee, S. Synthetic Chromosomes, Genomes, Viruses, and Cells. Cell 2022, 185 (15), 2708– 2724, DOI: 10.1016/j.cell.2022.06.046Google Scholar150https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhvV2lu7%252FP&md5=dc83f01578fe7603c336facd51bd82f3Synthetic chromosomes, genomes, viruses, and cellsVenter, J. Craig; Glass, John I.; Hutchison, Clyde A. III; Vashee, SanjayCell (Cambridge, MA, United States) (2022), 185 (15), 2708-2724CODEN: CELLB5; ISSN:0092-8674. (Cell Press)A review. Synthetic genomics is the construction of viruses, bacteria, and eukaryotic cells with synthetic genomes. It involves two basic processes: synthesis of complete genomes or chromosomes and booting up of those synthetic nucleic acids to make viruses or living cells. The first synthetic genomics efforts resulted in the construction of viruses. This led to a revolution in viral reverse genetics and improvements in vaccine design and manuf. The first bacterium with a synthetic genome led to construction of a minimal bacterial cell and recoded Escherichia coli strains able to incorporate multiple non-std. amino acids in proteins and resistant to phage infection. Further advances led to a yeast strain with a synthetic genome and new approaches for animal and plant artificial chromosomes. On the horizon there are dramatic advances in DNA synthesis that will enable extraordinary new opportunities in medicine, industry, agriculture, and research.
- 151Fredens, J.; Wang, K.; de la Torre, D.; Funke, L. F. H.; Robertson, W. E.; Christova, Y.; Chia, T.; Schmied, W. H.; Dunkelmann, D. L.; Beránek, V.; Uttamapinant, C.; Llamazares, A. G.; Elliott, T. S.; Chin, J. W. Total Synthesis of Escherichia Coli with a Recoded Genome. Nature 2019, 569 (7757), 514– 518, DOI: 10.1038/s41586-019-1192-5Google Scholar151https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXpslGmurg%253D&md5=0c419f23bf66ed52bb301287015c2b0aTotal synthesis of Escherichia coli with a recoded genomeFredens, Julius; Wang, Kaihang; de la Torre, Daniel; Funke, Louise F. H.; Robertson, Wesley E.; Christova, Yonka; Chia, Tiongsun; Schmied, Wolfgang H.; Dunkelmann, Daniel L.; Beranek, Vaclav; Uttamapinant, Chayasith; Llamazares, Andres Gonzalez; Elliott, Thomas S.; Chin, Jason W.Nature (London, United Kingdom) (2019), 569 (7757), 514-518CODEN: NATUAS; ISSN:0028-0836. (Nature Research)Nature uses 64 codons to encode the synthesis of proteins from the genome, and chooses 1 sense codon-out of up to 6 synonyms-to encode each amino acid. Synonymous codon choice has diverse and important roles, and many synonymous substitutions are detrimental. Here we demonstrate that the no. of codons used to encode the canonical amino acids can be reduced, through the genome-wide substitution of target codons by defined synonyms. We create a variant of Escherichia coli with a four-megabase synthetic genome through a high-fidelity convergent total synthesis. Our synthetic genome implements a defined recoding and refactoring scheme-with simple corrections at just seven positions-to replace every known occurrence of two sense codons and a stop codon in the genome. Thus, we recode 18,214 codons to create an organism with a 61-codon genome; this organism uses 59 codons to encode the 20 amino acids, and enables the deletion of a previously essential tRNA.
- 152Zhao, Y.; Coelho, C.; Hughes, A. L.; Lazar-Stefanita, L.; Yang, S.; Brooks, A. N.; Walker, R. S. K.; Zhang, W.; Lauer, S.; Hernandez, C.; Cai, J.; Mitchell, L. A.; Agmon, N.; Shen, Y.; Sall, J.; Fanfani, V.; Jalan, A.; Rivera, J.; Liang, F.-X.; Bader, J. S.; Stracquadanio, G.; Steinmetz, L. M.; Cai, Y.; Boeke, J. D. Debugging and Consolidating Multiple Synthetic Chromosomes Reveals Combinatorial Genetic Interactions. Cell 2023, 186 (24), 5220– 5236, DOI: 10.1016/j.cell.2023.09.025Google ScholarThere is no corresponding record for this reference.
- 153Woese, C. The Universal Ancestor. Proc. Natl. Acad. Sci. U. S. A. 1998, 95 (12), 6854– 6859, DOI: 10.1073/pnas.95.12.6854Google ScholarThere is no corresponding record for this reference.
- 154Woese, C. R. On the Evolution of Cells. Proc. Natl. Acad. Sci. U. S. A. 2002, 99 (13), 8742– 8747, DOI: 10.1073/pnas.132266999Google ScholarThere is no corresponding record for this reference.
- 155Sandberg, T. E.; Wise, K. S.; Dalldorf, C.; Szubin, R.; Feist, A. M.; Glass, J. I.; Palsson, B. O. Adaptive Evolution of a Minimal Organism with a Synthetic Genome. iScience 2023, 26 (9), 107500, DOI: 10.1016/j.isci.2023.107500Google ScholarThere is no corresponding record for this reference.
- 156Moger-Reischer, R. Z.; Glass, J. I.; Wise, K. S.; Sun, L.; Bittencourt, D. M. C.; Lehmkuhl, B. K.; Schoolmaster, D. R., Jr; Lynch, M.; Lennon, J. T. Evolution of a Minimal Cell. Nature 2023, 620 (7972), 122– 127, DOI: 10.1038/s41586-023-06288-xGoogle ScholarThere is no corresponding record for this reference.
- 157Strotz, L. C.; Simões, M.; Girard, M. G.; Breitkreuz, L.; Kimmig, J.; Lieberman, B. S. Getting Somewhere with the Red Queen: Chasing a Biologically Modern Definition of the Hypothesis. Biol. Lett. 2018, 14 (5), 20170734, DOI: 10.1098/rsbl.2017.0734Google ScholarThere is no corresponding record for this reference.
- 158Solé, R. Revisiting Leigh Van Valen’s “A New Evolutionary Law” (1973). Biol. Theory 2022, 17 (2), 120– 125, DOI: 10.1007/s13752-021-00391-wGoogle ScholarThere is no corresponding record for this reference.
- 159Hammerling, M. J.; Krüger, A.; Jewett, M. C. Strategies for in Vitro Engineering of the Translation Machinery. Nucleic Acids Res. 2020, 48 (3), 1068– 1083, DOI: 10.1093/nar/gkz1011Google Scholar159https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhs1Ogur3M&md5=90749c3fcd81c379acb645f02d89a421Strategies for in vitro engineering of the translation machineryHammerling, Michael J.; Krueger, Antje; Jewett, Michael C.Nucleic Acids Research (2020), 48 (3), 1068-1083CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)A review. Engineering the process of mol. translation, or protein biosynthesis, has emerged as a major opportunity in synthetic and chem. biol. to generate novel biol. insights and enable new applications (e.g. designer protein therapeutics). Here, we review methods for engineering the process of translation in vitro. We discuss the advantages and drawbacks of the two major strategies-purified and ext.-based systems-and how they may be used to manipulate and study translation. Techniques to engineer each component of the translation machinery are covered in turn, including tRNAs, translation factors, and the ribosome. Finally, future directions and enabling technol. advances for the field are discussed.
- 160Williams, T. C.; Averesch, N. J. H.; Winter, G.; Plan, M. R.; Vickers, C. E.; Nielsen, L. K.; Krömer, J. O. Quorum-Sensing Linked RNA Interference for Dynamic Metabolic Pathway Control in Saccharomyces Cerevisiae. Metab. Eng. 2015, 29, 124– 134, DOI: 10.1016/j.ymben.2015.03.008Google Scholar160https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXkvF2ru7w%253D&md5=c58b47da7c9140d71340afecce19f5e7Quorum-sensing linked RNA interference for dynamic metabolic pathway control in Saccharomyces cerevisiaeWilliams, T. C.; Averesch, N. J. H.; Winter, G.; Plan, M. R.; Vickers, C. E.; Nielsen, L. K.; Kromer, J. O.Metabolic Engineering (2015), 29 (), 124-134CODEN: MEENFM; ISSN:1096-7176. (Elsevier B. V.)Some of the most productive metabolic engineering strategies involve genetic modifications that cause severe metabolic burden on the host cell. Growth-limiting genetic modifications can be more effective if they are 'switched on' after a population growth phase has been completed. To address this problem we have engineered dynamic regulation using a previously developed synthetic quorum sensing circuit in Saccharomyces cerevisiae. The circuit autonomously triggers gene expression at a high population d., and was linked with an RNA interference module to enable target gene silencing. As a demonstration the circuit was used to control flux through the shikimate pathway for the prodn. of para-hydroxybenzoic acid (PHBA). Dynamic RNA repression allowed gene knock-downs which were identified by elementary flux mode anal. as highly productive but with low biomass formation to be implemented after a population growth phase, resulting in the highest published PHBA titer in yeast (1.1 mM).
- 161Robinson, A. O.; Venero, O. M.; Adamala, K. P. Toward Synthetic Life: Biomimetic Synthetic Cell Communication. Curr. Opin. Chem. Biol. 2021, 64, 165– 173, DOI: 10.1016/j.cbpa.2021.08.008Google Scholar161https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXitFejsr%252FL&md5=850478c427d6eabda0a43b5fcfdb4423Toward synthetic life: Biomimetic synthetic cell communicationRobinson, Abbey O.; Venero, Orion M.; Adamala, Katarzyna P.Current Opinion in Chemical Biology (2021), 64 (), 165-173CODEN: COCBF4; ISSN:1367-5931. (Elsevier B.V.)A review. Engineering synthetic minimal cells provide a controllable chassis for studying the biochem. principles of natural life, increasing our understanding of complex biol. processes. Recently, synthetic cell engineering has enabled communication between both natural live cells and other synthetic cells. A system such as these enable studying interactions between populations of cells, both natural and artificial, and engineering small mol. cell communication protocols for a variety of basic research and practical applications. In this review, we summarize recent progress in engineering communication between synthetic and natural cells, and we speculate about the possible future directions of this work.
- 162Garamella, J.; Majumder, S.; Liu, A. P.; Noireaux, V. An Adaptive Synthetic Cell Based on Mechanosensing, Biosensing, and Inducible Gene Circuits. ACS Synth. Biol. 2019, 8 (8), 1913– 1920, DOI: 10.1021/acssynbio.9b00204Google Scholar162https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtl2jtLnP&md5=d0c92306403ebbacb296c43bf9f5e9d0An Adaptive Synthetic Cell Based on Mechanosensing, Biosensing, and Inducible Gene CircuitsGaramella, Jonathan; Majumder, Sagardip; Liu, Allen P.; Noireaux, VincentACS Synthetic Biology (2019), 8 (8), 1913-1920CODEN: ASBCD6; ISSN:2161-5063. (American Chemical Society)The bottom-up assembly of synthetic cell systems capable of recapitulating biol. functions has become a means to understand living matter by construction. The integration of biomol. components into active, cell-sized, genetically programmed compartments remains, however, a major bottleneck for building synthetic cells. A primary feature of real cells is their ability to actively interact with their surroundings, particularly in stressed conditions. Here, we construct a synthetic cell equipped with an inducible genetic circuit that responds to changes in osmotic pressure through the mechanosensitive channel MscL. Liposomes loaded with an E. coli cell-free transcription-translation (TXTL) system are induced with IPTG when exposed to hypo-osmotic soln., resulting in the expression of a bacterial cytoskeletal protein MreB. MreB assocs. with the membrane to generate a cortex-like structure. Our work provides the first example of mol. integration that couples mechanosensitivity, gene expression, and self-assembly at the inner membrane of synthetic cells.
- 163Liu, H.; Yang, Q.; Peng, R.; Kuai, H.; Lyu, Y.; Pan, X.; Liu, Q.; Tan, W. Artificial Signal Feedback Network Mimicking Cellular Adaptivity. J. Am. Chem. Soc. 2019, 141 (16), 6458– 6461, DOI: 10.1021/jacs.8b13816Google Scholar163https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXms1Wqsr4%253D&md5=4f308379b93eae5483c35a93279cf605Artificial Signal Feedback Network Mimicking Cellular AdaptivityLiu, Hui; Yang, Qiuxia; Peng, Ruizi; Kuai, Hailan; Lyu, Yifan; Pan, Xiaoshu; Liu, Qiaoling; Tan, WeihongJournal of the American Chemical Society (2019), 141 (16), 6458-6461CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Inspired by this elegant system of cellular adaptivity, we herein report the rational design of a dynamic artificial adaptive system able to sense and respond to environmental stresses in a unique sense-and-respond mode. Utilizing DNA nanotechnol., we constructed an artificial signal feedback network and anchored it to surface membrane of a model giant membrane vesicle (GMV) protocell. Such system would need to both sense incoming stimuli and emit a feedback response to eliminate the stimuli. To accomplish this mechanistically, our DNA-based artificial signal system, hereinafter termed DASsys, was equipped with a DNA trig-ger-induced DNA polymer formation and dissocn. machinery. Thus, through a sequential cascade of stimulus-induced DNA strand displacement, DASsys could effectively sense and respond to incoming stimuli. Then, by eliminating the stimulus, the membrane surface would return to its initial state, realizing the formation of a cyclical feedback mechanism. Overall, our strategy opens up a route to the construction of artificial signaling system capable of maintaining homeostasis in the cellular micromilieu, and addresses important emerging challenges in bioinspired engineering.
- 164Gispert, I.; Hindley, J. W.; Pilkington, C. P.; Shree, H.; Barter, L. M. C.; Ces, O.; Elani, Y. Stimuli-Responsive Vesicles as Distributed Artificial Organelles for Bacterial Activation. Proc. Natl. Acad. Sci. U. S. A. 2022, 119 (42), e2206563119, DOI: 10.1073/pnas.2206563119Google ScholarThere is no corresponding record for this reference.
- 165Aufinger, L.; Simmel, F. C. Establishing Communication Between Artificial Cells. Chemistry 2019, 25 (55), 12659– 12670, DOI: 10.1002/chem.201901726Google Scholar165https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhsFSrt7vJ&md5=c7040b1a0bbee738b5c20f4a23e8d205Establishing Communication Between Artificial CellsAufinger, Lukas; Simmel, Friedrich C.Chemistry - A European Journal (2019), 25 (55), 12659-12670CODEN: CEUJED; ISSN:0947-6539. (Wiley-VCH Verlag GmbH & Co. KGaA)A review. Communication between artificial cells is essential for the realization of complex dynamical behaviors at the multi-cell level. It is also an important prerequisite for modular systems design, because it dets. how spatially sepd. functional modules can coordinate their actions. Among others, mol. communication is required for artificial cell signaling, synchronization of cellular behaviors, computation, group-level decision-making processes and pattern formation in artificial tissues. In this review, an overview of various recent approaches to create communicating artificial cellular systems is provided. In this context, important physicochem. boundary conditions that have to be considered for the design of the communicating cells are also described, and a survey of the most striking emergent behaviors that may be achieved in such systems is given.
- 166Pereyre, S.; Sirand-Pugnet, P.; Beven, L.; Charron, A.; Renaudin, H.; Barré, A.; Avenaud, P.; Jacob, D.; Couloux, A.; Barbe, V.; de Daruvar, A.; Blanchard, A.; Bébéar, C. Life on Arginine for Mycoplasma Hominis: Clues from Its Minimal Genome and Comparison with Other Human Urogenital Mycoplasmas. PLoS Genet. 2009, 5 (10), e1000677, DOI: 10.1371/journal.pgen.1000677Google ScholarThere is no corresponding record for this reference.
- 167Woese, C. R.; Maniloff, J.; Zablen, L. B. Phylogenetic Analysis of the Mycoplasmas. Proc. Natl. Acad. Sci. U. S. A. 1980, 77 (1), 494– 498, DOI: 10.1073/pnas.77.1.494Google ScholarThere is no corresponding record for this reference.
- 168Li, S.; Guan, J.-L.; Chien, S. Biochemistry and Biomechanics of Cell Motility. Annu. Rev. Biomed. Eng. 2005, 7, 105– 150, DOI: 10.1146/annurev.bioeng.7.060804.100340Google Scholar168https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXps12ltL8%253D&md5=c2f77f8a164b1ab53ed970c85e6d7ffbBiochemistry and biomechanics of cell motilityLi, Song; Guan, Jun-Lin; Chien, ShuAnnual Review of Biomedical Engineering (2005), 7 (), 105-150, 2 platesCODEN: ARBEF7; ISSN:1523-9829. (Annual Reviews Inc.)A review. Cell motility is an essential cellular process for a variety of biol. events. The process of cell migration requires the integration and coordination of complex biochem. and biomech. signals. The protrusion force at the leading edge of a cell is generated by the cytoskeleton, and this force generation is controlled by multiple signaling cascades. The formation of new adhesions at the front and the release of adhesions at the rear involve the outside-in and inside-out signaling mediated by integrins and other adhesion receptors. The traction force generated by the cell on the extracellular matrix (ECM) regulates cell-ECM adhesions, and the counter force exerted by ECM on the cell drives the migration. The polarity of cell migration can be amplified and maintained by the feedback loop between the cytoskeleton and cell-ECM adhesions. Cell migration in 3-dimensional ECM has characteristics distinct from that on 2-dimensional ECM. The migration of cells is initiated and modulated by external chem. and mech. factors, such as chemoattractants and the mech. forces acting on the cells and ECM, as well as the surface d., distribution, topog., and rigidity of the ECM.
- 169Brunet, T.; Albert, M.; Roman, W.; Coyle, M. C.; Spitzer, D. C.; King, N. A Flagellate-to-Amoeboid Switch in the Closest Living Relatives of Animals. eLife 2021, DOI: 10.7554/eLife.61037Google ScholarThere is no corresponding record for this reference.
- 170Siton-Mendelson, O.; Bernheim-Groswasser, A. Toward the Reconstitution of Synthetic Cell Motility. Cell Adh. Migr. 2016, 10 (5), 461– 474, DOI: 10.1080/19336918.2016.1170260Google Scholar170https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC28fkt1KitA%253D%253D&md5=614c8b2da2257939347704e78670e1f4Toward the reconstitution of synthetic cell motilitySiton-Mendelson Orit; Bernheim-Groswasser AnneCell adhesion & migration (2016), 10 (5), 461-474 ISSN:.Cellular motility is a fundamental process essential for embryonic development, wound healing, immune responses, and tissues development. Cells are mostly moving by crawling on external, or inside, substrates which can differ in their surface composition, geometry, and dimensionality. Cells can adopt different migration phenotypes, e.g., bleb-based and protrusion-based, depending on myosin contractility, surface adhesion, and cell confinement. In the few past decades, research on cell motility has focused on uncovering the major molecular players and their order of events. Despite major progresses, our ability to infer on the collective behavior from the molecular properties remains a major challenge, especially because cell migration integrates numerous chemical and mechanical processes that are coupled via feedbacks that span over large range of time and length scales. For this reason, reconstituted model systems were developed. These systems allow for full control of the molecular constituents and various system parameters, thereby providing insight into their individual roles and functions. In this review we describe the various reconstituted model systems that were developed in the past decades. Because of the multiple steps involved in cell motility and the complexity of the overall process, most of the model systems focus on very specific aspects of the individual steps of cell motility. Here we describe the main advancement in cell motility reconstitution and discuss the main challenges toward the realization of a synthetic motile cell.
- 171Blanken, D.; van Nies, P.; Danelon, C. Quantitative Imaging of Gene-Expressing Liposomes Reveals Rare Favorable Phenotypes. Phys. Biol. 2019, 16 (4), 045002, DOI: 10.1088/1478-3975/ab0c62Google Scholar171https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXitV2gs7Y%253D&md5=5cfe9787e67c5772d03f800f2a440dfcQuantitative imaging of gene-expressing liposomes reveals rare favorable phenotypesBlanken, Duco; van Nies, Pauline; Danelon, ChristophePhysical Biology (2019), 16 (4), 045002CODEN: PBHIAT; ISSN:1478-3975. (IOP Publishing Ltd.)We report on direct imaging of tens of thousands of gene-expressing liposomes per sample allowing us to assess sub-population features in a statistically relevant manner. Both the vesicle size(diam. <10μm) and lipid compn. (mixt. of phospholipids with zwitterionic and neg. charged headgroups, including cardiolipin) are compatible with the properties of bacterial cells. Therefore, our liposomes provide suitable chassis to host Escherichia coli-derived PURE translation machinery and other bacterial processes in future developments. The potential of high-content imaging to identify rare phenotypes is demonstrated by fact that a subset of the liposome population exhibits remarkably high yield of synthesized protein or prolonged expression lifespan that surpasses performance of ensemble liposome-averaged and bulk reactions. Among the three com. PURE systems tested, PUREfrex2.0 offers the most favorable phenotypes displaying both high yield and long protein synthesis lifespan. Moreover, probing membrane permeability reveals a large heterogeneity amongst liposomes. In situ expression and membrane embedding of the pore-forming connexin leads to a characteristic permeability time profile, while increasing the fraction of permeable liposomes in the population. We see diversity in gene expression dynamics and membrane permeability as an opportunity to complement a rational design approach aiming at further implementing biol. functions in liposome-based synthetic cells.
- 172Stano, P.; D’Aguanno, E.; Bolz, J.; Fahr, A.; Luisi, P. L. A Remarkable Self-Organization Process as the Origin of Primitive Functional Cells. Angew. Chem., Int. Ed. Engl. 2013, 52 (50), 13397– 13400, DOI: 10.1002/anie.201306613Google ScholarThere is no corresponding record for this reference.
- 173Weitz, M.; Kim, J.; Kapsner, K.; Winfree, E.; Franco, E.; Simmel, F. C. Diversity in the Dynamical Behaviour of a Compartmentalized Programmable Biochemical Oscillator. Nat. Chem. 2014, 6 (4), 295– 302, DOI: 10.1038/nchem.1869Google Scholar173https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXisFOjtbs%253D&md5=bc290cb6326fadb156dcb85af3eb587fDiversity in the dynamical behaviour of a compartmentalized programmable biochemical oscillatorWeitz, Maximilian; Kim, Jongmin; Kapsner, Korbinian; Winfree, Erik; Franco, Elisa; Simmel, Friedrich C.Nature Chemistry (2014), 6 (4), 295-302CODEN: NCAHBB; ISSN:1755-4330. (Nature Publishing Group)In vitro compartmentalization of biochem. reaction networks is a crucial step towards engineering artificial cell-scale devices and systems. At this scale the dynamics of mol. systems becomes stochastic, which introduces several engineering challenges and opportunities. Here we study a programmable transcriptional oscillator system that is compartmentalized into microemulsion droplets with vols. between 33 fl and 16 pl. Simultaneous measurement of large populations of droplets reveals major variations in the amplitude, frequency and damping of the oscillations. Variability increases for smaller droplets and depends on the operating point of the oscillator. Rather than reflecting the stochastic kinetics of the chem. reaction network itself, the variability can be attributed to the statistical variation of reactant concns. created during their partitioning into droplets. We anticipate that robustness to partitioning variability will be a crit. challenge for engineering cell-scale systems, and that highly parallel time-series acquisition from microemulsion droplets will become a key tool for characterization of stochastic circuit function.
- 174Hansen, M. M. K.; Meijer, L. H. H.; Spruijt, E.; Maas, R. J. M.; Rosquelles, M. V.; Groen, J.; Heus, H. A.; Huck, W. T. S. Macromolecular Crowding Creates Heterogeneous Environments of Gene Expression in Picolitre Droplets. Nat. Nanotechnol. 2016, 11 (2), 191– 197, DOI: 10.1038/nnano.2015.243Google Scholar174https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhslantL%252FL&md5=d2f5c923076100fe2836c79f5bf0bc1bMacromolecular crowding creates heterogeneous environments of gene expression in picolitre dropletsHansen, Maike M. K.; Meijer, Lenny H. H.; Spruijt, Evan; Maas, Roel J. M.; Rosquelles, Marta Ventosa; Groen, Joost; Heus, Hans A.; Huck, Wilhelm T. S.Nature Nanotechnology (2016), 11 (2), 191-197CODEN: NNAABX; ISSN:1748-3387. (Nature Publishing Group)Understanding the dynamics of complex enzymic reactions in highly crowded small vols. is crucial for the development of synthetic minimal cells. Compartmentalized biochem. reactions in cell-sized containers exhibit a degree of randomness due to the small no. of mols. involved. However, it is unknown how the phys. environment contributes to the stochastic nature of multistep enzymic processes. Here, we present a robust method to quantify gene expression noise in vitro using droplet microfluidics. We study the changes in stochasticity in the cell-free gene expression of two genes compartmentalized within droplets as a function of DNA copy no. and macromol. crowding. We find that decreased diffusion caused by a crowded environment leads to the spontaneous formation of heterogeneous microenvironments of mRNA as local prodn. rates exceed the diffusion rates of macromols. This heterogeneity leads to a higher probability of the mol. machinery staying in the same microenvironment, directly increasing the system's stochasticity.
- 175Rao, C. V. Expanding the Synthetic Biology Toolbox: Engineering Orthogonal Regulators of Gene Expression. Curr. Opin. Biotechnol. 2012, 23 (5), 689– 694, DOI: 10.1016/j.copbio.2011.12.015Google ScholarThere is no corresponding record for this reference.
- 176Yelleswarapu, M.; van der Linden, A. J.; van Sluijs, B.; Pieters, P. A.; Dubuc, E.; de Greef, T. F. A.; Huck, W. T. S. Sigma Factor-Mediated Tuning of Bacterial Cell-Free Synthetic Genetic Oscillators. ACS Synth. Biol. 2018, 7 (12), 2879– 2887, DOI: 10.1021/acssynbio.8b00300Google Scholar176https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitFaqsbfK&md5=95b792df5ec10e6ce8bbdca9264977abSigma factor-mediated tuning of bacterial cell-free synthetic genetic oscillatorsYelleswarapu, Maaruthy; van der Linden, Ardjan J.; van Sluijs, Bob; Pieters, Pascal A.; Dubuc, Emilien; de Greef, Tom F. A.; Huck, Wilhelm T. S.ACS Synthetic Biology (2018), 7 (12), 2879-2887CODEN: ASBCD6; ISSN:2161-5063. (American Chemical Society)Cell-free transcription-translation provides a simplified prototyping environment to rapidly design and study synthetic networks. Despite the presence of a well-characterized toolbox of genetic elements, examples of genetic networks that exhibit complex temporal behavior are scarce. Here, we present a genetic oscillator implemented in an Escherichia coli-based cell-free system under steady-state conditions using microfluidic flow reactors. The oscillator had an activator-repressor motif that utilizes the native transcriptional machinery of E. coli: the RNA polymerase (RNAP) and its assocd. σ-factors. We optimized a kinetic model with exptl. data using an evolutionary algorithm to quantify the key regulatory model parameters. The functional modulation of RNAP was investigated by coupling 2 oscillators driven by competing σ-factors, allowing the modification of network properties by means of passive transcriptional regulation.
- 177Li, J.; Zhang, C.; Huang, P.; Kuru, E.; Forster-Benson, E. T. C.; Li, T.; Church, G. M. Dissecting Limiting Factors of the Protein Synthesis Using Recombinant Elements (PURE) System. Translation (Austin) 2017, 5 (1), e1327006, DOI: 10.1080/21690731.2017.1327006Google ScholarThere is no corresponding record for this reference.
- 178Lavickova, B.; Maerkl, S. J. A Simple, Robust, and Low-Cost Method To Produce the PURE Cell-Free System. ACS Synth. Biol. 2019, 8 (2), 455– 462, DOI: 10.1021/acssynbio.8b00427Google Scholar178https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXnvF2gtA%253D%253D&md5=510c38e95769a3ea6ba79ea2238c7d87A Simple, Robust, and Low-Cost Method To Produce the PURE Cell-Free SystemLavickova, Barbora; Maerkl, Sebastian J.ACS Synthetic Biology (2019), 8 (2), 455-462CODEN: ASBCD6; ISSN:2161-5063. (American Chemical Society)We demonstrate a simple, robust, and low-cost method for producing the PURE cell-free transcription-translation system. Our OnePot PURE system achieved a protein synthesis yield of 156 μg/mL at a cost of 0.09 USD/μL, leading to a 14-fold improvement in cost normalized protein synthesis yield over existing PURE systems. The one-pot method makes the PURE system easy to generate and allows it to be readily optimized and modified.
- 179Li, J.; Gu, L.; Aach, J.; Church, G. M. Improved Cell-Free RNA and Protein Synthesis System. PLoS One 2014, 9 (9), e106232, DOI: 10.1371/journal.pone.0106232Google Scholar179https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhs12isr7F&md5=4eb9de2e59d20518523324e7ee54a406Improved cell-free RNA and protein synthesis systemLi, Jun; Gu, Liangcai; Aach, John; Church, George M.PLoS One (2014), 9 (9), e106232/1-e106232/11, 11 pp.CODEN: POLNCL; ISSN:1932-6203. (Public Library of Science)Cell-free RNA and protein synthesis (CFPS) is becoming increasingly used for protein prodn. as yields increase and costs decrease. Advances in reconstituted CFPS systems such as the Protein synthesis Using Recombinant Elements (PURE) system offer new opportunities to tailor the reactions for specialized applications including in vitro protein evolution, protein microarrays, isotopic labeling, and incorporating unnatural amino acids. In this study, using firefly luciferase synthesis as a reporter system, we improved PURE system productivity up to 5 fold by adding or adjusting a variety of factors that affect transcription and translation, including Elongation factors (EF-Ts, EF-Tu, EF-G and EF4), ribosome recycling factor (RRF), release factors (RF1, RF2, RF3), chaperones (GroEL/ES), BSA and tRNAs. The work provides a more efficient defined in vitro transcription and translation system and a deeper understanding of the factors that limit the whole system efficiency.
- 180Jewett, M. C.; Calhoun, K. A.; Voloshin, A.; Wuu, J. J.; Swartz, J. R. An Integrated Cell-Free Metabolic Platform for Protein Production and Synthetic Biology. Mol. Syst. Biol. 2008, 4, 220, DOI: 10.1038/msb.2008.57Google Scholar180https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD1cnmtVWluw%253D%253D&md5=59a196db8dc23aaf194e876ce7fdd36bAn integrated cell-free metabolic platform for protein production and synthetic biologyJewett Michael C; Calhoun Kara A; Voloshin Alexei; Wuu Jessica J; Swartz James RMolecular systems biology (2008), 4 (), 220 ISSN:.Cell-free systems offer a unique platform for expanding the capabilities of natural biological systems for useful purposes, i.e. synthetic biology. They reduce complexity, remove structural barriers, and do not require the maintenance of cell viability. Cell-free systems, however, have been limited by their inability to co-activate multiple biochemical networks in a single integrated platform. Here, we report the assessment of biochemical reactions in an Escherichia coli cell-free platform designed to activate natural metabolism, the Cytomim system. We reveal that central catabolism, oxidative phosphorylation, and protein synthesis can be co-activated in a single reaction system. Never before have these complex systems been shown to be simultaneously activated without living cells. The Cytomim system therefore promises to provide the metabolic foundation for diverse ab initio cell-free synthetic biology projects. In addition, we describe an improved Cytomim system with enhanced protein synthesis yields (up to 1200 mg/l in 2 h) and lower costs to facilitate production of protein therapeutics and biochemicals that are difficult to make in vivo because of their toxicity, complexity, or unusual cofactor requirements.
- 181Silverman, A. D.; Karim, A. S.; Jewett, M. C. Cell-Free Gene Expression: An Expanded Repertoire of Applications. Nat. Rev. Genet. 2020, 21 (3), 151– 170, DOI: 10.1038/s41576-019-0186-3Google Scholar181https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXit1OitbbO&md5=e227ddcf47b0b961b2b423d414fa3795Cell-free gene expression: an expanded repertoire of applicationsSilverman, Adam D.; Karim, Ashty S.; Jewett, Michael C.Nature Reviews Genetics (2020), 21 (3), 151-170CODEN: NRGAAM; ISSN:1471-0056. (Nature Research)Cell-free biol. is the activation of biol. processes without the use of intact living cells. It has been used for more than 50 years across the life sciences as a foundational research tool, but a recent tech. renaissance has facilitated high-yielding (grams of protein per L), cell-free gene expression systems from model bacteria, the development of cell-free platforms from non-model organisms and multiplexed strategies for rapidly assessing biol. design. These advances provide exciting opportunities to profoundly transform synthetic biol. by enabling new approaches to the model-driven design of synthetic gene networks, the fast and portable sensing of compds., on-demand biomanufg., building cells from the bottom up, and next-generation educational kits.
- 182Ouyang, X.; Zhou, X.; Lai, S. N.; Liu, Q.; Zheng, B. Immobilization of Proteins of Cell Extract to Hydrogel Networks Enhances the Longevity of Cell-Free Protein Synthesis and Supports Gene Networks. ACS Synth. Biol. 2021, 10 (4), 749– 755, DOI: 10.1021/acssynbio.0c00541Google ScholarThere is no corresponding record for this reference.
- 183New England Biolabs. NEBExpress Cell-free ecoli Protein Synthesis System. https://www.neb.com/products/e5360-nebexpress-cell-free-ecoli-protein-synthesis-system (accessed on August 21, 2023).Google ScholarThere is no corresponding record for this reference.
- 184Zimmerman, S. B.; Trach, S. O. Estimation of Macromolecule Concentrations and Excluded Volume Effects for the Cytoplasm of Escherichia Coli. J. Mol. Biol. 1991, 222 (3), 599– 620, DOI: 10.1016/0022-2836(91)90499-VGoogle Scholar184https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK38XlsFanuw%253D%253D&md5=df94a9250fe979cc422c967c1a7332caEstimation of macromolecule concentrations and excluded volume effects for the cytoplasm of Escherichia coliZimmerman, Steven B.; Trach, Stefan O.Journal of Molecular Biology (1991), 222 (3), 599-620CODEN: JMOBAK; ISSN:0022-2836.The very high concn. of macromols. within cells can potentially have an overwhelming effect on the thermodn. activity of cellular components because of excluded vol. effects. To est. the magnitudes of such effects, an exptl. study was made of the cytoplasm of Escherichia coli. Parameters from cells and cell exts. are used to calc. approx. activity coeffs. for cytoplasmic conditions. These calcns. require a representation of the sizes, concns. and effective sp. vol. of the macromols. in the exts. Macromol. size representations are obtained either by applying a two-phase distribution assay to define a related homogeneous soln. or by using the mol. mass distribution of macromols. from gel filtration. Macromol. concns. in cytoplasm are obtained from analyses of exts. by applying a correction for the diln. that occurs during extn. That factor is detd. from expts. based upon the known impermeability of the cytoplasmic vol. to sucrose in intact E. coli. Macromol. concns. in the cytoplasm of E. coli in either exponential or stationary growth phase are estd. to be ≈0.3 to 0.4 g/mL. Macromol. sp. vol. are inferred from the compn. of close-packed ppts. induced by polyethylene glycol. Several well-characterized proteins which bind to DNA (lac repressor, RNA polymerase) are extremely sensitive to changes in salt concn. in studies in vitro, but are insensitive in studies in vivo. Application of the activity coeffs. from the present work indicates that at least part of this discrepancy arises from the differences in excluded vols. in these studies. Applications of the activity coeffs. to soly. or to assocn. reactions are also discussed, as are changes assocd. with cell growth phase and osmotic or other effects. The use of solns. of purified macromols. that emulate the crowding conditions inferred for cytoplasm is discussed.
- 185Record, M. T., Jr; Courtenay, E. S.; Cayley, D. S.; Guttman, H. J. Responses of E. Coli to Osmotic Stress: Large Changes in Amounts of Cytoplasmic Solutes and Water. Trends Biochem. Sci. 1998, 23 (4), 143– 148, DOI: 10.1016/S0968-0004(98)01196-7Google Scholar185https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXivFKmsLc%253D&md5=51c3002e687a30f01fccddaf77ca9ebbResponses of E. coli to osmotic stress: large changes in amounts of cytoplasmic solutes and waterRecord, M. Thomas, Jr.; Courtenay, Elizabeth S.; Cayley, D. Scott; Guttman, Harry J.Trends in Biochemical Sciences (1998), 23 (4), 143-148CODEN: TBSCDB; ISSN:0968-0004. (Elsevier Science Ltd.)A review with 37 refs. Escherichia coli is capable of growing in environments ranging from very dil. aq. solns. of essential nutrients to media contg. molar concns. of salts or nonelectrolyte solutes. Growth in environments with such a wide range (at least 100-fold) of osmolarities poses significant physiol. challenges for cells. To meet these challenges, E. coli adjusts a wide range of cytoplasmic soln. variables, including the cytoplasmic amts. both of water and of charged and uncharged solutes.
- 186Cayley, S.; Lewis, B. A.; Guttman, H. J.; Record, M. T. Jr. Characterization of the Cytoplasm of Escherichia Coli K-12 as a Function of External Osmolarity. Implications for Protein-DNA Interactions in Vivo. J. Mol. Biol. 1991, 222 (2), 281– 300, DOI: 10.1016/0022-2836(91)90212-OGoogle ScholarThere is no corresponding record for this reference.
- 187Li, J.; Haas, W.; Jackson, K.; Kuru, E.; Jewett, M. C.; Fan, Z. H.; Gygi, S.; Church, G. M. Cogenerating Synthetic Parts toward a Self-Replicating System. ACS Synth. Biol. 2017, 6 (7), 1327– 1336, DOI: 10.1021/acssynbio.6b00342Google Scholar187https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXks12htrg%253D&md5=6ecae4340f4b4616f4b3bd1f41d5a359Co-generating Synthetic Parts toward a Self-Replicating SystemLi, Jun; Haas, Wilhelm; Jackson, Kirsten; Kuru, Erkin; Jewett, Michael C.; Fan, Z. Hugh; Gygi, Steven; Church, George M.ACS Synthetic Biology (2017), 6 (7), 1327-1336CODEN: ASBCD6; ISSN:2161-5063. (American Chemical Society)To build replicating systems with new functions, the engineering of existing biol. machineries requires a sensible strategy. Protein synthesis Using Recombinant Elements (PURE) system consists of the desired components for transcription, translation, aminoacylation and energy regeneration. PURE might be the basis for a radically alterable, lifelike system after optimization. Here, the authors regenerated 54 E. coli ribosomal (r-) proteins individually from DNA templates in the PURE system. The authors show that using stable isotope labeling with amino acids, mass spectrometry based quant. proteomics could detect 26 of the 33 50S and 20 of the 21 30S subunit r-proteins when coexpressed in batch format PURE system. By optimizing DNA template concns. and adapting a miniaturized Fluid Array Device with optimized feeding soln., the authors were able to cogenerate and detect at least 29 of the 33 50S and all of the 21 30S subunit r-proteins in one pot. The boost on yield of a single r-protein in coexpression pool varied from ∼1.5 to 5-fold compared to the batch mode, with up to ∼2.4 μM yield for a single r-protein. Reconstituted ribosomes under physiol. condition from PURE system synthesized 30S r-proteins and native 16S rRNA showed ∼13% activity of native 70S ribosomes, which increased to 21% when supplemented with GroEL/ES. This work also points to what is still needed to obtain self-replicating synthetic ribosomes in situ in the PURE system.
- 188Murase, Y.; Nakanishi, H.; Tsuji, G.; Sunami, T.; Ichihashi, N. In Vitro Evolution of Unmodified 16S rRNA for Simple Ribosome Reconstitution. ACS Synth. Biol. 2018, 7 (2), 576– 583, DOI: 10.1021/acssynbio.7b00333Google ScholarThere is no corresponding record for this reference.
- 189Jewett, M. C.; Fritz, B. R.; Timmerman, L. E.; Church, G. M. In Vitro Integration of Ribosomal RNA Synthesis, Ribosome Assembly, and Translation. Mol. Syst. Biol. 2013, 9, 678, DOI: 10.1038/msb.2013.31Google Scholar189https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtFSqsLbM&md5=65e884f5f49acc8f173da9b949a81237In vitro integration of ribosomal RNA synthesis, ribosome assembly, and translationJewett, Michael C.; Fritz, Brian R.; Timmerman, Laura E.; Church, George M.Molecular Systems Biology (2013), 9 (), 678CODEN: MSBOC3; ISSN:1744-4292. (Nature Publishing Group)Purely in vitro ribosome synthesis could provide a crit. step towards unraveling the systems biol. of ribosome biogenesis, constructing minimal cells from defined components, and engineering ribosomes with new functions. Here, as an initial step towards this goal, we report a method for constructing Escherichia coli ribosomes in crude S150 E. coli exts. While conventional methods for E. coli ribosome reconstitution are non-physiol., our approach attempts to mimic chem. conditions in the cytoplasm, thus permitting several biol. processes to occur simultaneously. Specifically, our integrated synthesis, assembly, and translation (iSAT) technol. enables one-step co-activation of rRNA transcription, assembly of transcribed rRNA with native ribosomal proteins into functional ribosomes, and synthesis of active protein by these ribosomes in the same compartment. We show that iSAT makes possible the in vitro construction of modified ribosomes by introducing a 23S rRNA mutation that mediates resistance against clindamycin. We anticipate that iSAT will aid studies of ribosome assembly and open new avenues for making ribosomes with altered properties.
- 190Fritz, B. R.; Jamil, O. K.; Jewett, M. C. Implications of Macromolecular Crowding and Reducing Conditions for in Vitro Ribosome Construction. Nucleic Acids Res. 2015, 43 (9), 4774– 4784, DOI: 10.1093/nar/gkv329Google Scholar190https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsFaisbzI&md5=3183327da923d2c7d665b3b7a33c4299Implications of macromolecular crowding and reducing conditions for in vitro ribosome constructionFritz, Brian R.; Jamil, Osman K.; Jewett, Michael C.Nucleic Acids Research (2015), 43 (9), 4774-4784CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)The in vitro construction of Escherichia coli ribosomes could elucidate a deeper understanding of these complex mol. machines and make possible the prodn. of synthetic variants with new functions. Toward this goal, the authors recently developed an integrated synthesis, assembly, and translation (iSAT) system that allows for co-activation of rRNA transcription and ribosome assembly, mRNA transcription, and protein translation without intact cells. Here, the authors discovered that macromol. crowding and reducing agents increased overall iSAT protein synthesis; the combination of 6% Ficoll 400 and 2 mM DTBA yielded an approx. 5-fold increase in overall iSAT protein synthesis activity. By utilizing a fluorescent RNA aptamer, fluorescent reporter proteins, and ribosome sedimentation anal., the authors showed that crowding agents increased iSAT yields by enhancing translation while reducing agents increased rRNA transcription and ribosome assembly. Finally, the authors showed that iSAT ribosomes possessed ∼70% of the protein synthesis activity of in vivo-assembled E. coli ribosomes. This work improved iSAT protein synthesis through the addn. of crowding and reducing agents, provides a thorough understanding of the effect of these additives within the iSAT system and demonstrated how iSAT allows for manipulation and anal. of ribosome biogenesis in the context of an in vitro transcription-translation system.
- 191Hammerling, M. J.; Fritz, B. R.; Yoesep, D. J.; Kim, D. S.; Carlson, E. D.; Jewett, M. C. In Vitro Ribosome Synthesis and Evolution through Ribosome Display. Nat. Commun. 2020, 11 (1), 1108, DOI: 10.1038/s41467-020-14705-2Google Scholar191https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXkvFaisbY%253D&md5=7a7004c5d0c04e4727b54c6d3c5723ccIn-vitro ribosome synthesis and evolution through ribosome displayHammerling, Michael J.; Fritz, Brian R.; Yoesep, Danielle J.; Kim, Do Soon; Carlson, Erik D.; Jewett, Michael C.Nature Communications (2020), 11 (1), 1108CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)Directed evolution of the ribosome for expanded substrate incorporation and novel functions is challenging because the requirement of cell viability limits the mutations that can be made. Here we address this challenge by combining cell-free synthesis and assembly of translationally competent ribosomes with ribosome display to develop a fully in-vitro methodol. for ribosome synthesis and evolution (called RISE). We validate the RISE method by selecting active genotypes from a ∼1.7 X 107 member library of rRNA (rRNA) variants, as well as identifying mutant ribosomes resistant to the antibiotic clindamycin from a library of ∼4 X 103 rRNA variants. We further demonstrate the prevalence of pos. epistasis in resistant genotypes, highlighting the importance of such interactions in selecting for new function. We anticipate that RISE will facilitate understanding of mol. translation and enable selection of ribosomes with altered properties.
- 192Wang, T.; Lu, Y. Toward Minimal Transcription-Translation Machinery. ACS Synth. Biol. 2023, 12 (11), 3312– 3327, DOI: 10.1021/acssynbio.3c00324Google ScholarThere is no corresponding record for this reference.
- 193Oberholzer, T.; Albrizio, M.; Luisi, P. L. Polymerase Chain Reaction in Liposomes. Chem. Biol. 1995, 2 (10), 677– 682, DOI: 10.1016/1074-5521(95)90031-4Google Scholar193https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2MXptFygtbc%253D&md5=297f9e6efcf5d03c3ab4810317037320Polymerase chain reaction in liposomesOberholzer, Thomas; Albrizio, Maria; Luisi, Pier LuigiChemistry & Biology (1995), 2 (10), 677-82CODEN: CBOLE2; ISSN:1074-5521. (Current Biology)Background: Compartmentalization of biochem. reactions within a spherically closed bilayer is an important step in the mol. evolution of cells. Liposomes are the most suitable structures to model this kind of chem. We have used the polymerase chain reaction (PCR) to demonstrate that complex biochem. reactions such as DNA replication can be carried out inside these compartments. Results: We describe the first example of DNA amplification by the PCR occurring inside liposomes composed of 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC), or of a mixt. of POPC and phosphatidylserine. We show that these liposomes are stable even under the high temp. conditions used for PCR. Although only a very small fraction of liposomes contains all eight different reagents together, a significant amt. of DNA is produced which can be obsd. by polyacrylamide gel electrophoresis. Conclusions: This work shows that it is possible to carry out complex biochem. reactions within liposomes, which may be germane to the question of the origin of living cells. We have established the parameters and conditions that are crit. for carrying out this complex reaction within the liposome compartment.
- 194Kawska, A.; Carvalho, K.; Manzi, J.; Boujemaa-Paterski, R.; Blanchoin, L.; Martiel, J.-L.; Sykes, C. How Actin Network Dynamics Control the Onset of Actin-Based Motility. Proc. Natl. Acad. Sci. U. S. A. 2012, 109 (36), 14440– 14445, DOI: 10.1073/pnas.1117096109Google ScholarThere is no corresponding record for this reference.
- 195Hürtgen, D.; Härtel, T.; Murray, S. M.; Sourjik, V.; Schwille, P. Functional Modules of Minimal Cell Division for Synthetic Biology. Adv. Biosyst. 2019, 3 (6), e1800315, DOI: 10.1002/adbi.201800315Google ScholarThere is no corresponding record for this reference.
- 196Pelletier, J. F.; Sun, L.; Wise, K. S.; Assad-Garcia, N.; Karas, B. J.; Deerinck, T. J.; Ellisman, M. H.; Mershin, A.; Gershenfeld, N.; Chuang, R.-Y.; Glass, J. I.; Strychalski, E. A. Genetic Requirements for Cell Division in a Genomically Minimal Cell. Cell 2021, 184 (9), 2430– 2440, DOI: 10.1016/j.cell.2021.03.008Google Scholar196https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXns1Cisrs%253D&md5=f2e8dc01302d6eac0e75d1d4d1597dc2Genetic requirements for cell division in a genomically minimal cellPelletier, James F.; Sun, Lijie; Wise, Kim S.; Assad-Garcia, Nacyra; Karas, Bogumil J.; Deerinck, Thomas J.; Ellisman, Mark H.; Mershin, Andreas; Gershenfeld, Neil; Chuang, Ray-Yuan; Glass, John I.; Strychalski, Elizabeth A.Cell (Cambridge, MA, United States) (2021), 184 (9), 2430-2440.e16CODEN: CELLB5; ISSN:0092-8674. (Cell Press)Genomically minimal cells, such as JCVI-syn3.0, offer a platform to clarify genes underlying core physiol. processes. Although this minimal cell includes genes essential for population growth, the physiol. of its single cells remained uncharacterized. To investigate striking morphol. variation in JCVI-syn3.0 cells, we present an approach to characterize cell propagation and det. genes affecting cell morphol. Microfluidic chemostats allowed observation of intrinsic cell dynamics that result in irregular morphologies. A genome with 19 genes not retained in JCVI-syn3.0 generated JCVI-syn3A, which presents morphol. similar to that of JCVI-syn1.0. We further identified seven of these 19 genes, including two known cell division genes, ftsZ and sepF, a hydrolase of unknown substrate, and four genes that encode membrane-assocd. proteins of unknown function, which are required together to restore a phenotype similar to that of JCVI-syn1.0. This result emphasizes the polygenic nature of cell division and morphol. in a genomically minimal cell. The fully annotated genome sequence of JCVI-syn3A is deposited in NCBI (GenBank: CP016816.2).
- 197Zhang, S.; Contini, C.; Hindley, J. W.; Bolognesi, G.; Elani, Y.; Ces, O. Engineering Motile Aqueous Phase-Separated Droplets via Liposome Stabilisation. Nat. Commun. 2021, 12 (1), 1673, DOI: 10.1038/s41467-021-21832-xGoogle Scholar197https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXmslKhtbc%253D&md5=d911a2717fe36d0872045e44ef9edf56Engineering motile aqueous phase-separated droplets via liposome stabilisationZhang, Shaobin; Contini, Claudia; Hindley, James W.; Bolognesi, Guido; Elani, Yuval; Ces, OscarNature Communications (2021), 12 (1), 1673CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)There are increasing efforts to engineer functional compartments that mimic cellular behaviors from the bottom-up. One behavior that is receiving particular attention is motility, due to its biotechnol. potential and ubiquity in living systems. Many existing platforms make use of the Marangoni effect to achieve motion in water/oil (w/o) droplet systems. However, most of these systems are unsuitable for biol. applications due to biocompatibility issues caused by the presence of oil phases. Here we report a biocompatible all aq. (wt./wt.) PEG/dextran Pickering-like emulsion system consisting of liposome-stabilized cell-sized droplets, where the stability can be easily tuned by adjusting liposome compn. and concn. We demonstrate that the compartments are capable of neg. chemotaxis: these droplets can respond to a PEG/dextran polymer gradient through directional motion down to the gradient. The biocompatibility, motility and partitioning abilities of this droplet system offers new directions to pursue research in motion-related biol. processes.
- 198Li, M.; Brinkmann, M.; Pagonabarraga, I.; Seemann, R.; Fleury, J.-B. Spatiotemporal Control of Cargo Delivery Performed by Programmable Self-Propelled Janus Droplets. Commun. Phys. 2018, 1 (1), 1– 8, DOI: 10.1038/s42005-018-0025-4Google Scholar198https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXpvV2lurk%253D&md5=25c2517f251115af25f3cfced1c1ab32Breakdown of diffusivity-entropy scaling in colloidal glass-forming liquidsLi, Bo; Xiao, Xiuming; Lou, Kai; Wang, Shuxia; Wen, Weijia; Wang, ZirenCommunications Physics (2018), 1 (1), 1-9CODEN: CPOHDJ; ISSN:2399-3650. (Nature Research)Glass is a liq. that has lost its ability to flow. Why this particular substance undergoes such a dramatic kinetic slowdown yet remains barely distinguishable in structure from its fluid state upon cooling constitutes the central question of glass transition physics. Here, we investigate the pathway of kinetic slowdown in glass-forming liqs. that consist of monolayers of ellipsoidal or binary spherical colloids. In contrast to rotational motion, the dynamics of the translational motion begin to violently slow down at considerably low area fractions (.vphi.T). At .vphi.T, anomalous translation-rotation coupling is enhanced and the topog. of the free energy landscape become rugged. Based on the pos. correlation between .vphi.T and fragility, the measurement of .vphi.T offers a novel method for predicting glassy dynamics, circumventing the prohibitive increase in equil. times required in high-d. regions. Our results highlight the role that thermodynamical entropy plays in glass transitions.
- 199Litschel, T.; Kelley, C. F.; Holz, D.; Adeli Koudehi, M.; Vogel, S. K.; Burbaum, L.; Mizuno, N.; Vavylonis, D.; Schwille, P. Reconstitution of Contractile Actomyosin Rings in Vesicles. Nat. Commun. 2021, 12 (1), 2254, DOI: 10.1038/s41467-021-22422-7Google Scholar199https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXptFOqsL8%253D&md5=5f2aaa315f2c39163e5b7e59c7b2aacbReconstitution of contractile actomyosin rings in vesiclesLitschel, Thomas; Kelley, Charlotte F.; Holz, Danielle; Adeli Koudehi, Maral; Vogel, Sven K.; Burbaum, Laura; Mizuno, Naoko; Vavylonis, Dimitrios; Schwille, PetraNature Communications (2021), 12 (1), 2254CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)One of the grand challenges of bottom-up synthetic biol. is the development of minimal machineries for cell division. The mech. transformation of large-scale compartments, such as Giant Unilamellar Vesicles (GUVs), requires the geometry-specific coordination of active elements, several orders of magnitude larger than the mol. scale. Of all cytoskeletal structures, large-scale actomyosin rings appear to be the most promising cellular elements to accomplish this task. Here, we have adopted advanced encapsulation methods to study bundled actin filaments in GUVs and compare our results with theor. modeling. By changing few key parameters, actin polymn. can be differentiated to resemble various types of networks in living cells. Importantly, we find membrane binding to be crucial for the robust condensation into a single actin ring in spherical vesicles, as predicted by theor. considerations. Upon force generation by ATP-driven myosin motors, these ring-like actin structures contract and locally constrict the vesicle, forming furrow-like deformations. On the other hand, cortex-like actin networks are shown to induce and stabilize deformations from spherical shapes.
- 200Bashirzadeh, Y.; Moghimianavval, H.; Liu, A. P. Encapsulated Actomyosin Patterns Drive Cell-like Membrane Shape Changes. iScience 2022, 25 (5), 104236, DOI: 10.1016/j.isci.2022.104236Google Scholar200https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhslGksbjK&md5=ed9418fdac1a79597adabe19c16c5de2Encapsulated actomyosin patterns drive cell-like membrane shape changesBashirzadeh, Yashar; Moghimianavval, Hossein; Liu, Allen P.iScience (2022), 25 (5), 104236CODEN: ISCICE; ISSN:2589-0042. (Elsevier B.V.)Cell shape changes from locomotion to cytokinesis are, to a large extent, driven by myosin-driven remodeling of cortical actin patterns. Passive crosslinkers such as α-actinin and fascin as well as actin nucleator Arp2/3 complex largely det. actin network architecture and, consequently, membrane shape changes. Here we reconstitute actomyosin networks inside cell-sized lipid bilayer vesicles and show that depending on vesicle size and concns. of α-actinin and fascin actomyosin networks assemble into ring and aster-like patterns. Anchoring actin to the membrane does not change actin network architecture yet exerts forces and deforms the membrane when assembled in the form of a contractile ring. In the presence of α-actinin and fascin, an Arp2/3 complex-mediated actomyosin cortex is shown to assemble a ring-like pattern at the equatorial cortex followed by myosin-driven clustering and consequently blebbing. An active gel theory unifies a model for the obsd. membrane shape changes induced by the contractile cortex.
- 201Gardner, P. M.; Winzer, K.; Davis, B. G. Sugar Synthesis in a Protocellular Model Leads to a Cell Signalling Response in Bacteria. Nat. Chem. 2009, 1 (5), 377– 383, DOI: 10.1038/nchem.296Google Scholar201https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXptVSjsrY%253D&md5=ad496ab10f8ec2775e43fbae9d4b25e8Sugar synthesis in a protocellular model leads to a cell signalling response in bacteriaGardner, Paul M.; Winzer, Klaus; Davis, Benjamin G.Nature Chemistry (2009), 1 (5), 377-383, S377/1-S377/50CODEN: NCAHBB; ISSN:1755-4330. (Nature Publishing Group)The design of systems with life-like properties from simple chem. components may offer insights into biol. processes, with the ultimate goal of creating an artificial chem. cell that would be considered to be alive. Most efforts to create artificial cells have concd. on systems based on complex natural mols. such as DNA and RNA. Here we have constructed a lipid-bound protometabolism that synthesizes complex carbohydrates from simple feedstocks, which are capable of engaging the natural quorum sensing mechanism of the marine bacterium Vibrio harveyi and stimulating a proportional bioluminescent response. This encapsulated system may represent the first step towards the realization of a cellular mimic' and a starting point for bottom-up' designs of other chem. cells, which could perhaps display complex behaviors such as communication with natural cells.
- 202Lentini, R.; Santero, S. P.; Chizzolini, F.; Cecchi, D.; Fontana, J.; Marchioretto, M.; Del Bianco, C.; Terrell, J. L.; Spencer, A. C.; Martini, L.; Forlin, M.; Assfalg, M.; Dalla Serra, M.; Bentley, W. E.; Mansy, S. S. Integrating Artificial with Natural Cells to Translate Chemical Messages That Direct E. Coli Behaviour. Nat. Commun. 2014, 5, 4012, DOI: 10.1038/ncomms5012Google Scholar202https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhvF2mur3K&md5=d80e7b3bbd5f02d32806c289530c7bc0Integrating artificial with natural cells to translate chemical messages that direct E. coli behaviourLentini, Roberta; Santero, Silvia Perez; Chizzolini, Fabio; Cecchi, Dario; Fontana, Jason; Marchioretto, Marta; Del Bianco, Cristina; Terrell, Jessica L.; Spencer, Amy C.; Martini, Laura; Forlin, Michele; Assfalg, Michael; Dalla Serra, Mauro; Bentley, William E.; Mansy, Sheref S.Nature Communications (2014), 5 (), 4012CODEN: NCAOBW; ISSN:2041-1723. (Nature Publishing Group)Previous efforts to control cellular behavior have largely relied upon various forms of genetic engineering. Once the genetic content of a living cell is modified, the behavior of that cell typically changes as well. However, other methods of cellular control are possible. All cells sense and respond to their environment. Therefore, artificial, non-living cellular mimics could be engineered to activate or repress already existing natural sensory pathways of living cells through chem. communication. Here we describe the construction of such a system. The artificial cells expand the senses of Escherichia coli by translating a chem. message that E. coli cannot sense on its own to a mol. that activates a natural cellular response. This methodol. could open new opportunities in engineering cellular behavior without exploiting genetically modified organisms.
- 203Ding, Y.; Contreras-Llano, L. E.; Morris, E.; Mao, M.; Tan, C. Minimizing Context Dependency of Gene Networks Using Artificial Cells. ACS Appl. Mater. Interfaces 2018, 10 (36), 30137– 30146, DOI: 10.1021/acsami.8b10029Google ScholarThere is no corresponding record for this reference.
- 204Toparlak, Ö. D.; Zasso, J.; Bridi, S.; Serra, M. D.; Macchi, P.; Conti, L.; Baudet, M.-L.; Mansy, S. S. Artificial Cells Drive Neural Differentiation. Sci. Adv. 2020, DOI: 10.1126/sciadv.abb4920Google ScholarThere is no corresponding record for this reference.
- 205Fanalista, F.; Birnie, A.; Maan, R.; Burla, F.; Charles, K.; Pawlik, G.; Deshpande, S.; Koenderink, G. H.; Dogterom, M.; Dekker, C. Shape and Size Control of Artificial Cells for Bottom-Up Biology. ACS Nano 2019, 13 (5), 5439– 5450, DOI: 10.1021/acsnano.9b00220Google Scholar205https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXptlKrtLw%253D&md5=10854f36682b1ae84c7c9bea92f127edShape and Size Control of Artificial Cells for Bottom-Up BiologyFanalista, Federico; Birnie, Anthony; Maan, Renu; Burla, Federica; Charles, Kevin; Pawlik, Grzegorz; Deshpande, Siddharth; Koenderink, Gijsje H.; Dogterom, Marileen; Dekker, CeesACS Nano (2019), 13 (5), 5439-5450CODEN: ANCAC3; ISSN:1936-0851. (American Chemical Society)Bottom-up biol. is an expanding research field that aims to understand the mechanisms underlying biol. processes via in vitro assembly of their essential components in synthetic cells. As encapsulation and controlled manipulation of these elements is a crucial step in the recreation of such cell-like objects, microfluidics is increasingly used for the prodn. of minimal artificial containers such as single-emulsion droplets, double-emulsion droplets, and liposomes. Despite the importance of cell morphol. on cellular dynamics, current synthetic-cell studies mainly use spherical containers, and methods to actively shape manipulate these have been lacking. In this paper, the authors describe a microfluidic platform to deform the shape of artificial cells into a variety of shapes (rods and disks) with adjustable cell-like dimensions below 5 μm, thereby mimicking realistic cell morphologies. To illustrate the potential of the method, the authors reconstitute three biol. relevant protein systems (FtsZ, microtubules, collagen) inside rod-shaped containers and study the arrangement of the protein networks inside these synthetic containers with physiol. relevant morphologies resembling those found in living cells.
- 206Van de Cauter, L.; Fanalista, F.; van Buren, L.; De Franceschi, N.; Godino, E.; Bouw, S.; Danelon, C.; Dekker, C.; Koenderink, G. H.; Ganzinger, K. A. Optimized cDICE for Efficient Reconstitution of Biological Systems in Giant Unilamellar Vesicles. ACS Synth. Biol. 2021, 10 (7), 1690– 1702, DOI: 10.1021/acssynbio.1c00068Google Scholar206https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhsVWrt7zP&md5=b34708aa4daf3694358fa8fa0b4dfa3cOptimized cDICE for Efficient Reconstitution of Biological Systems in Giant Unilamellar VesiclesVan de Cauter, Lori; Fanalista, Federico; van Buren, Lennard; De Franceschi, Nicola; Godino, Elisa; Bouw, Sharon; Danelon, Christophe; Dekker, Cees; Koenderink, Gijsje H.; Ganzinger, Kristina A.ACS Synthetic Biology (2021), 10 (7), 1690-1702CODEN: ASBCD6; ISSN:2161-5063. (American Chemical Society)Giant unilamellar vesicles (GUVs) are often used to mimic biol. membranes in reconstitution expts. They are also widely used in research on synthetic cells, as they provide a mech. responsive reaction compartment that allows for controlled exchange of reactants with the environment. However, while many methods exist to encapsulate functional biomols. in GUVs, there is no one-size-fits-all soln. and reliable GUV fabrication still remains a major exptl. hurdle in the field. Here, we show that defect-free GUVs contg. complex biochem. systems can be generated by optimizing a double-emulsion method for GUV formation called continuous droplet interface crossing encapsulation (cDICE). By tightly controlling environmental conditions and tuning the lipid-in-oil dispersion, we show that it is possible to significantly improve the reproducibility of high-quality GUV formation as well as the encapsulation efficiency. We demonstrate efficient encapsulation for a range of biol. systems including a minimal actin cytoskeleton, membrane-anchored DNA nanostructures, and a functional PURE (protein synthesis using recombinant elements) system. Our optimized cDICE method displays promising potential to become a std. method in biophysics and bottom-up synthetic biol.
- 207Ganar, K. A.; Leijten, L.; Deshpande, S. Actinosomes: Condensate-Templated Containers for Engineering Synthetic Cells. ACS Synth. Biol. 2022, 11 (8), 2869– 2879, DOI: 10.1021/acssynbio.2c00290Google Scholar207https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XitVyjsLvM&md5=9c7b4edc9a9e23d40018ea0eef031085Actinosomes: Condensate-Templated Containers for Engineering Synthetic CellsGanar, Ketan A.; Leijten, Liza; Deshpande, SiddharthACS Synthetic Biology (2022), 11 (8), 2869-2879CODEN: ASBCD6; ISSN:2161-5063. (American Chemical Society)Engineering synthetic cells has a broad appeal, from understanding living cells to designing novel biomaterials for therapeutics, biosensing, and hybrid interfaces. A key prerequisite to creating synthetic cells is a three-dimensional container capable of orchestrating biochem. reactions. In this study, we present an easy and effective technique to make cell-sized porous containers, coined actinosomes, using the interactions between biomol. condensates and the actin cytoskeleton. This approach uses polypeptide/nucleoside triphosphate condensates and localizes actin monomers on their surface. By triggering actin polymn. and using osmotic gradients, the condensates are transformed into containers, with the boundary made up of actin filaments and polylysine polymers. We show that the guanosine triphosphate (GTP)-to-ATP (ATP) ratio is a crucial parameter for forming actinosomes: insufficient ATP prevents condensate dissoln., while excess ATP leads to undesired crumpling. Permeability studies reveal the porous surface of actinosomes, allowing small mols. to pass through while restricting bigger macromols. within the interior. We show the functionality of actinosomes as bioreactors by carrying out in vitro protein translation within them. Actinosomes are a handy addn. to the synthetic cell platform, with appealing properties like ease of prodn., inherent encapsulation capacity, and a potentially active surface to trigger signaling cascades and form multicellular assemblies, conceivably useful for biotechnol. applications.
- 208Frischmon, C.; Sorenson, C.; Winikoff, M.; Adamala, K. P. Build-a-Cell: Engineering a Synthetic Cell Community. Life 2021, 11 (11), 1176, DOI: 10.3390/life11111176Google ScholarThere is no corresponding record for this reference.
- 209Schwille, P.; Spatz, J.; Landfester, K.; Bodenschatz, E.; Herminghaus, S.; Sourjik, V.; Erb, T. J.; Bastiaens, P.; Lipowsky, R.; Hyman, A.; Dabrock, P.; Baret, J.-C.; Vidakovic-Koch, T.; Bieling, P.; Dimova, R.; Mutschler, H.; Robinson, T.; Tang, T.-Y. D.; Wegner, S.; Sundmacher, K. MaxSynBio: Avenues Towards Creating Cells from the Bottom Up. Angew. Chem., Int. Ed. Engl. 2018, 57 (41), 13382– 13392, DOI: 10.1002/anie.201802288Google Scholar209https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1MfitFWnsA%253D%253D&md5=869c39a9f651f1fcbb33665f2d5c97a9MaxSynBio: Avenues Towards Creating Cells from the Bottom UpSchwille Petra; Mutschler Hannes; Spatz Joachim; Landfester Katharina; Wegner Seraphine; Bodenschatz Eberhard; Herminghaus Stephan; Sourjik Victor; Erb Tobias J; Bastiaens Philippe; Bieling Peter; Lipowsky Reinhard; Dimova Rumiana; Robinson Tom; Hyman Anthony; Tang T-Y Dora; Dabrock Peter; Baret Jean-Christophe; Vidakovic-Koch Tanja; Sundmacher KaiAngewandte Chemie (International ed. in English) (2018), 57 (41), 13382-13392 ISSN:.A large German research consortium mainly within the Max Planck Society ("MaxSynBio") was formed to investigate living systems from a fundamental perspective. The research program of MaxSynBio relies solely on the bottom-up approach to synthetic biology. MaxSynBio focuses on the detailed analysis and understanding of essential processes of life through modular reconstitution in minimal synthetic systems. The ultimate goal is to construct a basic living unit entirely from non-living components. The fundamental insights gained from the activities in MaxSynBio could eventually be utilized for establishing a new generation of biotechnological processes, which would be based on synthetic cell constructs that replace the natural cells currently used in conventional biotechnology.
- 210Habets, M. G. J. L.; Zwart, H. A. E.; van Est, R. Why the Synthetic Cell Needs Democratic Governance. Trends Biotechnol. 2021, 39 (6), 539– 541, DOI: 10.1016/j.tibtech.2020.11.006Google ScholarThere is no corresponding record for this reference.
- 211Rasmussen, S. Protocells: Bridging Nonliving and Living Matter; MIT Press, 2008.Google ScholarThere is no corresponding record for this reference.
- 212Schwille, P. Bottom-up Synthetic Biology: Engineering in a Tinkerer’s World. Science 2011, 333 (6047), 1252– 1254, DOI: 10.1126/science.1211701Google Scholar212https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtV2jt7vF&md5=f390108f5f3ffacc15a14093ef4277c3Bottom-Up Synthetic Biology: Engineering in a Tinkerer's WorldSchwille, PetraScience (Washington, DC, United States) (2011), 333 (6047), 1252-1254CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)A review. How synthetic can "synthetic biol." be A literal interpretation of the name of this new life science discipline invokes expectations of the systematic construction of biol. systems with cells being built module by module-from the bottom up. But can this possibly be achieved, taking into account the enormous complexity and redundancy of living systems, which distinguish them quite remarkably from design features that characterize human inventions There are several recent developments in biol., in tight conjunction with quant. disciplines, that may bring this literal perspective into the realm of the possible. However, such bottom-up engineering requires tools that were originally designed by nature's greatest tinkerer: evolution.
- 213Abil, Z.; Danelon, C. Roadmap to Building a Cell: An Evolutionary Approach. Front. Bioeng. Biotechnol. 2020, 8, 927, DOI: 10.3389/fbioe.2020.00927Google Scholar213https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3s%252Fjt1Ontg%253D%253D&md5=1453ab51194499edbf69bf236a3e6c64Roadmap to Building a Cell: An Evolutionary ApproachAbil Zhanar; Danelon ChristopheFrontiers in bioengineering and biotechnology (2020), 8 (), 927 ISSN:2296-4185.Laboratory synthesis of an elementary biological cell from isolated components may aid in understanding of the fundamental principles of life and will provide a platform for a range of bioengineering and medical applications. In essence, building a cell consists in the integration of cellular modules into system's level functionalities satisfying a definition of life. To achieve this goal, we propose in this perspective to undertake a semi-rational, system's level evolutionary approach. The strategy would require iterative cycles of genetic integration of functional modules, diversification of hereditary information, compartmentalized gene expression, selection/screening, and possibly, assistance from open-ended evolution. We explore the underlying challenges to each of these steps and discuss possible solutions toward the bottom-up construction of an artificial living cell.
- 214Yarmuth; [d-Ky-3], J. A. Inflation Reduction Act of 2022 ; 2022. http://www.congress.gov/ (accessed on April 30, 2023).Google ScholarThere is no corresponding record for this reference.
- 215The White House. Executive order on advancing biotechnology and biomanufacturing innovation for a sustainable, safe, and secure American bioeconomy. The White House. https://www.whitehouse.gov/briefing-room/presidential-actions/2022/09/12/executive-order-on-advancing-biotechnology-and-biomanufacturing-innovation-for-a-sustainable-safe-and-secure-american-bioeconomy/ (accessed on April 30, 2023).Google ScholarThere is no corresponding record for this reference.
- 216BioMADE. https://www.biomade.org/ (accessed on April 30, 2023).Google ScholarThere is no corresponding record for this reference.
- 217Huang, A.; Nguyen, P. Q.; Stark, J. C.; Takahashi, M. K.; Donghia, N.; Ferrante, T.; Dy, A. J.; Hsu, K. J.; Dubner, R. S.; Pardee, K.; Jewett, M. C.; Collins, J. J. BioBitsTM Explorer: A Modular Synthetic Biology Education Kit. Sci. Adv. 2018, 4 (8), eaat5105, DOI: 10.1126/sciadv.aat5105Google ScholarThere is no corresponding record for this reference.
- 218Stark, J. C.; Huang, A.; Nguyen, P. Q.; Dubner, R. S.; Hsu, K. J.; Ferrante, T. C.; Anderson, M.; Kanapskyte, A.; Mucha, Q.; Packett, J. S.; Patel, P.; Patel, R.; Qaq, D.; Zondor, T.; Burke, J.; Martinez, T.; Miller-Berry, A.; Puppala, A.; Reichert, K.; Schmid, M.; Brand, L.; Hill, L. R.; Chellaswamy, J. F.; Faheem, N.; Fetherling, S.; Gong, E.; Gonzalzles, E. M.; Granito, T.; Koritsaris, J.; Nguyen, B.; Ottman, S.; Palffy, C.; Patel, A.; Skweres, S.; Slaton, A.; Woods, T.; Donghia, N.; Pardee, K.; Collins, J. J.; Jewett, M. C. BioBitsTM Bright: A Fluorescent Synthetic Biology Education Kit. Sci. Adv. 2018, 4 (8), eaat5107, DOI: 10.1126/sciadv.aat5107Google ScholarThere is no corresponding record for this reference.
- 219Stark, J. C.; Huang, A.; Hsu, K. J.; Dubner, R. S.; Forbrook, J.; Marshalla, S.; Rodriguez, F.; Washington, M.; Rybnicky, G. A.; Nguyen, P. Q.; Hasselbacher, B.; Jabri, R.; Kamran, R.; Koralewski, V.; Wightkin, W.; Martinez, T.; Jewett, M. C. BioBits Health: Classroom Activities Exploring Engineering, Biology, and Human Health with Fluorescent Readouts. ACS Synth. Biol. 2019, 8 (5), 1001– 1009, DOI: 10.1021/acssynbio.8b00381Google Scholar219https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXmt1Olu70%253D&md5=99551c2017559b0b1275e1d168dfb5a4BioBits Health: Classroom Activities Exploring Engineering, Biology, and Human Health with Fluorescent ReadoutsStark, Jessica C.; Huang, Ally; Hsu, Karen J.; Dubner, Rachel S.; Forbrook, Jason; Marshalla, Suzanne; Rodriguez, Faith; Washington, Mechelle; Rybnicky, Grant A.; Nguyen, Peter Q.; Hasselbacher, Brenna; Jabri, Ramah; Kamran, Rijha; Koralewski, Veronica; Wightkin, Will; Martinez, Thomas; Jewett, Michael C.ACS Synthetic Biology (2019), 8 (5), 1001-1009CODEN: ASBCD6; ISSN:2161-5063. (American Chemical Society)Recent advances in synthetic biol. have resulted in biol. technologies with the potential to reshape the way we understand and treat human disease. Educating students about the biol. and ethics underpinning these technologies is crit. to empower them to make informed future policy decisions regarding their use and to inspire the next generation of synthetic biologists. However, hands-on, educational activities that convey emerging synthetic biol. topics can be difficult to implement due to the expensive equipment and expertise required to grow living cells. We present BioBits Health, an educational kit contg. lab activities and supporting curricula for teaching antibiotic resistance mechanisms and CRISPR-Cas9 gene editing in high school classrooms. This kit links complex biol. concepts to visual, fluorescent readouts in user-friendly freeze-dried cell-free reactions. BioBits Health represents a set of educational resources that promises to encourage teaching of cutting-edge, health-related synthetic biol. topics in classrooms and other nonlab. settings.
- 220Rybnicky, G. A.; Dixon, R. A.; Kuhn, R. M.; Karim, A. S.; Jewett, M. C. Development of a Freeze-Dried CRISPR-Cas12 Sensor for Detecting Wolbachia in the Secondary Science Classroom. ACS Synth. Biol. 2022, 11 (2), 835– 842, DOI: 10.1021/acssynbio.1c00503Google ScholarThere is no corresponding record for this reference.
- 221Jung, K. J.; Rasor, B. J.; Rybnicky, G. A.; Silverman, A. D.; Standeven, J.; Kuhn, R.; Granito, T.; Ekas, H. M.; Wang, B. M.; Karim, A. S.; Lucks, J. B.; Jewett, M. C. At-Home, Cell-Free Synthetic Biology Education Modules for Transcriptional Regulation and Environmental Water Quality Monitoring. bioRxiv , January 9, 2023. DOI: 10.1101/2023.01.09.523248 .Google ScholarThere is no corresponding record for this reference.
- 222Perry, E.; Weber, J.; Pataranutaporn, P.; Volf, V.; Gonzalez, L. M.; Nejad, S.; Angleton, C.; Chen, J.-E.; Gabo, A.; Jammalamadaka, M. S. S.; Kuru, E.; Fortuna, P.; Rico, A.; Sulich, K.; Wawrzyniak, D.; Jacobson, J.; Church, G.; Kong, D. How to Grow (almost) Anything: A Hybrid Distance Learning Model for Global Laboratory-Based Synthetic Biology Education. Nat. Biotechnol. 2022, 40 (12), 1874– 1879, DOI: 10.1038/s41587-022-01601-xGoogle Scholar222https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XjtFSlsLzF&md5=5a8f9f979f93a31e57c86f0bca9f1a05How to grow (almost) anything: a hybrid distance learning model for global laboratory-based synthetic biology educationPerry, Eyal; Weber, Jessica; Pataranutaporn, Pat; Volf, Verena; Gonzalez, Laura Maria; Nejad, Sara; Angleton, Carolyn; Chen, Jia-En; Gabo, Ananda; Jammalamadaka, Mani Sai Suryateja; Kuru, Erkin; Fortuna, Patrick; Rico, Andres; Sulich, Karolina; Wawrzyniak, Dominika; Jacobson, Joseph; Church, George; Kong, DavidNature Biotechnology (2022), 40 (12), 1874-1879CODEN: NABIF9; ISSN:1087-0156. (Nature Portfolio)A pilot program for synthetic biol. education via a scalable distributed network model of distance-based lab. learning can be accessible globally across disciplines and backgrounds.
Cited By
Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by ACS Publications if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
This article is cited by 16 publications.
- Surendra Yadav, Alexander J. P. Perkins, Sahan B. W. Liyanagedera, Anthony Bougas, Nadanai Laohakunakorn. ATP Regeneration from Pyruvate in the PURE System. ACS Synthetic Biology 2025, 14
(1)
, 247-256. https://doi.org/10.1021/acssynbio.4c00697
- Manisha Kapasiawala, Richard M. Murray. Metabolic Perturbations to an Escherichia coli-based Cell-Free System Reveal a Trade-off between Transcription and Translation. ACS Synthetic Biology 2024, 13
(12)
, 3976-3990. https://doi.org/10.1021/acssynbio.4c00361
- Lan Hai Anh Tran, Lauren A. Lowe, Yaam Deckel, Matthew Turner, James Luong, Omar Abdullah A Khamis, Megan L. Amos, Anna Wang. Measuring Vesicle Loading with Holographic Microscopy and Bulk Light Scattering. ACS Physical Chemistry Au 2024, 4
(4)
, 400-407. https://doi.org/10.1021/acsphyschemau.4c00011
- Yuishin Kosaka, Yumi Miyawaki, Megumi Mori, Shunsuke Aburaya, Chisato Nishizawa, Takeshi Chujo, Tatsuya Niwa, Takumi Miyazaki, Takashi Sugita, Mao Fukuyama, Hideki Taguchi, Kazuhito Tomizawa, Kenji Sugase, Mitsuyoshi Ueda, Wataru Aoki. Autonomous ribosome biogenesis in vitro. Nature Communications 2025, 16
(1)
https://doi.org/10.1038/s41467-025-55853-7
- Jiawei Zhu, Yaru Meng, Wenli Gao, Shuo Yang, Wenjie Zhu, Xiangyang Ji, Xuanpei Zhai, Wan-Qiu Liu, Yuan Luo, Shengjie Ling, Jian Li, Yifan Liu. AI-driven high-throughput droplet screening of cell-free gene expression. Nature Communications 2025, 16
(1)
https://doi.org/10.1038/s41467-025-58139-0
- Michele Costantino, Eric J. Young, Abesh Banerjee, Cheryl A. Kerfeld, Giovanna Ghirlanda. Interfacing bacterial microcompartment shell proteins with genetically encoded condensates. Protein Science 2025, 34
(3)
https://doi.org/10.1002/pro.70061
- Neta Agmon, Liat Rahamim-Ben Navi, Yosi Shacham-Diamand, Yuval Dorfan, Ilana Kolodkin-Gal. From academia to real-life biotech: ‘It takes a village’. Trends in Biotechnology 2025, 7 https://doi.org/10.1016/j.tibtech.2025.02.014
- Marcelo Freire, Katarzyna Adamala, Alexandre Nepomuceno, Paul Christou, Elibio Rech. Engineering biology and the positive regulatory pathway in Brazil. Annals of Applied Biology 2025, 186
(2)
, 88-92. https://doi.org/10.1111/aab.12978
- Matthew E. Allen, James W. Hindley, Robert V. Law, Oscar Ces, Yuval Elani. Microfluidic Production of Spatially Structured Biomimetic Microgels as Compartmentalized Artificial Cells. Small Science 2025, 8 https://doi.org/10.1002/smsc.202400320
- Nuan Li, Xiaofeng Han, Xiaoli Mai, Peng Wang, Fangxu Wang, Linyuan Wu, Yuanyuan Xie, Bin Wang. Iron biomineralization by mediation of clMagR/clCry4 protein contribute to T2 contrast enhanced in MRI. Colloid and Interface Science Communications 2025, 64 , 100810. https://doi.org/10.1016/j.colcom.2024.100810
- Zhanar Abil, Simone Giaveri, Tobias J. Erb, Lynn J. Rothschild. Integrating metabolism and evolution towards the realization of synthetic life. Nature Reviews Bioengineering 2025, 3
(1)
, 9-10. https://doi.org/10.1038/s44222-024-00267-1
- Zhen‐Hong Luo, Gong‐Yu Shi, Nan‐Nan Deng. Easy, Rapid and High‐Throughput Production of Single and Multicompartment Liposomes by Vortex Emulsification. ChemSystemsChem 2024, 333 https://doi.org/10.1002/syst.202400085
- Souradeep Das, Gauri M. Patki, Vanthanaa Sridhar, Sahil Sunil Mulewar, Raya Roy, Udita Bandyopadhyay, Nishant Nitinidhi Kulshreshtha, Sudha Rajamani. Compartmentalization as a ubiquitous feature of life: from origins of life to biomimetics. The European Physical Journal Special Topics 2024, 233
(21-22)
, 3153-3172. https://doi.org/10.1140/epjs/s11734-024-01203-5
- Priyanshi Agnihotri, Divya Dheer, Anvi Sangwan, Vysakh C. Chandran, Nimisha A. Mavlankar, Gunjan Hooda, Debabrata Patra, Asish Pal. Design of multi-responsive and actuating microgels toward on-demand drug release. Nanoscale 2024, 16
(41)
, 19254-19265. https://doi.org/10.1039/D4NR02728K
- Sebastian Barthel, Luca Brenker, Christoph Diehl, Nitin Bohra, Simone Giaveri, Nicole Paczia, Tobias J Erb. In vitro
transcription-based biosensing of glycolate for prototyping of a complex enzyme cascade. Synthetic Biology 2024, 9
(1)
https://doi.org/10.1093/synbio/ysae013
- Surendra Yadav, Alexander J. P. Perkins, Sahan B. W. Liyanagedera, Anthony Bougas, Nadanai Laohakunakorn. ATP Regeneration from Pyruvate in the PURE System. 2024https://doi.org/10.1101/2024.09.06.611674
- Ion A. Ioannou, Carolina Monck, Francesca Ceroni, Nicholas J. Brooks, Marina K. Kuimova, Yuval Elani. Nucleated synthetic cells with genetically driven intercompartment communication. Proceedings of the National Academy of Sciences 2024, 121
(36)
https://doi.org/10.1073/pnas.2404790121
- Jiawei Zhu, Yaru Meng, Wenli Gao, Shuo Yang, Wenjie Zhu, Xiangyang Ji, Xuanpei Zhai, Wan-Qiu Liu, Yuan Luo, Shengjie Ling, Jian Li, Yifan Liu. AI-driven high-throughput droplet screening of cell-free gene expression. 2024https://doi.org/10.1101/2024.07.04.602084
- Sebastian Barthel, Luca Brenker, Christoph Diehl, Nitin Bohra, Simone Giaveri, Nicole Paczia, Tobias J Erb. In vitro
transcription-based biosensing of glycolate for prototyping of a complex enzyme cascade. 2024https://doi.org/10.1101/2024.04.26.591264
- Manisha Kapasiawala, Richard M. Murray. Metabolic perturbations to an
E. coli
-based cell-free system reveal a trade-off between transcription and translation. 2023https://doi.org/10.1101/2023.03.22.533877
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
Abstract
Figure 1
Figure 1. Our working definition of living systems, based on Tibor Gánti’s Chemoton model of life. Compartmentalization, replication, and metabolism together make up minimal criteria for a living system. From these other features emerge, including evolution, responsiveness to stimuli, and directed movement.
Figure 2
Figure 2. Four different molecular platforms for studying life. Outgoing from externally provided template DNA (1), a cell-free transcription/translation (Tx/Tl) system uses complex substrates and a chemical energy-donor (2) to synthesize RNA, proteins and potentially other biocatalytically (enzymatically) formed products (3). An encapsulated cell-free system is often a liposome enclosing a cell-free Tx/Tl system. A synthetic cell would function more like a natural cell in that it could be a self-sustaining system that is capable of replication.
Figure 3
Figure 3. Major approaches to building a synthetic cell: bottom-up (a), top-down (b), and middle-out (c) are distinguished as the basic concepts of currently ongoing research. Top-down approaches generally seek to find a “minimal genome” of an existing organism, while the bottom-up approach aspires to de novo create a cell “from scratch” or based on macromolecules. Middle-out approaches utilize modules of known function (e.g., organelles, extracts) to assemble a new cell or major elements/mechanisms thereof.
Figure 4
Figure 4. Milestones toward the de novo design and construction of a synthetic cell. Many of these steps can be performed in parallel with a mixing and matching approach to integration.
References
This article references 222 other publications.
- 1van Stevendaal, M. H. M. E.; Vasiukas, L.; Yewdall, N. A.; Mason, A. F.; van Hest, J. C. M. Engineering of Biocompatible Coacervate-Based Synthetic Cells. ACS Appl. Mater. Interfaces 2021, 13 (7), 7879– 7889, DOI: 10.1021/acsami.0c190521https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXjvFentr8%253D&md5=ee4e6e04fd446421302e5e3d773e6f8cEngineering of Biocompatible Coacervate-Based Synthetic Cellsvan Stevendaal, Marleen H. M. E.; Vasiukas, Laurynas; Yewdall, N. Amy; Mason, Alexander F.; van Hest, Jan C. M.ACS Applied Materials & Interfaces (2021), 13 (7), 7879-7889CODEN: AAMICK; ISSN:1944-8244. (American Chemical Society)Polymer-stabilized complex coacervate microdroplets have emerged as a robust platform for synthetic cell research. Their unique core-shell properties enable the sequestration of high concns. of biol. relevant macromols. and their subsequent release through the semipermeable membrane. These unique properties render the synthetic cell platform highly suitable for a range of biomedical applications, as long as its biocompatibility upon interaction with biol. cells is ensured. The purpose of this study is to investigate how the structure and formulation of these coacervate-based synthetic cells impact the viability of several different cell lines. Through careful examn. of the individual synthetic cell components, it became evident that the presence of free polycation and membrane-forming polymer had to be prevented to ensure cell viability. After closely examg. the structure-toxicity relationship, a set of conditions could be found whereby no detrimental effects were obsd., when the artificial cells were cocultured with RAW264.7 cells. This opens up a range of possibilities to use this modular system for biomedical applications and creates design rules for the next generation of coacervate-based, biomedically relevant particles.
- 2Tsokolov, S. A. Why Is the Definition of Life so Elusive? Epistemological Considerations. Astrobiology 2009, 9 (4), 401– 412, DOI: 10.1089/ast.2007.0201There is no corresponding record for this reference.
- 3Deplazes-Zemp, A. The Conception of Life in Synthetic Biology. Sci. Eng. Ethics 2012, 18 (4), 757– 774, DOI: 10.1007/s11948-011-9269-zThere is no corresponding record for this reference.
- 4Machery, E. Why I Stopped Worrying about the Definition of Life··· and Why You Should as Well. Synthese 2012, 185 (1), 145– 164, DOI: 10.1007/s11229-011-9880-1There is no corresponding record for this reference.
- 5Bird, A.; Tobin, E. Natural Kinds. In The Stanford Encyclopedia of Philosophy; Zalta, E. N., Nodelman, U., Eds.; Metaphysics Research Lab, Stanford University, 2023.There is no corresponding record for this reference.
- 6Cleland, C. E.; Chyba, C. F. Defining “Life.. Orig. Life Evol. Biosph. 2002, 32 (4), 387– 393, DOI: 10.1023/A:1020503324273There is no corresponding record for this reference.
- 7Ganti, T. The Principles of Life; OUP Oxford, 2003.There is no corresponding record for this reference.
- 8Vibhute, M. A.; Mutschler, H. A Primer on Building Life like Systems. ChemSystemsChem 2023, DOI: 10.1002/syst.202200033There is no corresponding record for this reference.
- 9Benner, S. A. Defining Life. Astrobiology 2010, 10 (10), 1021– 1030, DOI: 10.1089/ast.2010.05249https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC3M%252FkslGktg%253D%253D&md5=e392b1246f1c562063170009c5e89c0eDefining lifeBenner Steven AAstrobiology (2010), 10 (10), 1021-30 ISSN:.Any definition is intricately connected to a theory that gives it meaning. Accordingly, this article discusses various definitions of life held in the astrobiology community by considering their connected "theories of life." These include certain "list" definitions and a popular definition that holds that life is a "self-sustaining chemical system capable of Darwinian evolution." We then act as "anthropologists," studying what scientists do to determine which definition-theories of life they constructively hold as they design missions to seek non-terran life. We also look at how constructive beliefs about biosignatures change as observational data accumulate. And we consider how a definition centered on Darwinian evolution might itself be forced to change as supra-Darwinian species emerge, including in our descendents, and consider the chances of our encountering supra-Darwinian species in our exploration of the Cosmos. Last, we ask what chemical structures might support Darwinian evolution universally; these structures might be universal biosignatures.
- 10Cleland, C. E. The Quest for a Universal Theory of Life: Searching for Life As We Don’t Know It; Cambridge University Press, 2019.There is no corresponding record for this reference.
- 11Otto, S. An Approach to the De Novo Synthesis of Life. Acc. Chem. Res. 2022, 55 (2), 145– 155, DOI: 10.1021/acs.accounts.1c00534There is no corresponding record for this reference.
- 12Joyce, G. F. In Vitro Evolution of Nucleic Acids. Curr. Opin. Struct. Biol. 1994, 4, 331– 336, DOI: 10.1016/S0959-440X(94)90100-712https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2MXis1SntA%253D%253D&md5=d6ebdd652b2725a3e2e047f84cb5ed54In vitro evolution of nucleic acidsJoyce, Gerald F.Current Opinion in Structural Biology (1994), 4 (3), 331-6CODEN: COSBEF; ISSN:0959-440X.A review with 37 refs. An increasing no. of labs. are applying the principles of Darwinian evolution to large populations of RNA or DNA mols. to obtain compds. that bind a target ligand or catalyze a particular chem. reaction. These mols. may be useful as enzyme inhibitors or diagnostic agents.
- 13Agapakis, C. M. Designing Synthetic Biology. ACS Synth. Biol. 2014, 3 (3), 121– 128, DOI: 10.1021/sb400106813https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhs1KktbrM&md5=40ea81294cd017b2ff94389d5491d8b4Designing Synthetic BiologyAgapakis, Christina M.ACS Synthetic Biology (2014), 3 (3), 121-128CODEN: ASBCD6; ISSN:2161-5063. (American Chemical Society)A review. Synthetic biol. is frequently defined as the application of engineering design principles to biol. Such principles are intended to streamline the practice of biol. engineering, to shorten the time required to design, build, and test synthetic gene networks. This streamlining of iterative design cycles can facilitate the future construction of biol. systems for a range of applications in the prodn. of fuels, foods, materials, and medicines. The promise of these potential applications as well as the emphasis on design has prompted crit. reflection on synthetic biol. from design theorists and practicing designers from many fields, who can bring valuable perspectives to the discipline. While interdisciplinary connections between biologists and engineers have built synthetic biol. via the science and the technol. of biol., interdisciplinary collaboration with artists, designers, and social theorists can provide insight on the connections between technol. and society. Such collaborations can open up new avenues and new principles for research and design, as well as shed new light on the challenging context-dependence-both biol. and social-that face living technologies at many scales. This review is inspired by the session titled Design and Synthetic Biol.: Connecting People and Technol.at Synthetic Biol. 6.0 and covers a range of literature on design practice in synthetic biol. and beyond. Crit. engagement with how design is used to shape the discipline opens up new possibilities for how we might design the future of synthetic biol.
- 14Malaterre, C. Can Synthetic Biology Shed Light on the Origins of Life?. Biol. Theory 2009, 4 (4), 357– 367, DOI: 10.1162/BIOT_a_00002There is no corresponding record for this reference.
- 15Mukherji, S.; van Oudenaarden, A. Synthetic Biology: Understanding Biological Design from Synthetic Circuits. Nat. Rev. Genet. 2009, 10 (12), 859– 871, DOI: 10.1038/nrg269715https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhsVaksL%252FO&md5=27ea5f286f69609af97759d5e3effb1bSynthetic biology: understanding biological design from synthetic circuitsMukherji, Shankar; van Oudenaarden, AlexanderNature Reviews Genetics (2009), 10 (12), 859-871CODEN: NRGAAM; ISSN:1471-0056. (Nature Publishing Group)The article highlights how the process of engineering biol. systems has contributed to our understanding of how endogenous systems are put together and function - from a quant. description of gene expression and signal transduction to controlling spatial organization and cell-cell interactions. An important aim of synthetic biol. is to uncover the design principles of natural biol. systems through the rational design of gene and protein circuits. Here, we highlight how the process of engineering biol. systems - from synthetic promoters to the control of cell-cell interactions - has contributed to our understanding of how endogenous systems are put together and function. Synthetic biol. devices allow us to grasp intuitively the ranges of behavior generated by simple biol. circuits, such as linear cascades and interlocking feedback loops, as well as to exert control over natural processes, such as gene expression and population dynamics.
- 16Rothschild, L. J. The Role of Emergence in Biology; Oxford University Press, 2008.There is no corresponding record for this reference.
- 17Adamala, K.; Szostak, J. W. Nonenzymatic Template-Directed RNA Synthesis inside Model Protocells. Science 2013, 342 (6162), 1098– 1100, DOI: 10.1126/science.124188817https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhvVartr3K&md5=47b93286cfc8db8306f03be866470a70Nonenzymatic Template-Directed RNA Synthesis Inside Model ProtocellsAdamala, Katarzyna; Szostak, Jack W.Science (Washington, DC, United States) (2013), 342 (6162), 1098-1100CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Efforts to recreate a prebiotically plausible protocell, in which RNA replication occurs within a fatty acid vesicle, have been stalled by the destabilizing effect of Mg2+ on fatty acid membranes. Here we report that the presence of citrate protects fatty acid membranes from the disruptive effects of high Mg2+ ion concns. while allowing RNA copying to proceed, while also protecting single-stranded RNA from Mg2+-catalyzed degrdn. This combination of properties has allowed us to demonstrate the chem. copying of RNA templates inside fatty acid vesicles, which in turn allows for an increase in copying efficiency by bathing the vesicles in a continuously refreshed soln. of activated nucleotides.
- 18Deamer, D.; Dworkin, J. P.; Sandford, S. A.; Bernstein, M. P.; Allamandola, L. J. The First Cell Membranes. Astrobiology 2002, 2 (4), 371– 381, DOI: 10.1089/15311070276247048218https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXhtVamur4%253D&md5=263bd36bf3d352bb062f2d04c0406f6aThe first cell membranesDeamer, David; Dworkin, Jason P.; Sandford, Scott A.; Bernstein, Max P.; Allamandola, Louis J.Astrobiology (2002), 2 (4), 371-381CODEN: ASTRC4; ISSN:1531-1074. (Mary Ann Liebert, Inc.)A review. Org. compds. are synthesized in the interstellar medium and can be delivered to planetary surfaces such as the early Earth, where they mix with endogenous species. Some of these compds. are amphiphilic, having polar and nonpolar groups on the same mol. Amphiphilic compds. spontaneously self-assemble into more complex structures such as bimol. layers, which in turn form closed membranous vesicles. The 1st forms of cellular life required self-assembled membranes that were likely to have been produced from amphiphilic compds. on the prebiotic Earth. Lab. simulations show that such vesicles readily encapsulate functional macromols., including nucleic acids and polymerases. The goal of future investigations will be to fabricate artificial cells as models of the origin of life.
- 19Deamer, D. The Role of Lipid Membranes in Life’s Origin. Life 2017, 7 (1), 5, DOI: 10.3390/life7010005There is no corresponding record for this reference.
- 20Discher, B. M.; Bermudez, H.; Hammer, D. A.; Discher, D. E.; Won, Y.-Y.; Bates, F. S. Cross-Linked Polymersome Membranes: Vesicles with Broadly Adjustable Properties. J. Phys. Chem. B 2002, 106 (11), 2848– 2854, DOI: 10.1021/jp011958z20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XhsVGjt7s%253D&md5=66052831fe2660d1457252d266f0d7ffCross-linked Polymersome Membranes: Vesicles with Broadly Adjustable PropertiesDischer, Bohdana M.; Bermudez, Harry; Hammer, Daniel A.; Discher, Dennis E.; Won, You-Yeon; Bates, Frank S.Journal of Physical Chemistry B (2002), 106 (11), 2848-2854CODEN: JPCBFK; ISSN:1089-5647. (American Chemical Society)Massively cross-linked and property-tunable membranes have been fabricated by free radical polymn. of self-assembled, block copolymer vesicles - polymersomes. Similar efforts with cross-linkable lipids would appear frustrated in the past due to at least two factors: limited reactivity and membrane fragility under local stresses of nano-confined crosslinking. We describe here a diblock copolymer of poly(ethylene oxide)-polybutadiene that has a hydrophilic wt. fraction like that of lipids and forms robust fluid phase membranes in water. The polymersomes sustain free radical polymn. of the hydrophobic butadiene, thereby generating a semipermeable nano-shell. Cross-linked giant vesicles prove stable in chloroform and can also be dehydrated and re-hydrated without rendering the ∼9 nm thick membrane core; the results imply defect-free membranes many microns-squared in area. Surface elastic moduli as well as sustainable wall stresses up to 103 Atm, orders of magnitude greater than any natural lipid membrane, appear consistent with strong tethering between close-packed neighbors. The enormous stability of the giant vesicles can be tuned down for application: blending in the hydrogenated analog poly(ethylene oxide)-polyethylethylene modulates the effective elastic consts. as well as the rupture strength by orders of magnitude. The results appear consistent with rigidity percolation through a finite-layer stack of two-dimensional lattices. Moreover, below the percolation limit, a regime of hyper-instability emerges, reflecting perhaps nanoscale demixing and suggestive of the limitations encountered with low reactivity lipids. The results provide general insights into covalent crosslinking within self-assembled nanostructures.
- 21Kamat, N. P.; Katz, J. S.; Hammer, D. A. Engineering Polymersome Protocells. J. Phys. Chem. Lett. 2011, 2 (13), 1612– 1623, DOI: 10.1021/jz200640x21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXnsFWjtLs%253D&md5=3c4972f113a5618037a2c216667ba743Engineering Polymersome ProtocellsKamat, Neha P.; Katz, Joshua S.; Hammer, Daniel A.Journal of Physical Chemistry Letters (2011), 2 (13), 1612-1623CODEN: JPCLCD; ISSN:1948-7185. (American Chemical Society)A review. The field of biomimicry is embracing the construction of complex assemblies that imitate both biol. structure and function. Advancements in the design of these mimetics have generated a growing vision for creating an artificial cell or protocell. Polymersomes are vesicles that can be made from synthetic, biol., or hybrid polymers and can be used as a model template to build cell-like structures. In this perspective, we discuss various areas where polymersomes have been used to mimic cell functions as well as areas in which the synthetic flexibility of polymersomes would make them ideal candidates for a biomembrane mimetic. Designing a polymersome that comprehensively displays the behaviors discussed herein has the potential to lead to the development of an autonomous, responsive particle that resembles the intelligence of a biol. cell.
- 22Vickers, C. E. The Minimal Genome Comes of Age. Nat. Biotechnol. 2016, 34 (6), 623– 624, DOI: 10.1038/nbt.3593There is no corresponding record for this reference.
- 23Xu, X.; Meier, F.; Blount, B. A.; Pretorius, I. S.; Ellis, T.; Paulsen, I. T.; Williams, T. C. Trimming the Genomic Fat: Minimising and Re-Functionalising Genomes Using Synthetic Biology. Nat. Commun. 2023, 14 (1), 1984, DOI: 10.1038/s41467-023-37748-723https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3sXnslWksb0%253D&md5=44b6c482b42a43dbcf6a77510da09105Trimming the genomic fat: minimising and re-functionalising genomes using synthetic biologyXu, Xin; Meier, Felix; Blount, Benjamin A.; Pretorius, Isak S.; Ellis, Tom; Paulsen, Ian T.; Williams, Thomas C.Nature Communications (2023), 14 (1), 1984CODEN: NCAOBW; ISSN:2041-1723. (Nature Portfolio)A review. Naturally evolved organisms typically have large genomes that enable their survival and growth under various conditions. However, the complexity of genomes often precludes our complete understanding of them, and limits the success of biotechnol. designs. In contrast, minimal genomes have reduced complexity and therefore improved engineerability, increased biosynthetic capacity through the removal of unnecessary genetic elements, and less recalcitrance to complete characterization. Here, we review the past and current genome minimisation and re-functionalisation efforts, with an emphasis on the latest advances facilitated by synthetic genomics, and provide a crit. appraisal of their potential for industrial applications.
- 24Hutchison, C. A.; Peterson, S. N.; Gill, S. R.; Cline, R. T.; White, O.; Fraser, C. M.; Smith, H. O.; Venter, J. C. Global Transposon Mutagenesis and a Minimal Mycoplasma Genome. Science 1999, 286 (5447), 2165– 2169, DOI: 10.1126/science.286.5447.216524https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1MXotFGru70%253D&md5=863f5f2e7a6dcefb45cd5765e0f892e8Global transposon mutagenesis and a minimal mycoplasma genomeHutchison, Clyde A., III; Peterson, Scott N.; Gill, Steven R.; Cline, Robin T.; White, Owen; Fraser, Claire M.; Smith, Hamilton O.; Venter, J. CraigScience (Washington, D. C.) (1999), 286 (5447), 2165-2169CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Mycoplasma genitalium with 517 genes has the smallest gene complement of any independently replicating cell so far identified. Global transposon mutagenesis was used to identify nonessential genes in an effort to learn whether the naturally occurring gene complement is a true minimal genome under lab. growth conditions. The positions of 2209 transposon insertions in the completely sequenced genomes of M. genitalium and its close relative M. pneumoniae were detd. by sequencing across the junction of the transposon and the genomic DNA. These junctions defined 1354 distinct sites of insertion that were not lethal. The anal. suggests that 265 to 350 of the 480 protein-coding genes of M. genitalium are essential under lab. growth conditions, including about 100 genes of unknown function.
- 25Glass, J. I.; Assad-Garcia, N.; Alperovich, N.; Yooseph, S.; Lewis, M. R.; Maruf, M.; Hutchison, C. A., 3rd; Smith, H. O.; Venter, J. C. Essential Genes of a Minimal Bacterium. Proc. Natl. Acad. Sci. U. S. A. 2006, 103 (2), 425– 430, DOI: 10.1073/pnas.051001310325https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XpsVSltA%253D%253D&md5=a4eabb14b59f1b81454494b19cf287a4Essential genes of a minimal bacteriumGlass, John I.; Assad-Garcia, Nacyra; Alperovich, Nina; Yooseph, Shibu; Lewis, Matthew R.; Maruf, Mahir; Hutchison, Clyde A., III; Smith, Hamilton O.; Venter, J. CraigProceedings of the National Academy of Sciences of the United States of America (2006), 103 (2), 425-430CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Mycoplasma genitalium has the smallest genome of any organism that can be grown in pure culture. It has a minimal metab. and little genomic redundancy. Consequently, its genome is expected to be a close approxn. to the minimal set of genes needed to sustain bacterial life. Using global transposon mutagenesis, gene disruption mutants were isolated and characterized for 100 different nonessential protein-coding genes. None of the 43 RNA-coding genes were disrupted. Herein, 382 of the 482 M. genitalium protein-coding genes were identified as essential, plus 5 sets of disrupted genes that encode proteins with potentially redundant essential functions, such as phosphate transport. Genes encoding proteins of unknown function constitute 28% of the essential protein-coding genes set. Disruption of some genes accelerated M. genitalium growth. The genome of M. genitalium G37 (ATCC 33530) was resequenced and found to differ from the previously sequenced version at 34 sites; the new sequences replaces the original M. genitalium genome sequence in GenBank/EMBL/DDBJ under accession no. L43967.
- 26Hutchison, C. A., 3rd; Chuang, R.-Y.; Noskov, V. N.; Assad-Garcia, N.; Deerinck, T. J.; Ellisman, M. H.; Gill, J.; Kannan, K.; Karas, B. J.; Ma, L.; Pelletier, J. F.; Qi, Z.-Q.; Richter, R. A.; Strychalski, E. A.; Sun, L.; Suzuki, Y.; Tsvetanova, B.; Wise, K. S.; Smith, H. O.; Glass, J. I.; Merryman, C.; Gibson, D. G.; Venter, J. C. Design and Synthesis of a Minimal Bacterial Genome. Science 2016, 351 (6280), aad6253, DOI: 10.1126/science.aad6253There is no corresponding record for this reference.
- 27Hashimoto, M.; Ichimura, T.; Mizoguchi, H.; Tanaka, K.; Fujimitsu, K.; Keyamura, K.; Ote, T.; Yamakawa, T.; Yamazaki, Y.; Mori, H.; Katayama, T.; Kato, J.-I. Cell Size and Nucleoid Organization of Engineered Escherichia Coli Cells with a Reduced Genome. Mol. Microbiol. 2005, 55 (1), 137– 149, DOI: 10.1111/j.1365-2958.2004.04386.x27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXotlSrtw%253D%253D&md5=484e3e2ccc942985f12b165903bf23f2Cell size and nucleoid organization of engineered Escherichia coli cells with a reduced genomeHashimoto, Masayuki; Ichimura, Toshiharu; Mizoguchi, Hiroshi; Tanaka, Kimie; Fujimitsu, Kazuyuki; Keyamura, Kenji; Ote, Tomotake; Yamakawa, Takehiro; Yamazaki, Yukiko; Mori, Hideo; Katayama, Tsutomu; Kato, Jun-ichiMolecular Microbiology (2005), 55 (1), 137-149CODEN: MOMIEE; ISSN:0950-382X. (Blackwell Publishing Ltd.)The minimization of a genome is necessary to identify exptl. the minimal gene set that contains only those genes that are essential and sufficient to sustain a functioning cell. Recent developments in genetic techniques have made it possible to generate bacteria with a markedly reduced genome. We developed a simple system for formation of markerless chromosomal deletions, and constructed and characterized a series of large-scale chromosomal deletion mutants of Escherichia coli that lack between 2.4 and 29.7% of the parental chromosome. Combining deletion mutations changes cell length and width, and the mutant cells with larger deletions were even longer and wider than the parental cells. The nucleoid organization of the mutants is also changed: the nucleoids occur as multiple small nucleoids and are localized peripherally near the envelope. Inhibition of translation causes them to condense into one or two packed nucleoids, suggesting that the coupling of transcription and translation of membrane proteins peripherally localizes chromosomes. Because these phenotypes are similar to those of spherical cells, those may be a consequence of the morphol. change. Based on the nucleoid localization obsd. with these mutants, we discuss the cellular nucleoid dynamics.
- 28Pósfai, G.; Plunkett, G., 3rd; Fehér, T.; Frisch, D.; Keil, G. M.; Umenhoffer, K.; Kolisnychenko, V.; Stahl, B.; Sharma, S. S.; de Arruda, M.; Burland, V.; Harcum, S. W.; Blattner, F. R. Emergent Properties of Reduced-Genome Escherichia Coli. Science 2006, 312 (5776), 1044– 1046, DOI: 10.1126/science.1126439There is no corresponding record for this reference.
- 29Lartigue, C.; Glass, J. I.; Alperovich, N.; Pieper, R.; Parmar, P. P.; Hutchison, C. A., 3rd; Smith, H. O.; Venter, J. C. Genome Transplantation in Bacteria: Changing One Species to Another. Science 2007, 317 (5838), 632– 638, DOI: 10.1126/science.114462229https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2sXptVCgu7Y%253D&md5=3acd99db81d8698752ffad0636948da2Genome transplantation in bacteria: changing one species to anotherLartigue, Carole; Glass, John I.; Alperovich, Nina; Pieper, Rembert; Parmar, Prashanth P.; Hutchison, Clyde A., III; Smith, Hamilton O.; Venter, J. CraigScience (Washington, DC, United States) (2007), 317 (5838), 632-638CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)As a step toward propagation of synthetic genomes, the authors completely replaced the genome of a bacterial cell with one from another species by transplanting a whole genome as naked DNA. Intact genomic DNA from Mycoplasma mycoides large colony (LC), virtually free of protein, was transplanted into Mycoplasma capricolum cells by polyethylene glycol-mediated transformation. Cells selected for tetracycline resistance, carried by the M. mycoides LC chromosome, contain the complete donor genome and are free of detectable recipient genomic sequences. These cells that result from genome transplantation are phenotypically identical to the M. mycoides LC donor strain as judged by several criteria.
- 30Gibson, D. G.; Benders, G. A.; Andrews-Pfannkoch, C.; Denisova, E. A.; Baden-Tillson, H.; Zaveri, J.; Stockwell, T. B.; Brownley, A.; Thomas, D. W.; Algire, M. A.; Merryman, C.; Young, L.; Noskov, V. N.; Glass, J. I.; Venter, J. C.; Hutchison, C. A., 3rd; Smith, H. O. Complete Chemical Synthesis, Assembly, and Cloning of a Mycoplasma Genitalium Genome. Science 2008, 319 (5867), 1215– 1220, DOI: 10.1126/science.115172130https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXisVSksbs%253D&md5=c17e288749853e61fa00ffa9048e27ceComplete Chemical Synthesis, Assembly, and Cloning of a Mycoplasma genitalium GenomeGibson, Daniel G.; Benders, Gwynedd A.; Andrews-Pfannkoch, Cynthia; Denisova, Evgeniya A.; Baden-Tillson, Holly; Zaveri, Jayshree; Stockwell, Timothy B.; Brownley, Anushka; Thomas, David W.; Algire, Mikkel A.; Merryman, Chuck; Young, Lei; Noskov, Vladimir N.; Glass, John I.; Venter, J. Craig; Hutchison, Clyde A., III; Smith, Hamilton O.Science (Washington, DC, United States) (2008), 319 (5867), 1215-1220CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)We have synthesized a 582,970-base pair Mycoplasma genitalium genome. This synthetic genome, named M. genitalium JCVI-1.0, contains all the genes of wild-type M. genitalium G37 except MG408, which was disrupted by an antibiotic marker to block pathogenicity and to allow for selection. To identify the genome as synthetic, we inserted "watermarks" at intergenic sites known to tolerate transposon insertions. Overlapping "cassettes" of 5 to 7 kilobases (kb), assembled from chem. synthesized oligonucleotides, were joined by in vitro recombination to produce intermediate assemblies of approx. 24 kb, 72 kb ("1/8 genome"), and 144 kb ("1/4 genome"), which were all cloned as bacterial artificial chromosomes in Escherichia coli. Most of these intermediate clones were sequenced, and clones of all four 1/4 genomes with the correct sequence were identified. The complete synthetic genome was assembled by transformation-assocd. recombination cloning in the yeast Saccharomyces cerevisiae, then isolated and sequenced. A clone with the correct sequence was identified. The methods described here will be generally useful for constructing large DNA mols. from chem. synthesized pieces and also from combinations of natural and synthetic DNA segments.
- 31Lartigue, C.; Vashee, S.; Algire, M. A.; Chuang, R.-Y.; Benders, G. A.; Ma, L.; Noskov, V. N.; Denisova, E. A.; Gibson, D. G.; Assad-Garcia, N.; Alperovich, N.; Thomas, D. W.; Merryman, C.; Hutchison, C. A., 3rd; Smith, H. O.; Venter, J. C.; Glass, J. I. Creating Bacterial Strains from Genomes That Have Been Cloned and Engineered in Yeast. Science 2009, 325 (5948), 1693– 1696, DOI: 10.1126/science.117375931https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtFGqt7%252FK&md5=bfc12b44f02c66cdc491d57eee6e46fdCreating Bacterial Strains from Genomes That Have Been Cloned and Engineered in YeastLartigue, Carole; Vashee, Sanjay; Algire, Mikkel A.; Chuang, Ray-Yuan; Benders, Gwynedd A.; Ma, Li; Noskov, Vladimir N.; Denisova, Evgeniya A.; Gibson, Daniel G.; Assad-Garcia, Nacyra; Alperovich, Nina; Thomas, David W.; Merryman, Chuck; Hutchison, Clyde A., III; Smith, Hamilton O.; Venter, J. Craig; Glass, John I.Science (Washington, DC, United States) (2009), 325 (5948), 1693-1696CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)We recently reported the chem. synthesis, assembly, and cloning of a bacterial genome in yeast. To produce a synthetic cell, the genome must be transferred from yeast to a receptive cytoplasm. Here we describe methods to accomplish this. We cloned a Mycoplasma mycoides genome as a yeast centromeric plasmid and then transplanted it into Mycoplasma capricolum to produce a viable M. mycoides cell. While in yeast, the genome was altered by using yeast genetic systems and then transplanted to produce a new strain of M. mycoides. These methods allow the construction of strains that could not be produced with genetic tools available for this bacterium. The complete, annotated sequence of the transplanted M. mycoides capri genome is deposited in GenBank/EMBL/DDBJ with accession no. CP001668.
- 32Gibson, D. G.; Glass, J. I.; Lartigue, C.; Noskov, V. N.; Chuang, R.-Y.; Algire, M. A.; Benders, G. A.; Montague, M. G.; Ma, L.; Moodie, M. M.; Merryman, C.; Vashee, S.; Krishnakumar, R.; Assad-Garcia, N.; Andrews-Pfannkoch, C.; Denisova, E. A.; Young, L.; Qi, Z.-Q.; Segall-Shapiro, T. H.; Calvey, C. H.; Parmar, P. P.; Hutchison, C. A., 3rd; Smith, H. O.; Venter, J. C. Creation of a Bacterial Cell Controlled by a Chemically Synthesized Genome. Science 2010, 329 (5987), 52– 56, DOI: 10.1126/science.119071932https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXotVeqsLg%253D&md5=f978c98f3e5c3c9b14bd4c7d8a1eecc7Creation of a bacterial cell controlled by a chemically synthesized genomeGibson, Daniel G.; Glass, John I.; Lartigue, Carole; Noskov, Vladimir N.; Chuang, Ray-Yuan; Algire, Mikkel A.; Benders, Gwynedd A.; Montague, Michael G.; Ma, Li; Moodie, Monzia M.; Merryman, Chuck; Vashee, Sanjay; Krishnakumar, Radha; Assad-Garcia, Nacyra; Andrews-Pfannkoch, Cynthia; Denisova, Evgeniya A.; Young, Lei; Qi, Zhi-Qing; Segall-Shapiro, Thomas H.; Calvey, Christopher H.; Parmar, Prashanth P.; Hutchison, Clyde A., III; Smith, Hamilton O.; Venter, J. CraigScience (Washington, DC, United States) (2010), 329 (5987), 52-56CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)We report the design, synthesis, and assembly of the 1.08-mega-base pair Mycoplasma mycoides JCVI-syn1.0 genome starting from digitized genome sequence information and its transplantation into a M. capricolum recipient cell to create new M. mycoides cells that are controlled only by the synthetic chromosome. The only DNA in the cells is the designed synthetic DNA sequence, including "watermark" sequences and other designed gene deletions and polymorphisms, and mutations acquired during the building process. The new cells have expected phenotypic properties and are capable of continuous self-replication. The complete, annotated synthetic genome sequence is deposited in GenBank/EMBL/DDBJ with accession no. CP002027.
- 33Stano, P.; Luisi, P. L. Semi-Synthetic Minimal Cells: Origin and Recent Developments. Curr. Opin. Biotechnol. 2013, 24 (4), 633– 638, DOI: 10.1016/j.copbio.2013.01.00233https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhslCnu7w%253D&md5=0bb71b3497381842f77cfd6bc763c81cSemi-synthetic minimal cells: origin and recent developmentsStano, Pasquale; Luisi, Pier LuigiCurrent Opinion in Biotechnology (2013), 24 (4), 633-638CODEN: CUOBE3; ISSN:0958-1669. (Elsevier B.V.)A review. The notion of minimal cells refers to cellular structures that contain the minimal and sufficient complexity to still be defined as living, or at least capable to display the most important features of biol. cells. Here the authors briefly describe the lab. construction of minimal cells, a project within the broader field of synthetic biol. In particular the authors discuss the advancements in the prepn. of semi-synthetic cells based on the encapsulation of biochems. inside liposomes, illustrating from the one hand the origin of this research and the most recent developments; and from the other the difficulties and limits of the approach. The role of physicochem. understandings is greatly emphasized.
- 34Olivi, L.; Berger, M.; Creyghton, R. N. P.; De Franceschi, N.; Dekker, C.; Mulder, B. M.; Claassens, N. J.; Ten Wolde, P. R.; van der Oost, J. Towards a Synthetic Cell Cycle. Nat. Commun. 2021, 12 (1), 4531, DOI: 10.1038/s41467-021-24772-834https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhsleqs7jE&md5=dcc559e4d649037affd64b1951698d1fTowards a synthetic cell cycleOlivi, Lorenzo; Berger, Mareike; Creyghton, Ramon N. P.; De Franceschi, Nicola; Dekker, Cees; Mulder, Bela M.; Claassens, Nico J.; ten Wolde, Pieter Rein; van der Oost, JohnNature Communications (2021), 12 (1), 4531CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)Abstr.: Recent developments in synthetic biol. may bring the bottom-up generation of a synthetic cell within reach. A key feature of a living synthetic cell is a functional cell cycle, in which DNA replication and segregation as well as cell growth and division are well integrated. Here, we describe different approaches to recreate these processes in a synthetic cell, based on natural systems and/or synthetic alternatives. Although some individual machineries have recently been established, their integration and control in a synthetic cell cycle remain to be addressed. In this Perspective, we discuss potential paths towards an integrated synthetic cell cycle.
- 35Adamala, K. P.; Martin-Alarcon, D. A.; Guthrie-Honea, K. R.; Boyden, E. S. Engineering Genetic Circuit Interactions within and between Synthetic Minimal Cells. Nat. Chem. 2017, 9 (5), 431– 439, DOI: 10.1038/nchem.264435https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhvVGiur3E&md5=b362d8195003a28512978349366fa02fEngineering genetic circuit interactions within and between synthetic minimal cellsAdamala, Katarzyna P.; Martin-Alarcon, Daniel A.; Guthrie-Honea, Katriona R.; Boyden, Edward S.Nature Chemistry (2017), 9 (5), 431-439CODEN: NCAHBB; ISSN:1755-4330. (Nature Publishing Group)Genetic circuits and reaction cascades are of great importance for synthetic biol., biochem. and bioengineering. An open question is how to maximize the modularity of their design to enable the integration of different reaction networks and to optimize their scalability and flexibility. One option is encapsulation within liposomes, which enables chem. reactions to proceed in well-isolated environments. Here we adapt liposome encapsulation to enable the modular, controlled compartmentalization of genetic circuits and cascades. We demonstrate that it is possible to engineer genetic circuit-contg. synthetic minimal cells (synells) to contain multiple-part genetic cascades, and that these cascades can be controlled by external signals as well as inter-liposomal communication without crosstalk. We also show that liposomes that contain different cascades can be fused in a controlled way so that the products of incompatible reactions can be brought together. Synells thus enable a more modular creation of synthetic biol. cascades, an essential step towards their ultimate programmability.
- 36Fenz, S. F.; Sachse, R.; Schmidt, T.; Kubick, S. Cell-Free Synthesis of Membrane Proteins: Tailored Cell Models out of Microsomes. Biochim. Biophys. Acta 2014, 1838 (5), 1382– 1388, DOI: 10.1016/j.bbamem.2013.12.009There is no corresponding record for this reference.
- 37Kroll, A. V.; Fang, R. H.; Zhang, L. Biointerfacing and Applications of Cell Membrane-Coated Nanoparticles. Bioconjugate Chem. 2017, 28 (1), 23– 32, DOI: 10.1021/acs.bioconjchem.6b0056937https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhslKgtrbK&md5=aae744045651438cbf397dfce59bb3d7Biointerfacing and Applications of Cell Membrane-Coated NanoparticlesKroll, Ashley V.; Fang, Ronnie H.; Zhang, LiangfangBioconjugate Chemistry (2017), 28 (1), 23-32CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)The cell membrane-coated nanoparticle is a biomimetic platform consisting of a nanoparticulate core coated with membrane derived from a cell, such as a red blood cell, platelet, or cancer cell. The cell membrane "disguise" allows the particles to be perceived by the body as the source cell by interacting with its surroundings using the translocated surface membrane components. The newly bestowed characteristics of the membrane-coated nanoparticle can be utilized for biol. interfacing in the body, providing natural solns. to many biomedical issues. This Review will cover the interactions of these cell membrane-coated nanoparticles and their applications within three biomedical areas of interest, including (i) drug delivery, (ii) detoxification, and (iii) immune modulation.
- 38Gurramkonda, C.; Rao, A.; Borhani, S.; Pilli, M.; Deldari, S.; Ge, X.; Pezeshk, N.; Han, T.-C.; Tolosa, M.; Kostov, Y.; Tolosa, L.; Wood, D. W.; Vattem, K.; Frey, D. D.; Rao, G. Improving the Recombinant Human Erythropoietin Glycosylation Using Microsome Supplementation in CHO Cell-Free System. Biotechnol. Bioeng. 2018, 115 (5), 1253– 1264, DOI: 10.1002/bit.26554There is no corresponding record for this reference.
- 39Su’etsugu, M.; Takada, H.; Katayama, T.; Tsujimoto, H. Exponential Propagation of Large Circular DNA by Reconstitution of a Chromosome-Replication Cycle. Nucleic Acids Res. 2017, 45 (20), 11525– 11534, DOI: 10.1093/nar/gkx82239https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtVyisbvM&md5=97d99e9a4ac7d2baaa3820e6a1d474c8Exponential propagation of large circular DNA by reconstitution of a chromosome-replication cycleSu'etsugu, Masayuki; Takada, Hiraku; Katayama, Tsutomu; Tsujimoto, HirokoNucleic Acids Research (2017), 45 (20), 11525-11534CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)Propagation of genetic information is a fundamental property of living organisms. Escherichia coli has a 4.6 Mb circular chromosome with a replication origin, oriC. While the oriC replication has been reconstituted in vitro more than 30 years ago, continuous repetition of the replication cycle has not yet been achieved. Here, we reconstituted the entire replication cycle with 14 purified enzymes (25 polypeptides) that catalyze initiation at oriC, bidirectional fork progression, Okazaki-fragment maturation and decatenation of the replicated circular products. Because decatenation provides covalently closed supercoiled monomers that are competent for the next round of replication initiation, the replication cycle repeats autonomously and continuously in an isothermal condition. This replication-cycle reaction (RCR) propagates ∼10 kb circular DNA exponentially as intact covalently closed mols., even from a single DNA mol., with a doubling time of ∼8 min and extremely high fidelity. Very large DNA up to 0.2 Mb is successfully propagated within 3 h. We further demonstrate a cell-free cloning in which RCR selectively propagates circular mols. constructed by a multi-fragment assembly reaction. Our results define the min. element necessary for the repetition of the chromosome-replication cycle, and also provide a powerful in vitro tool to generate large circular DNA mols. without relying on conventional biol. cloning.
- 40Shimizu, Y.; Inoue, A.; Tomari, Y.; Suzuki, T.; Yokogawa, T.; Nishikawa, K.; Ueda, T. Cell-Free Translation Reconstituted with Purified Components. Nat. Biotechnol. 2001, 19 (8), 751– 755, DOI: 10.1038/9080240https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3MXlslekt7g%253D&md5=8560f1b7319ea88b4784a4f02bafcbafCell-free translation reconstituted with purified componentsShimizu, Yoshihiro; Inoue, Akio; Tomari, Yukihide; Suzuki, Tsutomu; Yokogawa, Takashi; Nishikawa, Kazuya; Ueda, TakuyaNature Biotechnology (2001), 19 (8), 751-755CODEN: NABIF9; ISSN:1087-0156. (Nature America Inc.)We have developed a protein-synthesizing system reconstituted from recombinant tagged protein factors purified to homogeneity. The system was able to produce protein at a rate of about 160 μg/mL/h in a batch mode without the need for any supplementary app. The protein products were easily purified within 1 h using affinity chromatog. to remove the tagged protein factors. Moreover, omission of a release factor allowed efficient incorporation of an unnatural amino acid using suppressor tRNA.
- 41Drienovská, I.; Roelfes, G. Expanding the Enzyme Universe with Genetically Encoded Unnatural Amino Acids. Nature Catalysis 2020, 3 (3), 193– 202, DOI: 10.1038/s41929-019-0410-8There is no corresponding record for this reference.
- 42Medina, E.; Yik, E. J.; Herdewijn, P.; Chaput, J. C. Functional Comparison of Laboratory-Evolved XNA Polymerases for Synthetic Biology. ACS Synth. Biol. 2021, 10 (6), 1429– 1437, DOI: 10.1021/acssynbio.1c00048There is no corresponding record for this reference.
- 43Theobald, D. L. A Formal Test of the Theory of Universal Common Ancestry. Nature 2010, 465 (7295), 219– 222, DOI: 10.1038/nature09014There is no corresponding record for this reference.
- 44Yang, Z.; Chen, F.; Alvarado, J. B.; Benner, S. A. Amplification, Mutation, and Sequencing of a Six-Letter Synthetic Genetic System. J. Am. Chem. Soc. 2011, 133 (38), 15105– 15112, DOI: 10.1021/ja204910n44https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtFaks7zL&md5=cc4561e9527ca359c82494deb9e0be2dAmplification, mutation, and sequencing of a six-letter synthetic genetic systemYang, Zun-Yi; Chen, Fei; Alvarado, J. Brian; Benner, Steven A.Journal of the American Chemical Society (2011), 133 (38), 15105-15112CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)The next goals in the development of a synthetic biol. that uses artificial genetic systems will require chem.-biol. combinations that allow the amplification of DNA contg. any no. of sequential and nonsequential nonstandard nucleotides. This amplification must ensure that the nonstandard nucleotides are not unidirectionally lost during PCR amplification (unidirectional loss would cause the artificial system to revert to an all-natural genetic system). Further, technol. is needed to sequence artificial genetic DNA mols. The work reported here meets all three of these goals for a six-letter artificially expanded genetic information system (AEGIS) that comprises four std. nucleotides (G, A, C, and T) and two addnl. nonstandard nucleotides (Z and P). We report polymerases and PCR conditions that amplify a wide range of GACTZP DNA sequences having multiple consecutive unnatural synthetic genetic components with low (0.2% per theor. cycle) levels of mutation. We demonstrate that residual mutation processes both introduce and remove unnatural nucleotides, allowing the artificial genetic system to evolve as such, rather than revert to a wholly natural system. We then show that mechanisms for these residual mutation processes can be exploited in a strategy to sequence "six-letter" GACTZP DNA. These are all not yet reported for any other synthetic genetic system.
- 45Malyshev, D. A.; Dhami, K.; Lavergne, T.; Chen, T.; Dai, N.; Foster, J. M.; Corrêa, I. R., Jr; Romesberg, F. E. A Semi-Synthetic Organism with an Expanded Genetic Alphabet. Nature 2014, 509 (7500), 385– 388, DOI: 10.1038/nature1331445https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXotVyqtb8%253D&md5=97b4b184cda52cc809b1705e5e88ad8eA semi-synthetic organism with an expanded genetic alphabetMalyshev, Denis A.; Dhami, Kirandeep; Lavergne, Thomas; Chen, Tingjian; Dai, Nan; Foster, Jeremy M.; Correa, Ivan R.; Romesberg, Floyd E.Nature (London, United Kingdom) (2014), 509 (7500), 385-388CODEN: NATUAS; ISSN:0028-0836. (Nature Publishing Group)Organisms are defined by the information encoded in their genomes, and since the origin of life this information has been encoded using a two-base-pair genetic alphabet (A-T and G-C). In vitro, the alphabet has been expanded to include several unnatural base pairs (UBPs). We have developed a class of UBPs formed between nucleotides bearing hydrophobic nucleobases, exemplified by the pair formed between d5SICS and dNaM (d5SICS-dNaM), which is efficiently PCR-amplified and transcribed in vitro, and whose unique mechanism of replication has been characterized. However, expansion of an organism's genetic alphabet presents new and unprecedented challenges: the unnatural nucleoside triphosphates must be available inside the cell; endogenous polymerases must be able to use the unnatural triphosphates to faithfully replicate DNA contg. the UBP within the complex cellular milieu; and finally, the UBP must be stable in the presence of pathways that maintain the integrity of DNA. Here we show that an exogenously expressed algal nucleotide triphosphate transporter efficiently imports the triphosphates of both d5SICS and dNaM (d5SICSTP and dNaMTP) into Escherichia coli, and that the endogenous replication machinery uses them to accurately replicate a plasmid contg. d5SICS-dNaM. Neither the presence of the unnatural triphosphates nor the replication of the UBP introduces a notable growth burden. Lastly, we find that the UBP is not efficiently excised by DNA repair pathways. Thus, the resulting bacterium is the first organism to propagate stably an expanded genetic alphabet.
- 46Rech, E. L.; Arber, W. Biodiversity as a Source for Synthetic Domestication of Useful Specific Traits. Ann. Appl. Biol. 2013, 162 (2), 141– 144, DOI: 10.1111/aab.12013There is no corresponding record for this reference.
- 47Rech, E. Genomics and Synthetic Biology as a Viable Option to Intensify Sustainable Use of Biodiversity. Nat. Precedings 2011, 1– 1, DOI: 10.1038/npre.2011.5759.1There is no corresponding record for this reference.
- 48Kiyama, H.; Kakizawa, S.; Sasajima, Y.; Tahara, Y. O.; Miyata, M. Reconstitution of a Minimal Motility System Based on Spiroplasma Swimming by Two Bacterial Actins in a Synthetic Minimal Bacterium. Sci. Adv. 2022, 8 (48), eabo7490, DOI: 10.1126/sciadv.abo7490There is no corresponding record for this reference.
- 49Geiger, O.; Sanchez-Flores, A.; Padilla-Gomez, J.; Degli Esposti, M. Multiple Approaches of Cellular Metabolism Define the Bacterial Ancestry of Mitochondria. Sci. Adv. 2023, 9 (32), eadh0066, DOI: 10.1126/sciadv.adh0066There is no corresponding record for this reference.
- 50Pinheiro, V. B.; Taylor, A. I.; Cozens, C.; Abramov, M.; Renders, M.; Zhang, S.; Chaput, J. C.; Wengel, J.; Peak-Chew, S.-Y.; McLaughlin, S. H.; Herdewijn, P.; Holliger, P. Synthetic Genetic Polymers Capable of Heredity and Evolution. Science 2012, 336 (6079), 341– 344, DOI: 10.1126/science.121762250https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XlslOqtL0%253D&md5=4402af6a172599d03c6b7f8a65b7cea0Synthetic Genetic Polymers Capable of Heredity and EvolutionPinheiro, Vitor B.; Taylor, Alexander I.; Cozens, Christopher; Abramov, Mikhail; Renders, Marleen; Zhang, Su; Chaput, John C.; Wengel, Jesper; Peak-Chew, Sew-Yeu; McLaughlin, Stephen H.; Herdewijn, Piet; Holliger, PhilippScience (Washington, DC, United States) (2012), 336 (6079), 341-344CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Genetic information storage and processing rely on just two polymers, DNA and RNA, yet whether their role reflects evolutionary history or fundamental functional constraints is currently unknown. With the use of polymerase evolution and design, we show that genetic information can be stored in and recovered from six alternative genetic polymers based on simple nucleic acid architectures not found in nature [xeno-nucleic acids (XNAs)]. We also select XNA aptamers, which bind their targets with high affinity and specificity, demonstrating that beyond heredity, specific XNAs have the capacity for Darwinian evolution and folding into defined structures. Thus, heredity and evolution, two hallmarks of life, are not limited to DNA and RNA but are likely to be emergent properties of polymers capable of information storage.
- 51Hoshika, S.; Leal, N. A.; Kim, M.-J.; Kim, M.-S.; Karalkar, N. B.; Kim, H.-J.; Bates, A. M.; Watkins, N. E., Jr; SantaLucia, H. A.; Meyer, A. J.; DasGupta, S.; Piccirilli, J. A.; Ellington, A. D.; SantaLucia, J., Jr; Georgiadis, M. M.; Benner, S. A. Hachimoji DNA and RNA: A Genetic System with Eight Building Blocks. Science 2019, 363 (6429), 884– 887, DOI: 10.1126/science.aat097151https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXjt1elu7c%253D&md5=1ae87a691cfdfbe36a819d4782c305b9Hachimoji DNA and RNA: A genetic system with eight building blocksHoshika, Shuichi; Leal, Nicole A.; Kim, Myong-Jung; Kim, Myong-Sang; Karalkar, Nilesh B.; Kim, Hyo-Joong; Bates, Alison M.; Watkins, Norman E., Jr.; SantaLucia, Holly A.; Meyer, Adam J.; DasGupta, Saurja; Piccirilli, Joseph A.; Ellington, Andrew D.; SantaLucia, John, Jr.; Georgiadis, Millie M.; Benner, Steven A.Science (Washington, DC, United States) (2019), 363 (6429), 884-887CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)We report DNA- and RNA-like systems built from eight nucleotide "letters" (hence the name "hachimoji") that form four orthogonal pairs. These synthetic systems meet the structural requirements needed to support Darwinian evolution, including a polyelectrolyte backbone, predictable thermodn. stability, and stereoregular building blocks that fit a Schroedinger aperiodic crystal. Measured thermodn. parameters predict the stability of hachimoji duplexes, allowing hachimoji DNA to increase the information d. of natural terran DNA. Three crystal structures show that the synthetic building blocks do not perturb the aperiodic crystal seen in the DNA double helix. Hachimoji DNA was then transcribed to give hachimoji RNA in the form of a functioning fluorescent hachimoji aptamer. These results expand the scope of mol. structures that might support life, including life throughout the cosmos.
- 52Jumper, J.; Evans, R.; Pritzel, A.; Green, T.; Figurnov, M.; Ronneberger, O.; Tunyasuvunakool, K.; Bates, R.; Žídek, A.; Potapenko, A.; Bridgland, A.; Meyer, C.; Kohl, S. A. A.; Ballard, A. J.; Cowie, A.; Romera-Paredes, B.; Nikolov, S.; Jain, R.; Adler, J.; Back, T.; Petersen, S.; Reiman, D.; Clancy, E.; Zielinski, M.; Steinegger, M.; Pacholska, M.; Berghammer, T.; Bodenstein, S.; Silver, D.; Vinyals, O.; Senior, A. W.; Kavukcuoglu, K.; Kohli, P.; Hassabis, D. Highly Accurate Protein Structure Prediction with AlphaFold. Nature 2021, 596 (7873), 583– 589, DOI: 10.1038/s41586-021-03819-252https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhvVaktrrL&md5=25964ab1157cd5b74a437333dd86650dHighly accurate protein structure prediction with AlphaFoldJumper, John; Evans, Richard; Pritzel, Alexander; Green, Tim; Figurnov, Michael; Ronneberger, Olaf; Tunyasuvunakool, Kathryn; Bates, Russ; Zidek, Augustin; Potapenko, Anna; Bridgland, Alex; Meyer, Clemens; Kohl, Simon A. A.; Ballard, Andrew J.; Cowie, Andrew; Romera-Paredes, Bernardino; Nikolov, Stanislav; Jain, Rishub; Adler, Jonas; Back, Trevor; Petersen, Stig; Reiman, David; Clancy, Ellen; Zielinski, Michal; Steinegger, Martin; Pacholska, Michalina; Berghammer, Tamas; Bodenstein, Sebastian; Silver, David; Vinyals, Oriol; Senior, Andrew W.; Kavukcuoglu, Koray; Kohli, Pushmeet; Hassabis, DemisNature (London, United Kingdom) (2021), 596 (7873), 583-589CODEN: NATUAS; ISSN:0028-0836. (Nature Portfolio)Proteins are essential to life, and understanding their structure can facilitate a mechanistic understanding of their function. Through an enormous exptl. effort, the structures of around 100,000 unique proteins have been detd., but this represents a small fraction of the billions of known protein sequences. Structural coverage is bottlenecked by the months to years of painstaking effort required to det. a single protein structure. Accurate computational approaches are needed to address this gap and to enable large-scale structural bioinformatics. Predicting the three-dimensional structure that a protein will adopt based solely on its amino acid sequence-the structure prediction component of the 'protein folding problem'-has been an important open research problem for more than 50 years. Despite recent progress, existing methods fall far short of at. accuracy, esp. when no homologous structure is available. Here we provide the first computational method that can regularly predict protein structures with at. accuracy even in cases in which no similar structure is known. We validated an entirely redesigned version of our neural network-based model, AlphaFold, in the challenging 14th Crit. Assessment of protein Structure Prediction (CASP14), demonstrating accuracy competitive with exptl. structures in a majority of cases and greatly outperforming other methods. Underpinning the latest version of AlphaFold is a novel machine learning approach that incorporates phys. and biol. knowledge about protein structure, leveraging multi-sequence alignments, into the design of the deep learning algorithm.
- 53Lee, J. W.; Na, D.; Park, J. M.; Lee, J.; Choi, S.; Lee, S. Y. Systems Metabolic Engineering of Microorganisms for Natural and Non-Natural Chemicals. Nat. Chem. Biol. 2012, 8 (6), 536– 546, DOI: 10.1038/nchembio.97053https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XntF2nt70%253D&md5=c9ca7c6d604a94b7539387f3ed463e6fSystems metabolic engineering of microorganisms for natural and non-natural chemicalsLee, Jeong Wook; Na, Dokyun; Park, Jong Myoung; Lee, Joungmin; Choi, Sol; Lee, Sang YupNature Chemical Biology (2012), 8 (6), 536-546CODEN: NCBABT; ISSN:1552-4450. (Nature Publishing Group)A review. Growing concerns over limited fossil resources and assocd. environmental problems are motivating the development of sustainable processes for the prodn. of chems., fuels and materials from renewable resources. Metabolic engineering is a key enabling technol. for transforming microorganisms into efficient cell factories for these compds. Systems metabolic engineering, which incorporates the concepts and techniques of systems biol., synthetic biol. and evolutionary engineering at the systems level, offers a conceptual and technol. framework to speed the creation of new metabolic enzymes and pathways or the modification of existing pathways for the optimal prodn. of desired products. Here we discuss the general strategies of systems metabolic engineering and examples of its application and offer insights as to when and how each of the different strategies should be used. Finally, we highlight the limitations and challenges to be overcome for the systems metabolic engineering of microorganisms at more advanced levels.
- 54Nielsen, J. Systems Biology of Metabolism. Annu. Rev. Biochem. 2017, 86, 245– 275, DOI: 10.1146/annurev-biochem-061516-04475754https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXksVCqsLc%253D&md5=7bcd6c37b4b2a4f31a09469c652baa0dSystems Biology of MetabolismNielsen, JensAnnual Review of Biochemistry (2017), 86 (), 245-275CODEN: ARBOAW; ISSN:0066-4154. (Annual Reviews)Metab. is highly complex and involves thousands of different connected reactions; it is therefore necessary to use math. models for holistic studies. The use of math. models in biol. is referred to as systems biol. In this review, the principles of systems biol. are described, and two different types of math. models used for studying metab. are discussed: kinetic models and genome-scale metabolic models. The use of different omics technologies, including transcriptomics, proteomics, metabolomics, and fluxomics, for studying metab. is presented. Finally, the application of systems biol. for analyzing global regulatory structures, engineering the metab. of cell factories, and analyzing human diseases is discussed.
- 55Choi, K. R.; Jang, W. D.; Yang, D.; Cho, J. S.; Park, D.; Lee, S. Y. Systems Metabolic Engineering Strategies: Integrating Systems and Synthetic Biology with Metabolic Engineering. Trends Biotechnol. 2019, 37 (8), 817– 837, DOI: 10.1016/j.tibtech.2019.01.00355https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhvF2ntLg%253D&md5=958b9cb4e7d60ebda46f6b086dcd7e97Systems Metabolic Engineering Strategies: Integrating Systems and Synthetic Biology with Metabolic EngineeringChoi, Kyeong Rok; Jang, Woo Dae; Yang, Dongsoo; Cho, Jae Sung; Park, Dahyeon; Lee, Sang YupTrends in Biotechnology (2019), 37 (8), 817-837CODEN: TRBIDM; ISSN:0167-7799. (Elsevier Ltd.)A review. Metabolic engineering allows development of microbial strains efficiently producing chems. and materials, but it requires much time, effort, and cost to make the strains industrially competitive. Systems metabolic engineering, which integrates tools and strategies of systems biol., synthetic biol., and evolutionary engineering with traditional metabolic engineering, has recently been used to facilitate development of high-performance strains. The past decade has witnessed this interdisciplinary strategy continuously being improved toward the development of industrially competitive overproducer strains. In this article, current trends in systems metabolic engineering including tools and strategies are reviewed, focusing on recent developments in selection of host strains, metabolic pathway reconstruction, tolerance enhancement, and metabolic flux optimization. Also, future challenges and prospects are discussed.
- 56Karim, A. S.; Dudley, Q. M.; Juminaga, A.; Yuan, Y.; Crowe, S. A.; Heggestad, J. T.; Garg, S.; Abdalla, T.; Grubbe, W. S.; Rasor, B. J.; Coar, D. N.; Torculas, M.; Krein, M.; Liew, F. E.; Quattlebaum, A.; Jensen, R. O.; Stuart, J. A.; Simpson, S. D.; Köpke, M.; Jewett, M. C. In Vitro Prototyping and Rapid Optimization of Biosynthetic Enzymes for Cell Design. Nat. Chem. Biol. 2020, 16 (8), 912– 919, DOI: 10.1038/s41589-020-0559-056https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtFKgtL3K&md5=aac0bdcd46766b221480e35b41f9639cIn vitro prototyping and rapid optimization of biosynthetic enzymes for cell designKarim, Ashty S.; Dudley, Quentin M.; Juminaga, Alex; Yuan, Yongbo; Crowe, Samantha A.; Heggestad, Jacob T.; Garg, Shivani; Abdalla, Tanus; Grubbe, William S.; Rasor, Blake J.; Coar, David N.; Torculas, Maria; Krein, Michael; Liew, FungMin; Quattlebaum, Amy; Jensen, Rasmus O.; Stuart, Jeffrey A.; Simpson, Sean D.; Kopke, Michael; Jewett, Michael C.Nature Chemical Biology (2020), 16 (8), 912-919CODEN: NCBABT; ISSN:1552-4450. (Nature Research)The design and optimization of biosynthetic pathways for industrially relevant, non-model organisms is challenging due to transformation idiosyncrasies, reduced nos. of validated genetic parts and a lack of high-throughput workflows. Here the authors describe a platform for in vitro prototyping and rapid optimization of biosynthetic enzymes (iPROBE) to accelerate this process. In iPROBE, cell lysates are enriched with biosynthetic enzymes by cell-free protein synthesis and then metabolic pathways are assembled in a mix-and-match fashion to assess pathway performance. The authors demonstrate iPROBE by screening 54 different cell-free pathways for 3-hydroxybutyrate prodn. and optimizing a six-step butanol pathway across 205 permutations using data-driven design. Observing a strong correlation (r = 0.79) between cell-free and cellular performance, the authors then scaled up the authors' highest-performing pathway, which improved in vivo 3-HB prodn. in Clostridium by 20-fold to 14.63 ± 0.48 g L-1. The authors expect iPROBE to accelerate design-build-test cycles for industrial biotechnol.
- 57Dudley, Q. M.; Karim, A. S.; Nash, C. J.; Jewett, M. C. In Vitro Prototyping of Limonene Biosynthesis Using Cell-Free Protein Synthesis. Metab. Eng. 2020, 61, 251– 260, DOI: 10.1016/j.ymben.2020.05.00657https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhtl2itrzI&md5=bc5f4261c3a62805f1c904e797ec2134In vitro prototyping of limonene biosynthesis using cell-free protein synthesisDudley, Quentin M.; Karim, Ashty S.; Nash, Connor J.; Jewett, Michael C.Metabolic Engineering (2020), 61 (), 251-260CODEN: MEENFM; ISSN:1096-7176. (Elsevier B.V.)Metabolic engineering of microorganisms to produce sustainable chems. has emerged as an important part of the global bioeconomy. Unfortunately, efforts to design and engineer microbial cell factories are challenging because design-build-test cycles, iterations of re-engineering organisms to test and optimize new sets of enzymes, are slow. To alleviate this challenge, we demonstrate a cell-free approach termed in vitro Prototyping and Rapid Optimization of Biosynthetic Enzymes (or iPROBE). In iPROBE, a large no. of pathway combinations can be rapidly built and optimized. The key idea is to use cell-free protein synthesis (CFPS) to manuf. pathway enzymes in sep. reactions that are then mixed to modularly assemble multiple, distinct biosynthetic pathways. As a model, we apply our approach to the 9-step heterologous enzyme pathway to limonene in exts. from Escherichia coli. In iterative cycles of design, we studied the impact of 54 enzyme homologs, multiple enzyme levels, and cofactor concns. on pathway performance. In total, we screened over 150 unique sets of enzymes in 580 unique pathway conditions to increase limonene prodn. in 24 h from 0.2 to 4.5 mM (23-610 mg/L). Finally, to demonstrate the modularity of this pathway, we also synthesized the biofuel precursors pinene and bisabolene. We anticipate that iPROBE will accelerate design-build-test cycles for metabolic engineering, enabling data-driven multiplexed cell-free methods for testing large combinations of biosynthetic enzymes to inform cellular design.
- 58Averesch, N. J. H.; Krömer, J. O. Metabolic Engineering of the Shikimate Pathway for Production of Aromatics and Derived Compounds-Present and Future Strain Construction Strategies. Front. Bioeng. Biotechnol. 2018, 6, 32, DOI: 10.3389/fbioe.2018.0003258https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1MjgsVGqtw%253D%253D&md5=413681743faac53a82a74216557d0b12Metabolic Engineering of the Shikimate Pathway for Production of Aromatics and Derived Compounds-Present and Future Strain Construction StrategiesAveresch Nils J H; Kromer Jens OFrontiers in bioengineering and biotechnology (2018), 6 (), 32 ISSN:2296-4185.The aromatic nature of shikimate pathway intermediates gives rise to a wealth of potential bio-replacements for commonly fossil fuel-derived aromatics, as well as naturally produced secondary metabolites. Through metabolic engineering, the abundance of certain intermediates may be increased, while draining flux from other branches off the pathway. Often targets for genetic engineering lie beyond the shikimate pathway, altering flux deep in central metabolism. This has been extensively used to develop microbial production systems for a variety of compounds valuable in chemical industry, including aromatic and non-aromatic acids like muconic acid, para-hydroxybenzoic acid, and para-coumaric acid, as well as aminobenzoic acids and aromatic α-amino acids. Further, many natural products and secondary metabolites that are valuable in food- and pharma-industry are formed outgoing from shikimate pathway intermediates. (Re)construction of such routes has been shown by de novo production of resveratrol, reticuline, opioids, and vanillin. In this review, strain construction strategies are compared across organisms and put into perspective with requirements by industry for commercial viability. Focus is put on enhancing flux to and through shikimate pathway, and engineering strategies are assessed in order to provide a guideline for future optimizations.
- 59Jang, W. D.; Kim, G. B.; Lee, S. Y. An Interactive Metabolic Map of Bio-Based Chemicals. Trends Biotechnol. 2023, 41 (1), 10– 14, DOI: 10.1016/j.tibtech.2022.07.013There is no corresponding record for this reference.
- 60Lai, H.-E.; Obled, A. M. C.; Chee, S. M.; Morgan, R. M.; Lynch, R.; Sharma, S. V.; Moore, S. J.; Polizzi, K. M.; Goss, R. J. M.; Freemont, P. S. GenoChemetic Strategy for Derivatization of the Violacein Natural Product Scaffold. ACS Chem. Biol. 2021, 16 (11), 2116– 2123, DOI: 10.1021/acschembio.1c0048360https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXit1ehtr3I&md5=c0abd319d896030a21f20c0660a2b8a1GenoChemetic Strategy for Derivatization of the Violacein Natural Product ScaffoldLai, Hung-En; Obled, Alan M. C.; Chee, Soo Mei; Morgan, Rhodri M.; Lynch, Rosemary; Sharma, Sunil V.; Moore, Simon J.; Polizzi, Karen M.; Goss, Rebecca J. M.; Freemont, Paul S.ACS Chemical Biology (2021), 16 (11), 2116-2123CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)Natural products and their analogs are often challenging to synthesize due to their complex scaffolds and embedded functional groups. Solely relying on engineering the biosynthesis of natural products may lead to limited compd. diversity. Integrating synthetic biol. with synthetic chem. allows rapid access to much more diverse portfolios of xenobiotic compds., which may accelerate the discovery of new therapeutics. As a proof-of-concept, by supplementing an Escherichia coli strain expressing the violacein biosynthesis pathway with 5-bromo-tryptophan in vitro or tryptophan 7-halogenase RebH in vivo, six halogenated analogs of violacein or deoxyviolacein were generated, demonstrating the promiscuity of the violacein biosynthesis pathway. Furthermore, 20 new derivs. were generated from 5-brominated violacein analogs via the Suzuki-Miyaura cross-coupling reaction directly using the crude ext. without prior purifn. Herein we demonstrate a flexible and rapid approach to access a diverse chem. space that can be applied to a wide range of natural product scaffolds.
- 61Galanie, S.; Thodey, K.; Trenchard, I. J.; Filsinger Interrante, M.; Smolke, C. D. Complete Biosynthesis of Opioids in Yeast. Science 2015, 349 (6252), 1095– 1100, DOI: 10.1126/science.aac937361https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsVyntbbK&md5=7304186260192653b8c7c78b9119fde3Complete biosynthesis of opioids in yeastGalanie, Stephanie; Thodey, Kate; Trenchard, Isis J.; Filsinger Interrante, Maria; Smolke, Christina D.Science (Washington, DC, United States) (2015), 349 (6252), 1095-1100CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Opioids are the primary drugs used in Western medicine for pain management and palliative care. Farming of opium poppies remains the sole source of these essential medicines, despite diverse market demands and uncertainty in crop yields due to weather, climate change, and pests. We engineered yeast to produce the selected opioid compds. thebaine and hydrocodone starting from sugar. All work was conducted in a lab. that is permitted and secured for work with controlled substances. We combined enzyme discovery, enzyme engineering, and pathway and strain optimization to realize full opiate biosynthesis in yeast. The resulting opioid biosynthesis strains required the expression of 21 (thebaine) and 23 (hydrocodone) enzyme activities from plants, mammals, bacteria, and yeast itself. This is a proof of principle, and major hurdles remain before optimization and scale-up could be achieved. Open discussions of options for governing this technol. are also needed in order to responsibly realize alternative supplies for these medically relevant compds.
- 62Trenchard, I. J.; Siddiqui, M. S.; Thodey, K.; Smolke, C. D. De Novo Production of the Key Branch Point Benzylisoquinoline Alkaloid Reticuline in Yeast. Metab. Eng. 2015, 31, 74– 83, DOI: 10.1016/j.ymben.2015.06.01062https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtFOktL%252FE&md5=e07d3d284b8d8cb60631dd1045b2d68aDe novo production of the key branch point benzylisoquinoline alkaloid reticuline in yeastTrenchard, Isis J.; Siddiqui, Michael S.; Thodey, Kate; Smolke, Christina D.Metabolic Engineering (2015), 31 (), 74-83CODEN: MEENFM; ISSN:1096-7176. (Elsevier B. V.)Microbial biosynthesis for plant-based natural products, such as the benzylisoquinoline alkaloids (BIAs), has the potential to address limitations in plant-based supply of established drugs and make new mols. available for drug discovery. While yeast strains have been engineered to produce a variety of downstream BIAs including the opioids, these strains have relied on feeding an early BIA substrate. We describe the de novo synthesis of the major BIA branch point intermediate reticuline via norcoclaurine in Saccharomyces cerevisiae. Modifications were introduced into yeast central metab. to increase supply of the BIA precursor tyrosine, allowing us to achieve a 60-fold increase in prodn. of the early benzylisoquinoline scaffold from fed dopamine with no supply of exogenous tyrosine. Yeast strains further engineered to express a mammalian tyrosine hydroxylase, 4 mammalian tetrahydrobiopterin biosynthesis and recycling enzymes, and a bacterial DOPA decarboxylase produced norcoclaurine de novo. We further increased prodn. of early benzylisoquinoline scaffolds by 160-fold through introducing mutant tyrosine hydroxylase enzymes, an optimized plant norcoclaurine synthase variant, and optimizing culture conditions. Finally, we incorporated 5 addnl. plant enzymes (3 methyltransferases, a cytochrome P 450, and its reductase partner) to achieve de novo prodn. of the key branch point mol. reticuline with a titer of 19.2 μg/L. These strains and reconstructed pathways will serve as a platform for the biosynthesis of diverse natural and novel BIAs.
- 63Chan, C. T. Y.; Lee, J. W.; Cameron, D. E.; Bashor, C. J.; Collins, J. J. Deadman” and “Passcode” Microbial Kill Switches for Bacterial Containment. Nat. Chem. Biol. 2016, 12 (2), 82– 86, DOI: 10.1038/nchembio.197963https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhvFemsbrE&md5=c1cd6452791fc321b2432e61ad682557'Deadman' and 'Passcode' microbial kill switches for bacterial containmentChan, Clement T. Y.; Lee, Jeong Wook; Cameron, D. Ewen; Bashor, Caleb J.; Collins, James J.Nature Chemical Biology (2016), 12 (2), 82-86CODEN: NCBABT; ISSN:1552-4450. (Nature Publishing Group)Biocontainment systems that couple environmental sensing with circuit-based control of cell viability could be used to prevent escape of genetically modified microbes into the environment. Here we present two engineered safeguard systems known as the 'Deadman' and 'Passcode' kill switches. The Deadman kill switch uses unbalanced reciprocal transcriptional repression to couple a specific input signal with cell survival. The Passcode kill switch uses a similar two-layered transcription design and incorporates hybrid LacI-GalR family transcription factors to provide diverse and complex environmental inputs to control circuit function. These synthetic gene circuits efficiently kill Escherichia coli and can be readily reprogrammed to change their environmental inputs, regulatory architecture and killing mechanism.
- 64Caliando, B. J.; Voigt, C. A. Targeted DNA Degradation Using a CRISPR Device Stably Carried in the Host Genome. Nat. Commun. 2015, 6, 6989, DOI: 10.1038/ncomms798964https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhtF2kt7fJ&md5=a01b28683e14e8f6c185dbdfcc300293Targeted DNA degradation using a CRISPR device stably carried in the host genomeCaliando, Brian J.; Voigt, Christopher A.Nature Communications (2015), 6 (), 6989CODEN: NCAOBW; ISSN:2041-1723. (Nature Publishing Group)Once an engineered organism completes its task, it is useful to degrade the assocd. DNA to reduce environmental release and protect intellectual property. Here we present a genetically encoded device (DNAi) that responds to a transcriptional input and degrades user-defined DNA. This enables engineered regions to be obscured when the cell enters a new environment. DNAi is based on type-IE CRISPR biochem. and a synthetic CRISPR array defines the DNA target(s). When the input is on, plasmid DNA is degraded 108-fold. When the genome is targeted, this causes cell death, reducing viable cells by a factor of 108. Further, the CRISPR nuclease can direct degrdn. to specific genomic regions (for example, engineered or inserted DNA), which could be used to complicate recovery and sequencing efforts. DNAi can be stably carried in an engineered organism, with no impact on cell growth, plasmid stability or DNAi inducibility even after passaging for >2 mo.
- 65Nyerges, A.; Vinke, S.; Flynn, R.; Owen, S. V.; Rand, E. A.; Budnik, B.; Keen, E.; Narasimhan, K.; Marchand, J. A.; Baas-Thomas, M.; Liu, M.; Chen, K.; Chiappino-Pepe, A.; Hu, F.; Baym, M.; Church, G. M. A Swapped Genetic Code Prevents Viral Infections and Gene Transfer. Nature 2023, 615 (7953), 720– 727, DOI: 10.1038/s41586-023-05824-zThere is no corresponding record for this reference.
- 66Scrinis, G.; Lyons, K. The Emerging Nano-Corporate Paradigm: Nanotechnology and the Transformation of Nature, Food and Agri-Food Systems. Int. J. Sociol. Agric. Food 2007, 15 (2), 22– 44There is no corresponding record for this reference.
- 67Nitrogen fixing bacteria - microbial fertilizer. https://www.pivotbio.com/ (accessed on April 30, 2023).There is no corresponding record for this reference.
- 68Kaul, S.; Choudhary, M.; Gupta, S.; Dhar, M. K. Engineering Host Microbiome for Crop Improvement and Sustainable Agriculture. Front. Microbiol. 2021, 12, 635917, DOI: 10.3389/fmicb.2021.635917There is no corresponding record for this reference.
- 69Mueller, U. G.; Sachs, J. L. Engineering Microbiomes to Improve Plant and Animal Health. Trends Microbiol. 2015, 23 (10), 606– 617, DOI: 10.1016/j.tim.2015.07.00969https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsFKhtL7L&md5=8743f515980f584435a175d2b74a33bdEngineering Microbiomes to Improve Plant and Animal HealthMueller, U. G.; Sachs, J. L.Trends in Microbiology (2015), 23 (10), 606-617CODEN: TRMIEA; ISSN:0966-842X. (Elsevier Ltd.)A review. Animal and plant microbiomes encompass diverse microbial communities that colonize every accessible host tissue. These microbiomes enhance host functions, contributing to host health and fitness. A novel approach to improve animal and plant fitness is to artificially select upon microbiomes, thus engineering evolved microbiomes with specific effects on host fitness. The authors call this engineering approach host-mediated microbiome selection, because this method selects upon microbial communities indirectly through the host and leverages host traits that evolved to influence microbiomes. In essence, host phenotypes were used as probes to gauge and manipulate those microbiome functions that impact host fitness. To facilitate research on host-mediated microbiome engineering, the authors explain and compare the principal methods to impose artificial selection on microbiomes; discuss advantages and potential challenges of each method; offer a skeptical appraisal of each method in light of these potential challenges; and outline exptl. strategies to optimize microbiome engineering. Finally, the authors develop a predictive framework for microbiome engineering that organizes research around principles of artificial selection, quant. genetics, and microbial community ecol.
- 70Jin Song, S.; Woodhams, D. C.; Martino, C.; Allaband, C.; Mu, A.; Javorschi-Miller-Montgomery, S.; Suchodolski, J. S.; Knight, R. Engineering the Microbiome for Animal Health and Conservation. Exp. Biol. Med. 2019, 244 (6), 494– 504, DOI: 10.1177/1535370219830075There is no corresponding record for this reference.
- 71Aziz, C. E.; Borden, R. C.; Coates, J. D.; Cox, E. E.; Downey, D. C.; Evans, P. J.; Hatzinger, P. B.; Henry, B. M.; Andrew Jackson, W.; Krug, T. A.; Tony Lieberman, M.; Loehr, R. C.; Norris, R. D.; Nzengung, V. A.; Perlmutter, M. W.; Schaefer, C. E.; Stroo, H. F.; Herb Ward, C.; Winstead, C. J.; Wolfe, C. In Situ Bioremediation of Perchlorate in Groundwater; Springer: New York.There is no corresponding record for this reference.
- 72Hou, D.; O’Connor, D.; Igalavithana, A. D.; Alessi, D. S.; Luo, J.; Tsang, D. C. W.; Sparks, D. L.; Yamauchi, Y.; Rinklebe, J.; Ok, Y. S. Metal Contamination and Bioremediation of Agricultural Soils for Food Safety and Sustainability. Nature Reviews Earth & Environment 2020, 1 (7), 366– 381, DOI: 10.1038/s43017-020-0061-yThere is no corresponding record for this reference.
- 73Maity, W.; Maity, S.; Bera, S.; Roy, A. Emerging Roles of PETase and MHETase in the Biodegradation of Plastic Wastes. Appl. Biochem. Biotechnol. 2021, 193 (8), 2699– 2716, DOI: 10.1007/s12010-021-03562-4There is no corresponding record for this reference.
- 74de Lorenzo, V.; Prather, K. L.; Chen, G.-Q.; O’Day, E.; von Kameke, C.; Oyarzún, D. A.; Hosta-Rigau, L.; Alsafar, H.; Cao, C.; Ji, W.; Okano, H.; Roberts, R. J.; Ronaghi, M.; Yeung, K.; Zhang, F.; Lee, S. Y. The Power of Synthetic Biology for Bioproduction, Remediation and Pollution Control: The UN’s Sustainable Development Goals Will Inevitably Require the Application of Molecular Biology and Biotechnology on a Global Scale. EMBO Rep 2018, DOI: 10.15252/embr.201745658There is no corresponding record for this reference.
- 75Thavarajah, W.; Verosloff, M. S.; Jung, J. K.; Alam, K. K.; Miller, J. D.; Jewett, M. C.; Young, S. L.; Lucks, J. B. A Primer on Emerging Field-Deployable Synthetic Biology Tools for Global Water Quality Monitoring. NPJ. Clean Water 2020, DOI: 10.1038/s41545-020-0064-8There is no corresponding record for this reference.
- 76Gupta, R. M.; Schnitzler, G. R.; Fang, S.; Lee-Kim, V. S.; Barry, A. Multiomic Analysis and CRISPR Perturbation Screens Identify Endothelial Cell Programs and Novel Therapeutic Targets for Coronary Artery Disease. Arterioscler. Thromb. Vasc. Biol. 2023, 43 (5), 600– 608, DOI: 10.1161/ATVBAHA.123.318328There is no corresponding record for this reference.
- 77Virchow, R. L. K. Disease, Life and Man: Selected Essays; Rather, L. J., Translator; Stanford University Press, 1958.There is no corresponding record for this reference.
- 78Center for Biologics Evaluation; Research. Approved Cellular and Gene Therapy Products. U.S. Food and Drug Administration. https://www.fda.gov/vaccines-blood-biologics/cellular-gene-therapy-products/approved-cellular-and-gene-therapy-products (accessed on August 21, 2023).There is no corresponding record for this reference.
- 79Bashor, C. J.; Hilton, I. B.; Bandukwala, H.; Smith, D. M.; Veiseh, O. Engineering the next Generation of Cell-Based Therapeutics. Nat. Rev. Drug Discovery 2022, 21 (9), 655– 675, DOI: 10.1038/s41573-022-00476-6There is no corresponding record for this reference.
- 80Rosenbaum, L. Tragedy, Perseverance, and Chance - The Story of CAR-T Therapy. N. Engl. J. Med. 2017, 377 (14), 1313– 1315, DOI: 10.1056/NEJMp1711886There is no corresponding record for this reference.
- 81Hay, A. E.; Cheung, M. C. CAR T-Cells: Costs, Comparisons, and Commentary. J. Med. Econ. 2019, 22 (7), 613– 615, DOI: 10.1080/13696998.2019.1582059There is no corresponding record for this reference.
- 82Sampson, K.; Sorenson, C.; Adamala, K. FDA needs to get ready to evaluate synthetic cells, the next generation of therapeutics. STAT. https://www.statnews.com/2022/07/26/fda-develop-framework-evaluate-synthetic-cells/ (accessed on January 6, 2024).There is no corresponding record for this reference.
- 83Lussier, F.; Staufer, O.; Platzman, I.; Spatz, J. P. Can Bottom-Up Synthetic Biology Generate Advanced Drug-Delivery Systems?. Trends Biotechnol. 2021, 39 (5), 445– 459, DOI: 10.1016/j.tibtech.2020.08.00283https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhs1OltLfJ&md5=7fae97c9cc9dc7fcaec9b229371a4c69Can Bottom-Up Synthetic Biology Generate Advanced Drug-Delivery SystemsLussier, Felix; Staufer, Oskar; Platzman, Ilia; Spatz, Joachim P.Trends in Biotechnology (2021), 39 (5), 445-459CODEN: TRBIDM; ISSN:0167-7799. (Elsevier Ltd.)A review. Creating a magic bullet that can selectively kill cancer cells while sparing nearby healthy cells remains one of the most ambitious objectives in pharmacol. Nanomedicine, which relies on the use of nanotechnologies to fight disease, was envisaged to fulfill this coveted goal. Despite substantial progress, the structural complexity of therapeutic vehicles impedes their broad clin. application. Novel modular manufg. approaches for engineering programmable drug carriers may be able to overcome some fundamental limitations of nanomedicine. We discuss how bottom-up synthetic biol. principles, empowered by microfluidics, can palliate current drug carrier assembly limitations, and we demonstrate how such a magic bullet could be engineered from the bottom up to ultimately improve clin. outcomes for patients.
- 84Staufer, O.; Dietrich, F.; Rimal, R.; Schröter, M.; Fabritz, S.; Boehm, H.; Singh, S.; Möller, M.; Platzman, I.; Spatz, J. P. Bottom-up Assembly of Biomedical Relevant Fully Synthetic Extracellular Vesicles. Sci. Adv. 2021, 7 (36), eabg6666, DOI: 10.1126/sciadv.abg6666There is no corresponding record for this reference.
- 85Ghaemmaghamian, Z.; Zarghami, R.; Walker, G.; O’Reilly, E.; Ziaee, A. Stabilizing Vaccines via Drying: Quality by Design Considerations. Adv. Drug Delivery Rev. 2022, 187, 114313, DOI: 10.1016/j.addr.2022.11431385https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhsFKjsLfM&md5=268c97346f1c1595a47a83b73069aaa1Stabilizing vaccines via drying: Quality by design considerationsGhaemmaghamian, Zahra; Zarghami, Reza; Walker, Gavin; O'Reilly, Emmet; Ziaee, AhmadAdvanced Drug Delivery Reviews (2022), 187 (), 114313CODEN: ADDREP; ISSN:0169-409X. (Elsevier B.V.)A review. Pandemics and epidemics are continually challenging human beings' health and imposing major stresses on the societies particularly over the last few decades, when their frequency has increased significantly. Protecting humans from multiple diseases is best achieved through vaccination. However, vaccines thermal instability has always been a hurdle in their widespread application, esp. in less developed countries. Furthermore, insufficient vaccine processing capacity is also a major challenge for global vaccination programs. Continuous drying of vaccine formulations is one of the potential solns. to these challenges. This review highlights the challenges on implementing the continuous drying techniques for drying vaccines. The conventional drying methods, emerging technologies and their adaptation by biopharmaceutical industry are investigated considering the patented technologies for drying of vaccines. Moreover, the current progress in applying Quality by Design (QbD) in each of the drying techniques considering the crit. quality attributes (CQAs), crit. process parameters (CPPs) are comprehensively reviewed. An expert advice is presented on the required actions to be taken within the biopharmaceutical industry to move towards continuous stabilization of vaccines in the realm of QbD.
- 86Adiga, R.; Al-Adhami, M.; Andar, A.; Borhani, S.; Brown, S.; Burgenson, D.; Cooper, M. A.; Deldari, S.; Frey, D. D.; Ge, X.; Guo, H.; Gurramkonda, C.; Jensen, P.; Kostov, Y.; LaCourse, W.; Liu, Y.; Moreira, A.; Mupparapu, K.; Peñalber-Johnstone, C.; Pilli, M.; Punshon-Smith, B.; Rao, A.; Rao, G.; Rauniyar, P.; Snovida, S.; Taurani, K.; Tilahun, D.; Tolosa, L.; Tolosa, M.; Tran, K.; Vattem, K.; Veeraraghavan, S.; Wagner, B.; Wilhide, J.; Wood, D. W.; Zuber, A. Point-of-Care Production of Therapeutic Proteins of Good-Manufacturing-Practice Quality. Nat. Biomed Eng. 2018, 2 (9), 675– 686, DOI: 10.1038/s41551-018-0259-186https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtFensr3N&md5=bbe1f9134ed2590f5d0c500997cb50b5Point-of-care production of therapeutic proteins of good-manufacturing-practice qualityAdiga, Rajani; Al-adhami, Mustafa; Andar, Abhay; Borhani, Shayan; Brown, Sheniqua; Burgenson, David; Cooper, Merideth A.; Deldari, Sevda; Frey, Douglas D.; Ge, Xudong; Guo, Hui; Gurramkonda, Chandrasekhar; Jensen, Penny; Kostov, Yordan; LaCourse, William; Liu, Yang; Moreira, Antonio; Mupparapu, KarunaSri; Penalber-Johnstone, Chariz; Pilli, Manohar; Punshon-Smith, Benjamin; Rao, Aniruddha; Rao, Govind; Rauniyar, Priyanka; Snovida, Sergei; Taurani, Kanika; Tilahun, Dagmawi; Tolosa, Leah; Tolosa, Michael; Tran, Kevin; Vattem, Krishna; Veeraraghavan, Sudha; Wagner, Brandon; Wilhide, Joshua; Wood, David W.; Zuber, AdilNature Biomedical Engineering (2018), 2 (9), 675-686CODEN: NBEAB3; ISSN:2157-846X. (Nature Research)Manufg. technologies for biologics rely on large, centralized, good-manufg.-practice (GMP) prodn. facilities and on a cumbersome product-distribution network. Here, we report the development of an automated and portable medicines-on-demand device that enables consistent, small-scale GMP manufg. of therapeutic-grade biologics on a timescale of hours. The device couples the in vitro translation of target proteins from ribosomal DNA, using exts. from reconstituted lyophilized Chinese hamster ovary cells, with the continuous purifn. of the proteins. We used the device to reproducibly manuf. His-tagged granulocyte-colony stimulating factor, erythropoietin, glucose-binding protein and diphtheria toxoid DT5. Medicines-on-demand technol. may enable the rapid manufg. of biologics at the point of care.
- 87Ginsburg, G. S.; Phillips, K. A. Precision Medicine: From Science To Value. Health Aff. 2018, 37 (5), 694– 701, DOI: 10.1377/hlthaff.2017.1624There is no corresponding record for this reference.
- 88Agarwal, S.; Saha, S.; Balla, V. K.; Pal, A.; Barui, A.; Bodhak, S. Current Developments in 3D Bioprinting for Tissue and Organ Regeneration-A Review. Front. Mech. Eng. Chin 2020, DOI: 10.3389/fmech.2020.589171There is no corresponding record for this reference.
- 89Ghosh, S.; Yi, H.-G. A Review on Bioinks and Their Application in Plant Bioprinting. Int. J. Bioprint 2022, 8 (4), 612, DOI: 10.18063/ijb.v8i4.612There is no corresponding record for this reference.
- 90Santomartino, R.; Averesch, N. J. H.; Bhuiyan, M.; Cockell, C. S.; Colangelo, J.; Gumulya, Y.; Lehner, B.; Lopez-Ayala, I.; McMahon, S.; Mohanty, A.; Santa Maria, S. R.; Urbaniak, C.; Volger, R.; Yang, J.; Zea, L. Toward Sustainable Space Exploration: A Roadmap for Harnessing the Power of Microorganisms. Nat. Commun. 2023, 14 (1), 1391, DOI: 10.1038/s41467-023-37070-2There is no corresponding record for this reference.
- 91Rothschild, L. J. Synthetic Biology Meets Bioprinting: Enabling Technologies for Humans on Mars (and Earth). Biochem. Soc. Trans. 2016, 44 (4), 1158– 1164, DOI: 10.1042/BST2016006791https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhtlCju7jO&md5=e97bf6b90993c7664400df619e087f6cSynthetic biology meets bioprinting: enabling technologies for humans on Mars (and Earth)Rothschild, Lynn J.Biochemical Society Transactions (2016), 44 (4), 1158-1164CODEN: BCSTB5; ISSN:0300-5127. (Portland Press Ltd.)Human exploration off planet is severely limited by the cost of launching materials into space and by re-supply. Thus materials brought from Earth must be light, stable and reliable at destination. Using traditional approaches, a lunar or Mars base would require either transporting a hefty store of metals or heavy manufg. equipment and construction materials for in situ extn.; both would severely limit any other mission objectives. Long-term human space presence requires periodic replenishment, adding a massive cost overhead. Even robotic missions often sacrifice science goals for heavy radiation and thermal protection. Biol. has the potential to solve these problems because life can replicate and repair itself, and perform a wide variety of chem. reactions including making food, fuel and materials. Synthetic biol. enhances and expands life's evolved repertoire. Using organisms as feedstock, additive manufg. through bioprinting will make possible the dream of producing bespoke tools, food, smart fabrics and even replacement organs on demand. This new approach and the resulting novel products will enable human exploration and settlement on Mars, while providing new manufg. approaches for life on Earth.
- 92Averesch, N. J. H.; Berliner, A. J.; Nangle, S. N.; Zezulka, S.; Vengerova, G. L.; Ho, D.; Casale, C. A.; Lehner, B. A. E.; Snyder, J. E.; Clark, K. B.; Dartnell, L. R.; Criddle, C. S.; Arkin, A. P. Microbial Biomanufacturing for Space-Exploration-What to Take and When to Make. Nat. Commun. 2023, 14 (1), 2311, DOI: 10.1038/s41467-023-37910-1There is no corresponding record for this reference.
- 93Averesch, N. J. H. Choice of Microbial System for in-Situ Resource Utilization on Mars. Front. Astron. Space Sci. 2021, DOI: 10.3389/fspas.2021.700370There is no corresponding record for this reference.
- 94Cockell, C. S. Bridging the Gap between Microbial Limits and Extremes in Space: Space Microbial Biotechnology in the next 15 Years. Microb. Biotechnol. 2022, 15 (1), 29– 41, DOI: 10.1111/1751-7915.13927There is no corresponding record for this reference.
- 95Buddingh’, B. C.; van Hest, J. C. M. Artificial Cells: Synthetic Compartments with Life-like Functionality and Adaptivity. Acc. Chem. Res. 2017, 50 (4), 769– 777, DOI: 10.1021/acs.accounts.6b0051295https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXps1SisQ%253D%253D&md5=979537efa2cf22428e5de31f8267fb31Artificial Cells: Synthetic Compartments with Life-like Functionality and AdaptivityBuddingh', Bastiaan C.; van Hest, Jan C. M.Accounts of Chemical Research (2017), 50 (4), 769-777CODEN: ACHRE4; ISSN:0001-4842. (American Chemical Society)A review. Cells are highly advanced microreactors that form the basis of all life. Their fascinating complexity has inspired scientists to create analogs from synthetic and natural components using a bottom-up approach. The ultimate goal here is to assemble a fully man-made cell that displays functionality and adaptivity as advanced as that found in nature, which will not only provide insight into the fundamental processes in natural cells but also pave the way for new applications of such artificial cells. In this Account, the authors highlight their recent work and that of others on the construction of artificial cells. First, the authors will introduce the key features that characterize a living system; next, the authors will discuss how these have been imitated in artificial cells. First, compartmentalization is crucial to sep. the inner chem. milieu from the external environment. Current state-of-the-art artificial cells comprise subcompartments to mimic the hierarchical architecture of eukaryotic cells and tissue. Furthermore, synthetic gene circuits have been used to encode genetic information that creates complex behavior like pulses or feedback. Addnl., artificial cells have to reproduce to maintain a population. Controlled growth and fission of synthetic compartments have been demonstrated, but the extensive regulation of cell division in nature is still unmatched. Here, the authors also point out important challenges the field needs to overcome to realize its full potential. As artificial cells integrate increasing orders of functionality, maintaining a supporting metab. that can regenerate key metabolites becomes crucial. Furthermore, life does not operate in isolation. Natural cells constantly sense their environment, exchange (chem.) signals, and can move toward a chemoattractant. Here, the authors specifically explore recent efforts to reproduce such adaptivity in artificial cells. For instance, synthetic compartments have been produced that can recruit proteins to the membrane upon an external stimulus or modulate their membrane compn. and permeability to control their interaction with the environment. A next step would be the communication of artificial cells with either bacteria or another artificial cell. Indeed, examples of such primitive chem. signaling are presented. Finally, motility is important for many organisms and has, therefore, also been pursued in synthetic systems. Synthetic compartments that were designed to move in a directed, controlled manner have been assembled, and directed movement toward a chem. attractant is among one of the most life-like directions currently under research. Although the bottom-up construction of an artificial cell that can be truly considered "alive" is still an ambitious goal, the recent work discussed in this Account shows that this is an active field with contributions from diverse disciplines like materials chem. and biochem. Notably, research during the past decade has already provided valuable insights into complex synthetic systems with life-like properties. In the future, artificial cells are thought to contribute to an increased understanding of processes in natural cells and provide opportunities to create smart, autonomous, cell-like materials.
- 96Ichihashi, N. What Can We Learn from the Construction of in Vitro Replication Systems?. Ann. N.Y. Acad. Sci. 2019, 1447 (1), 144– 156, DOI: 10.1111/nyas.1404296https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3M%252Fis1yqtw%253D%253D&md5=0cb66816ca21190294e9a09ce2768e10What can we learn from the construction of in vitro replication systems?Ichihashi NorikazuAnnals of the New York Academy of Sciences (2019), 1447 (1), 144-156 ISSN:.Replication is a central function of living organisms. Several types of replication systems have been constructed in vitro from various molecules, including peptides, DNA, RNA, and proteins. In this review, I summarize the progress in the construction of replication systems over the past few decades and discuss what we can learn from their construction. I introduce various types of replication systems, supporting the feasibility of the spontaneous appearance of replication early in Earth's history. In the latter part of the review, I focus on parasitic replicators, one of the largest obstacles for sustainable replication. Compartmentalization is discussed as a possible solution.
- 97Groaz, A.; Moghimianavval, H.; Tavella, F.; Giessen, T. W.; Vecchiarelli, A. G.; Yang, Q.; Liu, A. P. Engineering Spatiotemporal Organization and Dynamics in Synthetic Cells. WIREs Nanomed. Nanobiotechnol. 2021, 13 (3), e1685, DOI: 10.1002/wnan.1685There is no corresponding record for this reference.
- 98Gaut, N. J.; Adamala, K. P. Reconstituting Natural Cell Elements in Synthetic Cells. Adv. Biol. 2021, 5 (3), e2000188, DOI: 10.1002/adbi.202000188There is no corresponding record for this reference.
- 99Stal, L. J. Nitrogen Fixation in Cyanobacteria. In eLS; John Wiley & Sons, Ltd: Chichester, UK, 2015; pp 1– 9. DOI: 10.1002/9780470015902.a0021159.pub2 .There is no corresponding record for this reference.
- 100Poirier, Y.; Antonenkov, V. D.; Glumoff, T.; Hiltunen, J. K. Peroxisomal β-oxidation─A Metabolic Pathway with Multiple Functions. Biochimica et Biophysica Acta (BBA) - Molecular Cell Research 2006, 1763 (12), 1413– 1426, DOI: 10.1016/j.bbamcr.2006.08.034100https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xhtlakur%252FK&md5=6d7a975e32e574ff04ead583cf4ff881Peroxisomal β-oxidation-A metabolic pathway with multiple functionsPoirier, Yves; Antonenkov, Vasily D.; Glumoff, Tuomo; Hiltunen, J. KalervoBiochimica et Biophysica Acta, Molecular Cell Research (2006), 1763 (12), 1413-1426CODEN: BBAMCO; ISSN:0167-4889. (Elsevier Ltd.)A review. Fatty acid degrdn. in most organisms occurs primarily via the β-oxidn. cycle. In mammals, β-oxidn. occurs in both mitochondria and peroxisomes, whereas plants and most fungi harbor the β-oxidn. cycle only in the peroxisomes. Although several of the enzymes participating in this pathway in both organelles are similar, some distinct physiol. roles have been uncovered. Recent advances in the structural elucidation of numerous mammalian and yeast enzymes involved in β-oxidn. have shed light on the basis of the substrate specificity for several of them. Of particular interest is the structural organization and function of the type 1 and 2 multifunctional enzyme (MFE-1 and MFE-2), two enzymes evolutionarily distant yet catalyzing the same overall enzymic reactions but via opposite stereochem. New data on the physiol. roles of the various enzymes participating in β-oxidn. have been gathered through the anal. of knockout mutants in plants, yeast and animals, as well as by the use of polyhydroxyalkanoate synthesis from β-oxidn. intermediates as a tool to study carbon flux through the pathway. In plants, both forward and reverse genetics performed on the model plant Arabidopsis thaliana have revealed novel roles for β-oxidn. in the germination process that is independent of the generation of carbohydrates for growth, as well as in embryo and flower development, and the generation of the phytohormone indole-3-acetic acid and the signal mol. jasmonic acid.
- 101Murat, D.; Byrne, M.; Komeili, A. Cell Biology of Prokaryotic Organelles. Cold Spring Harb. Perspect. Biol. 2010, 2 (10), a000422, DOI: 10.1101/cshperspect.a000422There is no corresponding record for this reference.
- 102Nott, T. J.; Craggs, T. D.; Baldwin, A. J. Membraneless Organelles Can Melt Nucleic Acid Duplexes and Act as Biomolecular Filters. Nat. Chem. 2016, 8 (6), 569– 575, DOI: 10.1038/nchem.2519102https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XotV2nt70%253D&md5=0224be564579d21e8d4dd9c8bbc42fcdMembraneless organelles can melt nucleic acid duplexes and act as biomolecular filtersNott, Timothy J.; Craggs, Timothy D.; Baldwin, Andrew J.Nature Chemistry (2016), 8 (6), 569-575CODEN: NCAHBB; ISSN:1755-4330. (Nature Publishing Group)Membraneless organelles are cellular compartments made from drops of liq. protein inside a cell. These compartments assemble via the phase sepn. of disordered regions of proteins in response to changes in the cellular environment and the cell cycle. Here we demonstrate that the solvent environment within the interior of these cellular bodies behaves more like an org. solvent than like water. One of the most-stable biol. structures known, the DNA double helix, can be melted once inside the liq. droplet, and simultaneously structures formed from regulatory single-stranded nucleic acids are stabilized. Moreover, proteins are shown to have a wide range of absorption or exclusion from these bodies, and can act as importers for otherwise-excluded nucleic acids, which suggests the existence of a protein-mediated trafficking system. A common strategy in org. chem. is to utilize different solvents to influence the behavior of mols. and reactions. These results reveal that cells have also evolved this capability by exploiting the interiors of membraneless organelles.
- 103Lyon, A. S.; Peeples, W. B.; Rosen, M. K. A Framework for Understanding the Functions of Biomolecular Condensates across Scales. Nat. Rev. Mol. Cell Biol. 2021, 22 (3), 215– 235, DOI: 10.1038/s41580-020-00303-z103https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXit1yht7rP&md5=36e48922d2d86afd61d54380ef468ab5A framework for understanding the functions of biomolecular condensates across scalesLyon, Andrew S.; Peeples, William B.; Rosen, Michael K.Nature Reviews Molecular Cell Biology (2021), 22 (3), 215-235CODEN: NRMCBP; ISSN:1471-0072. (Nature Research)Abstr.: Biomol. condensates are found throughout eukaryotic cells, including in the nucleus, in the cytoplasm and on membranes. They are also implicated in a wide range of cellular functions, organizing mols. that act in processes ranging from RNA metab. to signalling to gene regulation. Early work in the field focused on identifying condensates and understanding how their phys. properties and regulation arise from mol. constituents. Recent years have brought a focus on understanding condensate functions. Studies have revealed functions that span different length scales: from mol. (modulating the rates of chem. reactions) to mesoscale (organizing large structures within cells) to cellular (facilitating localization of cellular materials and homeostatic responses). In this Roadmap, we discuss representative examples of biochem. and cellular functions of biomol. condensates from the recent literature and organize these functions into a series of non-exclusive classes across the different length scales. We conclude with a discussion of areas of current interest and challenges in the field, and thoughts about how progress may be made to further our understanding of the widespread roles of condensates in cell biol.
- 104Protter, D. S. W.; Parker, R. Principles and Properties of Stress Granules. Trends Cell Biol. 2016, 26 (9), 668– 679, DOI: 10.1016/j.tcb.2016.05.004104https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XosVSnsLw%253D&md5=799c56059a5fcd164ddb5fb50a9348cbPrinciples and Properties of Stress GranulesProtter, David S. W.; Parker, RoyTrends in Cell Biology (2016), 26 (9), 668-679CODEN: TCBIEK; ISSN:0962-8924. (Elsevier Ltd.)A review. Stress granules are assemblies of untranslating messenger ribonucleoproteins (mRNPs) that form from mRNAs stalled in translation initiation. Stress granules form through interactions between mRNA-binding proteins that link together populations of mRNPs. Interactions promoting stress granule formation include conventional protein-protein interactions as well as interactions involving intrinsically disordered regions (IDRs) of proteins. Assembly and disassembly of stress granules are modulated by various post-translational modifications as well as numerous ATP-dependent RNP or protein remodeling complexes, illustrating that stress granules represent an active liq. wherein energy input maintains their dynamic state. Stress granule formation modulates the stress response, viral infection, and signaling pathways. Persistent or aberrant stress granule formation contributes to neurodegenerative disease and some cancers.
- 105Roger, A. J.; Muñoz-Gómez, S. A.; Kamikawa, R. The Origin and Diversification of Mitochondria. Curr. Biol. 2017, 27 (21), R1177– R1192, DOI: 10.1016/j.cub.2017.09.015105https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXhslygsL%252FO&md5=4611fac5e6db78c3eb8bb4bc313ceb32The Origin and Diversification of MitochondriaRoger, Andrew J.; Munoz-Gomez, Sergio A.; Kamikawa, RyomaCurrent Biology (2017), 27 (21), R1177-R1192CODEN: CUBLE2; ISSN:0960-9822. (Cell Press)A review. Mitochondria are best known for their role in the generation of ATP by aerobic respiration. Yet, research in the past half century has shown that they perform a much larger suite of functions and that these functions can vary substantially among diverse eukaryotic lineages. Despite this diversity, all mitochondria derive from a common ancestral organelle that originated from the integration of an endosymbiotic alphaproteobacterium into a host cell related to Asgard Archaea. The transition from endosymbiotic bacterium to permanent organelle entailed a massive no. of evolutionary changes including the origins of hundreds of new genes and a protein import system, insertion of membrane transporters, integration of metab. and reprodn., genome redn., endosymbiotic gene transfer, lateral gene transfer and the retargeting of proteins. These changes occurred incrementally as the endosymbiont and the host became integrated. Although many insights into this transition have been gained, controversy persists regarding the nature of the original endosymbiont, its initial interactions with the host and the timing of its integration relative to the origin of other features of eukaryote cells. Since the establishment of the organelle, proteins have been gained, lost, transferred and retargeted as mitochondria have specialized into the spectrum of functional types seen across the eukaryotic tree of life.
- 106Mehta, A. P.; Supekova, L.; Chen, J.-H.; Pestonjamasp, K.; Webster, P.; Ko, Y.; Henderson, S. C.; McDermott, G.; Supek, F.; Schultz, P. G. Engineering Yeast Endosymbionts as a Step toward the Evolution of Mitochondria. Proc. Natl. Acad. Sci. U. S. A. 2018, 115 (46), 11796– 11801, DOI: 10.1073/pnas.1813143115106https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXit1Sgt7fE&md5=fb74ba83e9dc4402b3d436758bafb90fEngineering yeast endosymbionts as a step toward the evolution of mitochondriaMehta, Angad P.; Supekova, Lubica; Chen, Jian-Hua; Pestonjamasp, Kersi; Webster, Paul; Ko, Yeonjin; Henderson, Scott C.; McDermott, Gerry; Supek, Frantisek; Schultz, Peter G.Proceedings of the National Academy of Sciences of the United States of America (2018), 115 (46), 11796-11801CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)It has been hypothesized that mitochondria evolved from a bacterial ancestor that initially became established in an archaeal host cell as an endosymbiont. Here we model this first stage of mitochondrial evolution by engineering endosymbiosis between Escherichia coli and Saccharomyces cerevisiae. An ADP/ATP translocase-expressing E. coli provided ATP to a respiration-deficient cox2 yeast mutant and enabled growth of a yeast-E. coli chimera on a nonfermentable carbon source. In a reciprocal fashion, yeast provided thiamin to an endosymbiotic E. coli thiamin auxotroph. Expression of several SNARE-like proteins in E. coli was also required, likely to block lysosomal degrdn. of intracellular bacteria. This chimeric system was stable for more than 40 doublings, and GFP-expressing E. coli endosymbionts could be obsd. in the yeast by fluorescence microscopy and X-ray tomog. This readily manipulated system should allow exptl. delineation of host-endosymbiont adaptations that occurred during evolution of the current, highly reduced mitochondrial genome.
- 107Headen, D. M.; Aubry, G.; Lu, H.; García, A. J. Microfluidic-Based Generation of Size-Controlled, Biofunctionalized Synthetic Polymer Microgels for Cell Encapsulation. Adv. Mater. 2014, 26 (19), 3003– 3008, DOI: 10.1002/adma.201304880107https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXktVWmtL8%253D&md5=9f60d129d74f04352d95959a640285f0Microfluidic-based generation of size-controlled, biofunctionalized synthetic polymer microgels for cell encapsulationHeaden, Devon M.; Aubry, Guillaume; Lu, Hang; Garcia, Andres J.Advanced Materials (Weinheim, Germany) (2014), 26 (19), 3003-3008CODEN: ADVMEW; ISSN:0935-9648. (Wiley-VCH Verlag GmbH & Co. KGaA)The high potential of synthetic hydrogel microencapsulation for cell and protein therapeutics has been limited by the lack of synthetic polymer systems with tunable capsule size, cytocompatible crosslinking reactions, rapid crosslinking rates, adequate biomol. permeability, and ease of functionalization with bioactive mols. (e.g., adhesive peptides). Using a synthetic hydrogel system with tunable network and crosslinking characteristics and a microfluidics encapsulation platform, we have created an integrated and robust strategy for microencapsulation of cells in which we can control capsule size and local cellular microenvironment. Addnl., microgel network structure can be tuned to optimize permeability of the capsule to mols. of various sizes. We have demonstrated, proof of concept with two different clin. relevant human cell types, but the versatility of this strategy will allow it to be tailored to fit diverse engineering applications.
- 108Torre, P.; Keating, C. D.; Mansy, S. S. Multiphase Water-in-Oil Emulsion Droplets for Cell-Free Transcription-Translation. Langmuir 2014, 30 (20), 5695– 5699, DOI: 10.1021/la404146g108https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXns1Cns7w%253D&md5=d1aa45d707f57c13621a12046cc04eb0Multiphase water-in-oil emulsion droplets for cell-free transcription-translationTorre, Paola; Keating, Christine D.; Mansy, Sheref S.Langmuir (2014), 30 (20), 5695-5699CODEN: LANGD5; ISSN:0743-7463. (American Chemical Society)The construction of genetically encoded cellular mimics in compartments contg. organized synthetic cytosols is desirable for the development of artificial cells. Phase sepd. aq. domains were placed within water-in-oil emulsion droplets in a manner compatible with transcription and translation machinery. Aq. two-phase and three-phase systems (ATPS and A3PS) were assembled with dextran, poly(ethylene glycol), and Ficoll. Aq. two-phase systems were capable of supporting the cell-free expression of protein within water droplets, whereas the aq. three-phase-based system did not give rise to detectable protein synthesis. The expressed protein preferentially partitioned to the dextran-enriched phase. The system could serve as a foundation for building cellular mimics with liq. organelles.
- 109Zhang, Y.; Kojima, T.; Kim, G.-A.; McNerney, M. P.; Takayama, S.; Styczynski, M. P. Protocell Arrays for Simultaneous Detection of Diverse Analytes. Nat. Commun. 2021, 12 (1), 5724, DOI: 10.1038/s41467-021-25989-3109https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXitFCgtLjO&md5=310fc2fe6fcb267fbcf136e7d1951810Protocell arrays for simultaneous detection of diverse analytesZhang, Yan; Kojima, Taisuke; Kim, Ge-Ah; McNerney, Monica P.; Takayama, Shuichi; Styczynski, Mark P.Nature Communications (2021), 12 (1), 5724CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)Simultaneous detection of multiple analytes from a single sample (multiplexing), particularly when done at the point of need, can guide complex decision-making without increasing the required sample vol. or cost per test. Despite recent advances, multiplexed analyte sensing still typically faces the crit. limitation of measuring only one type of mol. (e.g., small mols. or nucleic acids) per assay platform. Here, we address this bottleneck with a customizable platform that integrates cell-free expression (CFE) with a polymer-based aq. two-phase system (ATPS), producing membrane-less protocells contg. transcription and translation machinery used for detection. We show that multiple protocells, each performing a distinct sensing reaction, can be arrayed in the same microwell to detect chem. diverse targets from the same sample. Furthermore, these protocell arrays are compatible with human biofluids, maintain function after lyophilization and rehydration, and can produce visually interpretable readouts, illustrating this platforms potential as a minimal-equipment, field-deployable, multi-analyte detection tool.
- 110Allen, M. E.; Hindley, J. W.; Baxani, D. K.; Ces, O.; Elani, Y. Hydrogels as Functional Components in Artificial Cell Systems. Nat. Rev. Chem. 2022, 6 (8), 562– 578, DOI: 10.1038/s41570-022-00404-7110https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhvFGnsbbK&md5=a18a79ffa095b7ee78a973929cdd92a2Hydrogels as functional components in artificial cell systemsAllen, Matthew E.; Hindley, James W.; Baxani, Divesh K.; Ces, Oscar; Elani, YuvalNature Reviews Chemistry (2022), 6 (8), 562-578CODEN: NRCAF7; ISSN:2397-3358. (Nature Portfolio)A review. Recent years have seen substantial efforts aimed at constructing artificial cells from various mol. components with the aim of mimicking the processes, behaviors and architectures found in biol. systems. Artificial cell development ultimately aims to produce model constructs that progress our understanding of biol., as well as forming the basis for functional bio-inspired devices that can be used in fields such as therapeutic delivery, biosensing, cell therapy and bioremediation. Typically, artificial cells rely on a bilayer membrane chassis and have fluid aq. interiors to mimic biol. cells. However, a desire to more accurately replicate the gel-like properties of intracellular and extracellular biol. environments has driven increasing efforts to build cell mimics based on hydrogels. This has enabled researchers to exploit some of the unique functional properties of hydrogels that have seen them deployed in fields such as tissue engineering, biomaterials and drug delivery. In this Review, we explore how hydrogels can be leveraged in the context of artificial cell development. We also discuss how hydrogels can potentially be incorporated within the next generation of artificial cells to engineer improved biol. mimics and functional microsystems.
- 111Has, C.; Sunthar, P. A Comprehensive Review on Recent Preparation Techniques of Liposomes. J. Liposome Res. 2020, 30 (4), 336– 365, DOI: 10.1080/08982104.2019.1668010111https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhvVGntL3N&md5=8a224f78b2356da1539e1a35bad7b076A comprehensive review on recent preparation techniques of liposomesHas, C.; Sunthar, P.Journal of Liposome Research (2020), 30 (4), 336-365CODEN: JLREE7; ISSN:0898-2104. (Taylor & Francis Ltd.)A review Liposomes (or lipid vesicles) are a versatile platform as carriers for the delivery of the drugs and other macromols. into human and animal bodies. Though the method of using liposomes has been known since 1960s, and major developments and commercialization of liposomal formulations took place in the late nineties (or early part of this century), newer methods of liposome synthesis and drug encapsulation continue to be an active area of research. With the developments in related fields, such as electrohydrodynamics and microfluidics, and a growing understanding of the mechanisms of lipid assembly from colloidal and intermol. forces, liposome prepn. techniques have been enriched and more predictable than before. This has led to better methods that can also scale at an industrial prodn. level. In this review, we present several novel methods that were introduced over the last decade and compare their advantages over conventional methods. Researchers beginning to explore liposomal formulations will find this resource useful to give an overall direction for an appropriate choice of method. Where possible, we have also provided the known mechanisms behind the prepn. methods.
- 112Luisi, P. L.; Allegretti, M.; Pereira de Souza, T.; Steiniger, F.; Fahr, A.; Stano, P. Spontaneous Protein Crowding in Liposomes: A New Vista for the Origin of Cellular Metabolism. Chembiochem 2010, 11 (14), 1989– 1992, DOI: 10.1002/cbic.201000381There is no corresponding record for this reference.
- 113Michelon, M.; Huang, Y.; de la Torre, L. G.; Weitz, D. A.; Cunha, R. L. Single-Step Microfluidic Production of W/O/W Double Emulsions as Templates for β-Carotene-Loaded Giant Liposomes Formation. Chem. Eng. J. 2019, 366, 27– 32, DOI: 10.1016/j.cej.2019.02.021113https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXjtFOns7Y%253D&md5=392e7b1f109abc07e1a4137388de4360Single-step microfluidic production of W/O/W double emulsions as templates for β-carotene-loaded giant liposomes formationMichelon, Mariano; Huang, Yuting; de la Torre, Lucimara Gaziola; Weitz, David A.; Cunha, Rosiane LopesChemical Engineering Journal (Amsterdam, Netherlands) (2019), 366 (), 27-32CODEN: CMEJAJ; ISSN:1385-8947. (Elsevier B.V.)We demonstrated the microfluidic prodn. of W/O/W double emulsion droplets aiming formation of β-carotene-incorporated giant liposomes for food and/or pharmaceutical applications. For this purpose, glass-capillary microfluidic devices were fabricated to create a truly three-dimensional flow aiming prodn. of giant unilamellar liposomes by solvent evapn. process after W/O/W double emulsion droplet templates formation. A great challenge of microfluidic prodn. of monodisperse and stable W/O/W double emulsion templates for this proposal is the replacement of org. solvents potentially toxic for phospholipids dissoln. Besides, the high cost of several semi-synthetic phospholipids commonly used for giant liposome formation remains as a major technol. challenge to be overcome. Thus, β-carotene-incorporated giant liposomes were generated using biocompatible solvents with low toxic potential (Et acetate and pentane) and non-purified soybean lecithin - a food-grade phospholipid mixt. with low cost - by dewetting and evapn. of the solvents forming the oily intermediate phase of W/O/W double emulsion droplet templates. Our results showed monodisperse β-carotene-loaded giant liposomes with diam. ranging between 100 μm and 180 μm and a stability of approx. 7 days. In this way, a single-step microfluidic process with highly accurate control of size distribution was developed. This microfluidic process proposed is potentially useful for a broad range of applications in protection and delivery of active compds.
- 114Elani, Y.; Trantidou, T.; Wylie, D.; Dekker, L.; Polizzi, K.; Law, R. V.; Ces, O. Constructing Vesicle-Based Artificial Cells with Embedded Living Cells as Organelle-like Modules. Sci. Rep. 2018, 8 (1), 4564, DOI: 10.1038/s41598-018-22263-3114https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1Mnht1yisw%253D%253D&md5=8e62e29719807e28d859d2c4d4edfe5bConstructing vesicle-based artificial cells with embedded living cells as organelle-like modulesElani Yuval; Trantidou Tatiana; Wylie Douglas; Law Robert V; Ces Oscar; Elani Yuval; Wylie Douglas; Ces Oscar; Dekker Linda; Polizzi KarenScientific reports (2018), 8 (1), 4564 ISSN:.There is increasing interest in constructing artificial cells by functionalising lipid vesicles with biological and synthetic machinery. Due to their reduced complexity and lack of evolved biochemical pathways, the capabilities of artificial cells are limited in comparison to their biological counterparts. We show that encapsulating living cells in vesicles provides a means for artificial cells to leverage cellular biochemistry, with the encapsulated cells serving organelle-like functions as living modules inside a larger synthetic cell assembly. Using microfluidic technologies to construct such hybrid cellular bionic systems, we demonstrate that the vesicle host and the encapsulated cell operate in concert. The external architecture of the vesicle shields the cell from toxic surroundings, while the cell acts as a bioreactor module that processes encapsulated feedstock which is further processed by a synthetic enzymatic metabolism co-encapsulated in the vesicle.
- 115Tayeb, H. H.; Sainsbury, F. Nanoemulsions in Drug Delivery: Formulation to Medical Application. Nanomedicine 2018, 13 (19), 2507– 2525, DOI: 10.2217/nnm-2018-0088115https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitV2isL7F&md5=10a78be18038f4a9b8a01afa21fe96feNanoemulsions in drug delivery: formulation to medical applicationTayeb, Hossam H.; Sainsbury, FrankNanomedicine (London, United Kingdom) (2018), 13 (19), 2507-2525CODEN: NLUKAC; ISSN:1748-6963. (Future Medicine Ltd.)Nanoscale oil-in-water emulsions (NEs), heterogeneous systems of two immiscible liqs. stabilized by emulsifiers or surfactants, show great potential in medical applications because of their attractive characteristics for drug delivery. NEs have been explored as therapeutic carriers for hydrophobic compds. via various routes of administration. NEs provide opportunities to improve drug delivery via alternative administration routes. However, deep understanding of the NE manufg. and functionalization fundamentals, and how they relate to the choice of administration route and pharmacol. profile is still needed to ease the clin. translation of NEs. Here, we review the diversity of medical applications for NEs and how that governs their formulation, route of administration, and the emergence of increasing sophistication in NE design for specific application.
- 116Vogele, K.; Frank, T.; Gasser, L.; Goetzfried, M. A.; Hackl, M. W.; Sieber, S. A.; Simmel, F. C.; Pirzer, T. Towards Synthetic Cells Using Peptide-Based Reaction Compartments. Nat. Commun. 2018, 9 (1), 3862, DOI: 10.1038/s41467-018-06379-8116https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3czhsV2jsA%253D%253D&md5=7ee8f4968fc15a261b9d2deb5b7d789aTowards synthetic cells using peptide-based reaction compartmentsVogele Kilian; Frank Thomas; Gasser Lukas; Goetzfried Marisa A; Simmel Friedrich C; Pirzer Tobias; Hackl Mathias W; Sieber Stephan A; Simmel Friedrich CNature communications (2018), 9 (1), 3862 ISSN:.Membrane compartmentalization and growth are central aspects of living cells, and are thus encoded in every cell's genome. For the creation of artificial cellular systems, genetic information and production of membrane building blocks will need to be coupled in a similar manner. However, natural biochemical reaction networks and membrane building blocks are notoriously difficult to implement in vitro. Here, we utilized amphiphilic elastin-like peptides (ELP) to create self-assembled vesicular structures of about 200 nm diameter. In order to genetically encode the growth of these vesicles, we encapsulate a cell-free transcription-translation system together with the DNA template inside the peptide vesicles. We show in vesiculo production of a functioning fluorescent RNA aptamer and a fluorescent protein. Furthermore, we implement in situ expression of the membrane peptide itself and finally demonstrate autonomous vesicle growth due to the incorporation of this ELP into the membrane.
- 117Mushnoori, S.; Lu, C. Y.; Schmidt, K.; Zang, E.; Dutt, M. Peptide-Based Vesicles and Droplets: A Review. J. Phys.: Condens. Matter 2020, 33 (5), 053002, DOI: 10.1088/1361-648X/abb995There is no corresponding record for this reference.
- 118Sharma, B.; Ma, Y.; Hiraki, H. L.; Baker, B. M.; Ferguson, A. L.; Liu, A. P. Facile Formation of Giant Elastin-like Polypeptide Vesicles as Synthetic Cells. Chem. Commun. 2021, 57 (97), 13202– 13205, DOI: 10.1039/D1CC05579H118https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXisFWlu7nI&md5=210d236dd96e6fcbdf19e6c8f2042f95Facile formation of giant elastin-like polypeptide vesicles as synthetic cellsSharma, Bineet; Ma, Yutao; Hiraki, Harrison L.; Baker, Brendon M.; Ferguson, Andrew L.; Liu, Allen P.Chemical Communications (Cambridge, United Kingdom) (2021), 57 (97), 13202-13205CODEN: CHCOFS; ISSN:1359-7345. (Royal Society of Chemistry)We demonstrate the facile and robust generation of giant peptide vesicles by using an emulsion transfer method. These robust vesicles can sustain chem. and phys. stresses. The peptide vesicles can host cell-free expression reactions by encapsulating essential ingredients. We show the incorporation of another cell-free expressed elastin-like polypeptide into the existing membrane of the peptide vesicles.
- 119Vieregg, J. R.; Lueckheide, M.; Marciel, A. B.; Leon, L.; Bologna, A. J.; Rivera, J. R.; Tirrell, M. V. Oligonucleotide-Peptide Complexes: Phase Control by Hybridization. J. Am. Chem. Soc. 2018, 140 (5), 1632– 1638, DOI: 10.1021/jacs.7b03567119https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXlvVChsQ%253D%253D&md5=7c78b1f95043da89c34c10a3d3543529Oligonucleotide-peptide complexes: Phase control by hybridizationVieregg, Jeffrey R.; Lueckheide, Michael; Marciel, Amanda B.; Leon, Lorraine; Bologna, Alex J.; Rivera, Josean Reyes; Tirrell, Matthew V.Journal of the American Chemical Society (2018), 140 (5), 1632-1638CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)When oppositely charged polymers are mixed, counterion release drives phase sepn.; understanding this process is a key unsolved problem in polymer science and biophys. chem., particularly for nucleic acids, polyanions whose biol. functions are intimately related to their high charge d. In the cell, complexation by basic proteins condenses DNA into chromatin, and membraneless organelles formed by liq.-liq. phase sepn. of RNA and proteins perform vital functions and have been linked to disease. Electrostatic interactions are also the primary method used for assembly of nanoparticles to deliver therapeutic nucleic acids into cells. This work describes complexation expts. with oligonucleotides and cationic peptides spanning a wide range of polymer lengths, concns., and structures, including RNA and methylphosphonate backbones. We find that the phase of the complexes is controlled by the hybridization state of the nucleic acid, with double-stranded nucleic acids forming solid ppts. while single-stranded oligonucleotides form liq. coacervates, apparently due to their lower charge d. Adding salt "melts" ppts. into coacervates, and oligonucleotides in coacervates remain competent for sequence-specific hybridization and phase change, suggesting the possibility of environmentally responsive complexes and nanoparticles for therapeutic or sensing applications.
- 120Fraccia, T. P.; Jia, T. Z. Liquid Crystal Coacervates Composed of Short Double-Stranded DNA and Cationic Peptides. ACS Nano 2020, 14 (11), 15071– 15082, DOI: 10.1021/acsnano.0c05083120https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhs12rtr7L&md5=04619533c8c1cacce2a6b7f7dd591b6eLiquid Crystal Coacervates Composed of Short Double-Stranded DNA and Cationic PeptidesFraccia, Tommaso P.; Jia, Tony Z.ACS Nano (2020), 14 (11), 15071-15082CODEN: ANCAC3; ISSN:1936-0851. (American Chemical Society)Phase sepn. of nucleic acids and proteins is a ubiquitous phenomenon regulating subcellular compartment structure and function. While complex coacervation of flexible single-stranded nucleic acids is broadly investigated, coacervation of double-stranded DNA (dsDNA) is less studied because of its propensity to generate solid ppts. Here, we reverse this perspective by showing that short dsDNA and poly-L-lysine coacervates can escape pptn. while displaying a surprisingly complex phase diagram, including the full set of liq. crystal (LC) mesophases obsd. to date in bulk dsDNA. Short dsDNA supramol. aggregation and packing in the dense coacervate phase are the main parameters regulating the global LC-coacervate phase behavior. LC-coacervate structure was characterized upon variations in temp. and monovalent salt, DNA, and peptide concns., which allow continuous reversible transitions between all accessible phases. A deeper understanding of LC-coacervates can gain insights to decipher structures and phase transition mechanisms within biomol. condensates, to design stimuli-responsive multiphase synthetic compartments with different degrees of order and to exploit self-assembly driven cooperative prebiotic evolution of nucleic acids and peptides.
- 121Aumiller, W. M., Jr; Pir Cakmak, F.; Davis, B. W.; Keating, C. D. RNA-Based Coacervates as a Model for Membraneless Organelles: Formation, Properties, and Interfacial Liposome Assembly. Langmuir 2016, 32 (39), 10042– 10053, DOI: 10.1021/acs.langmuir.6b02499121https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsVKrtrnO&md5=1298cfe8fb86980343543917b9831be1RNA-Based Coacervates as a Model for Membraneless Organelles: Formation, Properties, and Interfacial Liposome AssemblyAumiller, William M.; Pir Cakmak, Fatma; Davis, Bradley W.; Keating, Christine D.Langmuir (2016), 32 (39), 10042-10053CODEN: LANGD5; ISSN:0743-7463. (American Chemical Society)Liq.-liq. phase sepn. is responsible for the formation of P granules, nucleoli, and other membraneless subcellular organelles composed of RNA and proteins. Efforts to understand the phys. basis of liq. organelle formation have thus far focused on intrinsically disordered proteins (IDPs) as major components that dictate occurrence and properties. Here, the authors show that complex coacervates composed of low complexity RNA [polyuridylic acid, poly(U)] and short polyamines (spermine and spermidine) share many features of IDP-based coacervates. Poly(U)/polyamine coacervates compartmentalized biomols. (peptides, oligonucleotides) in a sequence- and length- dependent manner. These solutes retained mobility within the coacervate droplets, as demonstrated by rapid recovery from photobleaching. Coacervation was reversible with changes in soln. temp. due to changes in the poly(U) structure that impacted its interactions with polyamines. The authors further demonstrated that lipid vesicles assembled at the droplet interface without impeding RNA entry/egress. These vesicles remained intact at the interface and could be released upon temp.-induced droplet dissoln.
- 122Rimoli, M. G.; Rabaioli, M. R.; Melisi, D.; Curcio, A.; Mondello, S.; Mirabelli, R.; Abignente, E. Synthetic Zeolites as a New Tool for Drug Delivery. J. Biomed. Mater. Res., Part A 2008, 87 (1), 156– 164, DOI: 10.1002/jbm.a.31763122https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhtFCqsL7K&md5=9d159c5f96d5a848b766aa5e118a087aSynthetic zeolites as a new tool for drug deliveryRimoli, Maria G.; Rabaioli, Maria R.; Melisi, Daniela; Curcio, Annalisa; Mondello, Sandro; Mirabelli, Rosella; Abignente, EnricoJournal of Biomedical Materials Research, Part A (2008), 87A (1), 156-164CODEN: JBMRCH; ISSN:1549-3296. (John Wiley & Sons, Inc.)Synthetic zeolites were studied in order to investigate their ability to encapsulate and to release drugs. In particular, a zeolite X and a zeolitic product obtained from a cocrystn. of zeolite X and zeolite A were examd. These materials were characterized by chem. analyses (ICP-AES), x-ray diffraction, nitrogen adsorption isotherm, SEM, laser diffraction, and IR spectroscopy. Since ketoprofen was chosen as a model drug for the formulation of controlled-release dosage forms, it was encapsulated into these two types of synthetic zeolites by a soaking procedure. Drug-loaded matrixes were then characterized for entrapped drug amt. and thermogravimetric behavior. In both types of activated zeolites, the total amt. of ketoprofen (800 mg) was encapsulated in 2 g of matrix. By using HPLC measurements, ketoprofen release studies were done at different pH conditions so as to mimic gastrointestinal fluids. The absence of release in acid conditions and a double phased release, at two different pH values (5 and 6.8), suggest that after activation these materials offer good potential for a modified release delivery system of ketoprofen.
- 123Hyman, A. A.; Weber, C. A.; Jülicher, F. Liquid-Liquid Phase Separation in Biology. Annu. Rev. Cell Dev. Biol. 2014, 30, 39– 58, DOI: 10.1146/annurev-cellbio-100913-013325123https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXitVeit7vL&md5=a4d83a5d473be634e6a4d9bdbcc6e63aLiquid-liquid phase separation in biologyHyman, Anthony A.; Weber, Christoph A.; Juelicher, FrankAnnual Review of Cell and Developmental Biology (2014), 30 (), 39-58CODEN: ARDBF8; ISSN:1081-0706. (Annual Reviews)A review. Cells organize many of their biochem. reactions in non-membrane compartments. Recent evidence showed that many of these compartments are liqs. that form by phase sepn. from the cytoplasm. Here the basic phys. concepts necessary to understand the consequences of liq.-like states for biol. functions are discussed.
- 124Junge, F.; Haberstock, S.; Roos, C.; Stefer, S.; Proverbio, D.; Dötsch, V.; Bernhard, F. Advances in Cell-Free Protein Synthesis for the Functional and Structural Analysis of Membrane Proteins. N. Biotechnol. 2011, 28 (3), 262– 271, DOI: 10.1016/j.nbt.2010.07.002There is no corresponding record for this reference.
- 125Lee, K. Y.; Park, S.-J.; Lee, K. A.; Kim, S.-H.; Kim, H.; Meroz, Y.; Mahadevan, L.; Jung, K.-H.; Ahn, T. K.; Parker, K. K.; Shin, K. Photosynthetic Artificial Organelles Sustain and Control ATP-Dependent Reactions in a Protocellular System. Nat. Biotechnol. 2018, 36 (6), 530– 535, DOI: 10.1038/nbt.4140125https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhtVWktrzM&md5=38cc9b7aa604f285b5d84c793c9e26cfPhotosynthetic artificial organelles sustain and control ATP-dependent reactions in a protocellular systemLee, Keel Yong; Park, Sung-Jin; Lee, Keon Ah; Kim, Se-Hwan; Kim, Heeyeon; Meroz, Yasmine; Mahadevan, L.; Jung, Kwang-Hwan; Ahn, Tae Kyu; Parker, Kevin Kit; Shin, KwanwooNature Biotechnology (2018), 36 (6), 530-535CODEN: NABIF9; ISSN:1087-0156. (Nature Research)Inside cells, complex metabolic reactions are distributed across the modular compartments of organelles. Reactions in organelles have been recapitulated in vitro by reconstituting functional protein machineries into membrane systems. However, maintaining and controlling these reactions is challenging. Here we designed, built, and tested a switchable, light-harvesting organelle that provides both a sustainable energy source and a means of directing intravesicular reactions. An ATP (ATP) synthase and two photoconverters (plant-derived photosystem II and bacteria-derived proteorhodopsin) enable ATP synthesis. Independent optical activation of the two photoconverters allows dynamic control of ATP synthesis: red light facilitates and green light impedes ATP synthesis. We encapsulated the photosynthetic organelles in a giant vesicle to form a protocellular system and demonstrated optical control of two ATP-dependent reactions, carbon fixation and actin polymn., with the latter altering outer vesicle morphol. Switchable photosynthetic organelles may enable the development of biomimetic vesicle systems with regulatory networks that exhibit homeostasis and complex cellular behaviors.
- 126Jewett, M. C.; Swartz, J. R. Mimicking the Escherichia Coli Cytoplasmic Environment Activates Long-Lived and Efficient Cell-Free Protein Synthesis. Biotechnol. Bioeng. 2004, 86 (1), 19– 26, DOI: 10.1002/bit.20026126https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXis1aksrY%253D&md5=e8d45540857fdb7b7b269e2704afb08cMimicking the Escherichia coli cytoplasmic environment activates long-lived and efficient cell-free protein synthesisJewett, Michael C.; Swartz, James R.Biotechnology and Bioengineering (2004), 86 (1), 19-26CODEN: BIBIAU; ISSN:0006-3592. (John Wiley & Sons, Inc.)Cell-free translation systems generally utilize high-energy phosphate compds. to regenerate the ATP necessary to drive protein synthesis. This hampers the widespread use and practical implementation of this technol. in a batch format due to expensive reagent costs; the accumulation of inhibitory byproducts, such as phosphate; and pH change. To address these problems, a cell-free protein synthesis system has been engineered that is capable of using pyruvate as an energy source to produce high yields of protein. The "Cytomim" system, synthesizes chloramphenicol acetyl-transferase (CAT) for up to 6 h in a batch reaction to yield 700 μg/mL of protein. By more closely replicating the physiol. conditions of the cytoplasm of Escherichia coli, the Cytomim system provides a stable energy supply for protein expression without phosphate accumulation, pH change, exogenous enzyme addn., or the need for expensive high-energy phosphate compds.
- 127Caschera, F.; Noireaux, V. Synthesis of 2.3 Mg/mL of Protein with an All Escherichia Coli Cell-Free Transcription-Translation System. Biochimie 2014, 99, 162– 168, DOI: 10.1016/j.biochi.2013.11.025127https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhvFOhtrjM&md5=7d951478b25a8eaaba5a48e3ca59522cSynthesis of 2.3 mg/ml of protein with an all Escherichia coli cell-free transcription-translation systemCaschera, Filippo; Noireaux, VincentBiochimie (2014), 99 (), 162-168CODEN: BICMBE; ISSN:0300-9084. (Elsevier Masson SAS)Cell-free protein synthesis is becoming a useful technique for synthetic biol. As more applications are developed, the demand for novel and more powerful in vitro expression systems is increasing. In this work, an all Escherichia coli cell-free system, that uses the endogenous transcription and translation mol. machineries, is optimized to synthesize up to 2.3 mg/mL of a reporter protein in batch mode reactions. A new metab. based on maltose allows recycling of inorg. phosphate through its incorporation into newly available glucose mols., which are processed through the glycolytic pathway to produce more ATP. As a result, the ATP regeneration is more efficient and cell-free protein synthesis lasts up to 10 h. Using a com. E. coli strain, we show for the first time that more than 2 mg/mL of protein can be synthesized in run-off cell-free transcription-translation reactions by optimizing the energy regeneration and waste products recycling. This work suggests that endogenous enzymes present in the cytoplasmic ext. can be used to implement new metabolic pathways for increasing protein yields. This system is the new basis of a cell-free gene expression platform used to construct and to characterize complex biochem. processes in vitro such as gene circuits.
- 128Garamella, J.; Marshall, R.; Rustad, M.; Noireaux, V. The All E. Coli TX-TL Toolbox 2.0: A Platform for Cell-Free Synthetic Biology. ACS Synth. Biol. 2016, 5 (4), 344– 355, DOI: 10.1021/acssynbio.5b00296128https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhsFGqu74%253D&md5=609156affc1417f89485cfc7b001d95cThe All E. coli TX-TL Toolbox 2.0: A Platform for Cell-Free Synthetic BiologyGaramella, Jonathan; Marshall, Ryan; Rustad, Mark; Noireaux, VincentACS Synthetic Biology (2016), 5 (4), 344-355CODEN: ASBCD6; ISSN:2161-5063. (American Chemical Society)We report on and provide a detailed characterization of the performance and properties of a recently developed, all Escherichia coli, cell-free transcription and translation system. Gene expression is entirely based on the endogenous translation components and transcription machinery provided by an E. coli cytoplasmic ext., thus expanding the repertoire of regulatory parts to hundreds of elements. We use a powerful metab. for ATP regeneration to achieve more than 2 mg/mL of protein synthesis in batch mode reactions, and more than 6 mg/mL in semicontinuous mode. While the strength of cell-free expression is increased by a factor of 3 on av., the output signal of simple gene circuits and the synthesis of entire bacteriophages are increased by orders of magnitude compared to previous results. MRNAs and protein degrdn., resp. tuned using E. coli MazF interferase and ClpXP AAA+ proteases, are characterized over a much wider range of rates than the first version of the cell-free toolbox. This system is a highly versatile cell-free platform to construct complex biol. systems through the execution of DNA programs composed of synthetic and natural bacterial regulatory parts.
- 129Kim, D.-M.; Swartz, J. R. Efficient Production of a Bioactive, Multiple Disulfide-Bonded Protein Using Modified Extracts of Escherichia Coli. Biotechnol. Bioeng. 2004, 85 (2), 122– 129, DOI: 10.1002/bit.10865There is no corresponding record for this reference.
- 130Jeong, S.; Nguyen, H. T.; Kim, C. H.; Ly, M. N.; Shin, K. Toward Artificial Cells: Novel Advances in Energy Conversion and Cellular Motility. Adv. Funct. Mater. 2020, 30 (11), 1907182, DOI: 10.1002/adfm.201907182130https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhvFSls70%253D&md5=ff8cbe75b127ecc70c73f49fd1075be0Toward Artificial Cells: Novel Advances in Energy Conversion and Cellular MotilityJeong, Sungwoo; Nguyen, Huong Thanh; Kim, Chang Ho; Ly, Mai Nguyet; Shin, KwanwooAdvanced Functional Materials (2020), 30 (11), 1907182CODEN: AFMDC6; ISSN:1616-301X. (Wiley-VCH Verlag GmbH & Co. KGaA)A review. The demand to discover every single cellular component has been continuously increasing along with the development of biol. techniques. The bottom-up approach to construct a cell-mimicking system from well-defined and tunable compns. is accelerating, with the ultimate goal of comprehending a biol. cell. From among the available techniques, the artificial cell has been increasingly recognized as one of the most powerful tools for building a cell-like system from scratch. This review summarizes the development of artificial cells, from a pure giant unilamellar vesicle (GUV) to a controllable, self-fueled proteoliposome, both of which are highly compartmentalized. The basic components of an artificial cell, as well as the optimal conditions required for successful, reproducible GUV formation and protein reconstitution, are discussed. Most importantly, progress in studying the metabolic reactions in and the motility of a reconstituted artificial cell are the main focus of the review. The ability to perform a complicated reaction cascade in a controllable manner is highlighted as a promising perspective to obtaining an autonomous and movable GUV.
- 131Hanczyc, M. M.; Fujikawa, S. M.; Szostak, J. W. Experimental Models of Primitive Cellular Compartments: Encapsulation, Growth, and Division. Science 2003, 302 (5645), 618– 622, DOI: 10.1126/science.1089904131https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3sXotlWqt74%253D&md5=647b217542d67ffe265a705e5231f180Experimental Models of Primitive Cellular Compartments: Encapsulation, Growth, and DivisionHanczyc, Martin M.; Fujikawa, Shelly M.; Szostak, Jack W.Science (Washington, DC, United States) (2003), 302 (5645), 618-622CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)The clay montmorillonite is known to catalyze the polymn. of RNA from activated ribonucleotides. Here we report that montmorillonite accelerates the spontaneous conversion of fatty acid micelles into vesicles. Clay particles often become encapsulated in these vesicles, thus providing a pathway for the prebiotic encapsulation of catalytically active surfaces within membrane vesicles. In addn., RNA adsorbed to clay can be encapsulated within vesicles. Once formed, such vesicles can grow by incorporating fatty acid supplied as micelles and can divide without diln. of their contents by extrusion through small pores. These processes mediate vesicle replication through cycles of growth and division. The formation, growth, and division of the earliest cells may have occurred in response to similar interactions with mineral particles and inputs of material and energy.
- 132Kretschmer, S.; Ganzinger, K. A.; Franquelim, H. G.; Schwille, P. Synthetic Cell Division via Membrane-Transforming Molecular Assemblies. BMC Biol. 2019, 17 (1), 43, DOI: 10.1186/s12915-019-0665-1132https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3M7psFSntg%253D%253D&md5=834c319f13f711a883cb26e659512c92Synthetic cell division via membrane-transforming molecular assembliesKretschmer Simon; Ganzinger Kristina A; Franquelim Henri G; Schwille Petra; Kretschmer Simon; Ganzinger Kristina ABMC biology (2019), 17 (1), 43 ISSN:.Reproduction, i.e. the ability to produce new individuals from a parent organism, is a hallmark of living matter. Even the simplest forms of reproduction require cell division: attempts to create a designer cell therefore should include a synthetic cell division machinery. In this review, we will illustrate how nature solves this task, describing membrane remodelling processes in general and focusing on bacterial cell division in particular. We discuss recent progress made in their in vitro reconstitution, identify open challenges, and suggest how purely synthetic building blocks could provide an additional and attractive route to creating artificial cell division machineries.
- 133Anzai, K.; Yoshida, M.; Kirino, Y. Change in Intravesicular Volume of Liposomes by Freeze-Thaw Treatment as Studied by the ESR Stopped-Flow Technique. Biochimica et Biophysica Acta (BBA) - Biomembranes 1990, 1021 (1), 21– 26, DOI: 10.1016/0005-2736(90)90378-2There is no corresponding record for this reference.
- 134van der Valk, T.; Pečnerová, P.; Díez-Del-Molino, D.; Bergström, A.; Oppenheimer, J.; Hartmann, S.; Xenikoudakis, G.; Thomas, J. A.; Dehasque, M.; Sağlıcan, E.; Fidan, F. R.; Barnes, I.; Liu, S.; Somel, M.; Heintzman, P. D.; Nikolskiy, P.; Shapiro, B.; Skoglund, P.; Hofreiter, M.; Lister, A. M.; Götherström, A.; Dalén, L. Million-Year-Old DNA Sheds Light on the Genomic History of Mammoths. Nature 2021, 591 (7849), 265– 269, DOI: 10.1038/s41586-021-03224-9134https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXktlGhuro%253D&md5=a3cb5a5555d6390029f7aa91ec3671f2Million-year-old DNA sheds light on the genomic history of mammothsvan der Valk, Tom; Pecnerova, Patricia; Diez-del-Molino, David; Bergstroem, Anders; Oppenheimer, Jonas; Hartmann, Stefanie; Xenikoudakis, Georgios; Thomas, Jessica A.; Dehasque, Marianne; Saglican, Ekin; Fidan, Fatma Rabia; Barnes, Ian; Liu, Shanlin; Somel, Mehmet; Heintzman, Peter D.; Nikolskiy, Pavel; Shapiro, Beth; Skoglund, Pontus; Hofreiter, Michael; Lister, Adrian M.; Goetherstroem, Anders; Dalen, LoveNature (London, United Kingdom) (2021), 591 (7849), 265-269CODEN: NATUAS; ISSN:0028-0836. (Nature Research)Temporal genomic data hold great potential for studying evolutionary processes such as speciation. However, sampling across speciation events would, in many cases, require genomic time series that stretch well back into the Early Pleistocene subepoch. Although theor. models suggest that DNA should survive on this timescale1, the oldest genomic data recovered so far are from a horse specimen dated to 780-560 thousand years ago2. Here we report the recovery of genome-wide data from three mammoth specimens dating to the Early and Middle Pleistocene subepochs, two of which are more than one million years old. We find that two distinct mammoth lineages were present in eastern Siberia during the Early Pleistocene. One of these lineages gave rise to the woolly mammoth and the other represents a previously unrecognized lineage that was ancestral to the first mammoths to colonize North America. Our analyses reveal that the Columbian mammoth of North America traces its ancestry to a Middle Pleistocene hybridization between these two lineages, with roughly equal admixt. proportions. Finally, we show that the majority of protein-coding changes assocd. with cold adaptation in woolly mammoths were already present one million years ago. These findings highlight the potential of deep-time palaeogenomics to expand our understanding of speciation and long-term adaptive evolution.
- 135Li, Z.; Deutscher, M. P. Analyzing the Decay of Stable RNAs in E. Coli. Methods Enzymol. 2008, 447, 31– 45, DOI: 10.1016/S0076-6879(08)02202-7There is no corresponding record for this reference.
- 136Chan, L. Y.; Mugler, C. F.; Heinrich, S.; Vallotton, P.; Weis, K. Non-Invasive Measurement of mRNA Decay Reveals Translation Initiation as the Major Determinant of mRNA Stability. eLife 2018, DOI: 10.7554/eLife.32536There is no corresponding record for this reference.
- 137Paul, N.; Joyce, G. F. A Self-Replicating Ligase Ribozyme. Proc. Natl. Acad. Sci. U. S. A. 2002, 99 (20), 12733– 12740, DOI: 10.1073/pnas.202471099There is no corresponding record for this reference.
- 138Robertson, M. P.; Joyce, G. F. Highly Efficient Self-Replicating RNA Enzymes. Chem. Biol. 2014, 21 (2), 238– 245, DOI: 10.1016/j.chembiol.2013.12.004138https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXisV2isg%253D%253D&md5=5a1af26a9543c01dd17ab35366474b4fHighly Efficient Self-Replicating RNA EnzymesRobertson, Michael P.; Joyce, Gerald F.Chemistry & Biology (Oxford, United Kingdom) (2014), 21 (2), 238-245CODEN: CBOLE2; ISSN:1074-5521. (Elsevier Ltd.)An RNA enzyme has been developed that catalyzes the joining of oligonucleotide substrates to form addnl. copies of itself, undergoing self-replication with exponential growth. The enzyme also can cross-replicate with a partner enzyme, resulting in their mutual exponential growth and enabling self-sustained Darwinian evolution. The opportunity for inventive evolution within this synthetic genetic system depends on the diversity of the evolving population, which is limited by the catalytic efficiency of the enzyme. Directed evolution was used to improve the efficiency of the enzyme and increase its exponential growth rate to 0.14 min-1, corresponding to a doubling time of 5 min. This is close to the limit of 0.21 min-1 imposed by the rate of product release, but sufficient to enable more than 80 logs of growth per day.
- 139Chen, I. A.; Salehi-Ashtiani, K.; Szostak, J. W. RNA Catalysis in Model Protocell Vesicles. J. Am. Chem. Soc. 2005, 127 (38), 13213– 13219, DOI: 10.1021/ja051784p139https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXpsVGhtr0%253D&md5=02ae4e206b5b8529dd41d7df46c56793RNA Catalysis in Model Protocell VesiclesChen, Irene A.; Salehi-Ashtiani, Kourosh; Szostak, Jack W.Journal of the American Chemical Society (2005), 127 (38), 13213-13219CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)We are engaged in a long-term effort to synthesize chem. systems capable of Darwinian evolution, based on the encapsulation of self-replicating nucleic acids in self-replicating membrane vesicles. Here, we address the issue of the compatibility of these two replicating systems. Fatty acids form vesicles that are able to grow and divide, but vesicles composed solely of fatty acids are incompatible with the folding and activity of most ribozymes, because low concns. of divalent cations (e.g., Mg2+) cause fatty acids to ppt. Furthermore, vesicles that grow and divide must be permeable to the cations and substrates required for internal metab. We used a mixt. of myristoleic acid and its glycerol monoester to construct vesicles that were Mg2+-tolerant and found that Mg2+ cations can permeate the membrane and equilibrate within a few minutes. In vesicles encapsulating a hammerhead ribozyme, the addn. of external Mg2+ led to the activation and self-cleavage of the ribozyme mols. Vesicles composed of these amphiphiles grew spontaneously through osmotically driven competition between vesicles, and further modification of the membrane compn. allowed growth following mixed micelle addn. Our results show that membranes made from simple amphiphiles can form vesicles that are stable enough to retain encapsulated RNAs in the presence of divalent cations, yet dynamic enough to grow spontaneously and allow the passage of Mg2+ and mononucleotides without specific macromol. transporters. This combination of stability and dynamics is crit. for building model protocells in the lab. and may have been important for early cellular evolution.
- 140Gorbalenya, A. E.; Enjuanes, L.; Ziebuhr, J.; Snijder, E. J. Nidovirales: Evolving the Largest RNA Virus Genome. Virus Res. 2006, 117 (1), 17– 37, DOI: 10.1016/j.virusres.2006.01.017There is no corresponding record for this reference.
- 141Joyce, G. F.; Szostak, J. W. Protocells and RNA Self-Replication. Cold Spring Harb. Perspect. Biol. 2018, 10 (9), a034801, DOI: 10.1101/cshperspect.a034801141https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhslCltrfL&md5=db161c0090d2a46ee85da2e6e93dba23Protocells and RNA self-replicationJoyce, Gerald F.; Szostak, Jack W.Cold Spring Harbor Perspectives in Biology (2018), 10 (9), a034801/1-a034801/20CODEN: CSHPEU; ISSN:1943-0264. (Cold Spring Harbor Laboratory Press)The general notion of an "RNA world" is that, in the early development of life on the Earth, genetic continuity was assured by the replication of RNA, and RNA mols. were the chief agents of catalytic function. Assuming that all of the components of RNA were available in some prebiotic locale, these components could have assembled into activated nucleotides that condensed to form RNA polymers, setting the stage for the chem. replication of polynucleotides through RNA-templated RNA polymn. If a sufficient diversity of RNAs could be copied with reasonable rate and fidelity, then Darwinian evolution would begin with RNAs that facilitated their own reprodn. enjoying a selective advantage. The concept of a "protocell" refers to a compartment where replication of the primitive genetic material took place and where primitive catalysts gave rise to products that accumulated locally for the benefit of the replicating cellular entity. Replication of both the protocell and its encapsulated genetic material would have enabled natural selection to operate based on the differential fitness of competing cellular entities, ultimately giving rise to modern cellular life.
- 142Jahn, M.; Vorpahl, C.; Hübschmann, T.; Harms, H.; Müller, S. Copy Number Variability of Expression Plasmids Determined by Cell Sorting and Droplet Digital PCR. Microb. Cell Fact. 2016, 15 (1), 211, DOI: 10.1186/s12934-016-0610-8There is no corresponding record for this reference.
- 143Nielsen, A. A. K.; Der, B. S.; Shin, J.; Vaidyanathan, P.; Paralanov, V.; Strychalski, E. A.; Ross, D.; Densmore, D.; Voigt, C. A. Genetic Circuit Design Automation. Science 2016, 352 (6281), aac7341, DOI: 10.1126/science.aac7341There is no corresponding record for this reference.
- 144Salis, H. M.; Mirsky, E. A.; Voigt, C. A. Automated Design of Synthetic Ribosome Binding Sites to Control Protein Expression. Nat. Biotechnol. 2009, 27 (10), 946– 950, DOI: 10.1038/nbt.1568144https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXht1WgsbzO&md5=2e61e8668c2011f8d0afa0b1378a3f25Automated design of synthetic ribosome binding sites to control protein expressionSalis, Howard M.; Mirsky, Ethan A.; Voigt, Christopher A.Nature Biotechnology (2009), 27 (10), 946-950CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)Microbial engineering often requires fine control over protein expression-for example, to connect genetic circuits or control flux through a metabolic pathway. To circumvent the need for trial and error optimization, we developed a predictive method for designing synthetic ribosome binding sites, enabling a rational control over the protein expression level. Exptl. validation of >100 predictions in Escherichia coli showed that the method is accurate to within a factor of 2.3 over a range of 100,000-fold. The design method also correctly predicted that reusing identical ribosome binding site sequences in different genetic contexts can result in different protein expression levels. We demonstrate the method's utility by rationally optimizing protein expression to connect a genetic sensor to a synthetic circuit. The proposed forward engineering approach should accelerate the construction and systematic optimization of large genetic systems.
- 145Ostrov, N.; Beal, J.; Ellis, T.; Gordon, D. B.; Karas, B. J.; Lee, H. H.; Lenaghan, S. C.; Schloss, J. A.; Stracquadanio, G.; Trefzer, A.; Bader, J. S.; Church, G. M.; Coelho, C. M.; Efcavitch, J. W.; Güell, M.; Mitchell, L. A.; Nielsen, A. A. K.; Peck, B.; Smith, A. C.; Stewart, C. N., Jr; Tekotte, H. Technological Challenges and Milestones for Writing Genomes. Science 2019, 366 (6463), 310– 312, DOI: 10.1126/science.aay0339145https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhvFyntrnK&md5=24eb50b4c0f38df350da15331a3e6838Technological challenges and milestones for writing genomesOstrov, Nili; Beal, Jacob; Ellis, Tom; Gordon, D. Benjamin; Karas, Bogumil J.; Lee, Henry H.; Lenaghan, Scott C.; Schloss, Jeffery A.; Stracquadanio, Giovanni; Trefzer, Axel; Bader, Joel S.; Church, George M.; Coelho, Cintia M.; Efcavitch, J. William; Guell, Marc; Mitchell, Leslie A.; Nielsen, Alec A. K.; Peck, Bill; Smith, Alexander C.; Stewart, C. Neal, Jr.; Tekotte, HilleScience (Washington, DC, United States) (2019), 366 (6463), 310-312CODEN: SCIEAS; ISSN:1095-9203. (American Association for the Advancement of Science)A review. Engineering biol. with recombinant DNA, broadly called synthetic biol., has progressed tremendously in the last decade, owing to continued industrialization of DNA synthesis, discovery and development of mol. tools and organisms, and increasingly sophisticated modeling and analytic tools. However, we have yet to understand the full potential of engineering biol. because of our inability to write and test whole genomes, which we call synthetic genomics. Substantial improvements are needed to reduce the cost and increase the speed and reliability of genetic tools. Here, we identify emerging technologies and improvements to existing methods that will be needed in four major areas to advance synthetic genomics within the next 10 years: genome design, DNA synthesis, genome editing, and chromosome construction (see table). Similar to other large-scale projects for responsible advancement of innovative technologies, such as the Human Genome Project, an international, cross-disciplinary effort consisting of public and private entities will likely yield maximal return on investment and open new avenues of research and biotechnol.
- 146Farasat, I.; Kushwaha, M.; Collens, J.; Easterbrook, M.; Guido, M.; Salis, H. M. Efficient Search, Mapping, and Optimization of Multi-Protein Genetic Systems in Diverse Bacteria. Mol. Syst. Biol. 2014, 10 (6), 731, DOI: 10.15252/msb.20134955146https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2cfkvFGlug%253D%253D&md5=3531fb8c73dfed9285fea7b9e68b307cEfficient search, mapping, and optimization of multi-protein genetic systems in diverse bacteriaFarasat Iman; Easterbrook Michael; Guido Matthew; Kushwaha Manish; Collens Jason; Salis Howard MMolecular systems biology (2014), 10 (), 731 ISSN:.Developing predictive models of multi-protein genetic systems to understand and optimize their behavior remains a combinatorial challenge, particularly when measurement throughput is limited. We developed a computational approach to build predictive models and identify optimal sequences and expression levels, while circumventing combinatorial explosion. Maximally informative genetic system variants were first designed by the RBS Library Calculator, an algorithm to design sequences for efficiently searching a multi-protein expression space across a > 10,000-fold range with tailored search parameters and well-predicted translation rates. We validated the algorithm's predictions by characterizing 646 genetic system variants, encoded in plasmids and genomes, expressed in six gram-positive and gram-negative bacterial hosts. We then combined the search algorithm with system-level kinetic modeling, requiring the construction and characterization of 73 variants to build a sequence-expression-activity map (SEAMAP) for a biosynthesis pathway. Using model predictions, we designed and characterized 47 additional pathway variants to navigate its activity space, find optimal expression regions with desired activity response curves, and relieve rate-limiting steps in metabolism. Creating sequence-expression-activity maps accelerates the optimization of many protein systems and allows previous measurements to quantitatively inform future designs.
- 147Pretorius, I. S.; Boeke, J. D. Yeast 2.0-Connecting the Dots in the Construction of the World’s First Functional Synthetic Eukaryotic Genome. FEMS Yeast Res. 2018, DOI: 10.1093/femsyr/foy032There is no corresponding record for this reference.
- 148Blight, K. J.; Kolykhalov, A. A.; Rice, C. M. Efficient Initiation of HCV RNA Replication in Cell Culture. Science 2000, 290 (5498), 1972– 1974, DOI: 10.1126/science.290.5498.1972148https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXoslKgsL4%253D&md5=67eb65d15efc36854faf3940db9a5d91Efficient initiation of HCV RNA replication in cell cultureBlight, Keril J.; Kolykhalov, Alexander A.; Rice, Charles M.Science (Washington, D. C.) (2000), 290 (5498), 1972-1974CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Hepatitis C virus (HCV) infection is a global health problem affecting an estd. 170 million individuals worldwide. We report the identification of multiple independent adaptive mutations that cluster in the HCV nonstructural protein NS5A and confer increased replicative ability in vitro. Among these adaptive mutations were a single amino acid substitution that allowed HCV RNA replication in 10% of transfected hepatoma cells and a deletion of 47 amino acids encompassing the interferon (IFN) sensitivity detg. region (ISDR). Independent of the ISDR, IFN-α rapidly inhibited HCV RNA replication in vitro. This work establishes a robust, cell-based system for genetic and functional analyses of HCV replication.
- 149Gibson, D. G.; Young, L.; Chuang, R.-Y.; Venter, J. C.; Hutchison, C. A., 3rd; Smith, H. O. Enzymatic Assembly of DNA Molecules up to Several Hundred Kilobases. Nat. Methods 2009, 6 (5), 343– 345, DOI: 10.1038/nmeth.1318149https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXksVemsbw%253D&md5=46284924c7d73c47cfb490983338e480Enzymatic assembly of DNA molecules up to several hundred kilobasesGibson, Daniel G.; Young, Lei; Chuang, Ray-Yuan; Venter, J. Craig; Hutchison, Clyde A.; Smith, Hamilton O.Nature Methods (2009), 6 (5), 343-345CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)The authors describe an isothermal, single-reaction method for assembling multiple overlapping DNA mols. by the concerted action of a 5' exonuclease, a DNA polymerase and a DNA ligase. First they recessed DNA fragments, yielding single-stranded DNA overhangs that specifically annealed, and then covalently joined them. This assembly method can be used to seamlessly construct synthetic and natural genes, genetic pathways and entire genomes, and could be a useful mol. engineering tool.
- 150Venter, J. C.; Glass, J. I.; Hutchison, C. A., 3rd; Vashee, S. Synthetic Chromosomes, Genomes, Viruses, and Cells. Cell 2022, 185 (15), 2708– 2724, DOI: 10.1016/j.cell.2022.06.046150https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhvV2lu7%252FP&md5=dc83f01578fe7603c336facd51bd82f3Synthetic chromosomes, genomes, viruses, and cellsVenter, J. Craig; Glass, John I.; Hutchison, Clyde A. III; Vashee, SanjayCell (Cambridge, MA, United States) (2022), 185 (15), 2708-2724CODEN: CELLB5; ISSN:0092-8674. (Cell Press)A review. Synthetic genomics is the construction of viruses, bacteria, and eukaryotic cells with synthetic genomes. It involves two basic processes: synthesis of complete genomes or chromosomes and booting up of those synthetic nucleic acids to make viruses or living cells. The first synthetic genomics efforts resulted in the construction of viruses. This led to a revolution in viral reverse genetics and improvements in vaccine design and manuf. The first bacterium with a synthetic genome led to construction of a minimal bacterial cell and recoded Escherichia coli strains able to incorporate multiple non-std. amino acids in proteins and resistant to phage infection. Further advances led to a yeast strain with a synthetic genome and new approaches for animal and plant artificial chromosomes. On the horizon there are dramatic advances in DNA synthesis that will enable extraordinary new opportunities in medicine, industry, agriculture, and research.
- 151Fredens, J.; Wang, K.; de la Torre, D.; Funke, L. F. H.; Robertson, W. E.; Christova, Y.; Chia, T.; Schmied, W. H.; Dunkelmann, D. L.; Beránek, V.; Uttamapinant, C.; Llamazares, A. G.; Elliott, T. S.; Chin, J. W. Total Synthesis of Escherichia Coli with a Recoded Genome. Nature 2019, 569 (7757), 514– 518, DOI: 10.1038/s41586-019-1192-5151https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXpslGmurg%253D&md5=0c419f23bf66ed52bb301287015c2b0aTotal synthesis of Escherichia coli with a recoded genomeFredens, Julius; Wang, Kaihang; de la Torre, Daniel; Funke, Louise F. H.; Robertson, Wesley E.; Christova, Yonka; Chia, Tiongsun; Schmied, Wolfgang H.; Dunkelmann, Daniel L.; Beranek, Vaclav; Uttamapinant, Chayasith; Llamazares, Andres Gonzalez; Elliott, Thomas S.; Chin, Jason W.Nature (London, United Kingdom) (2019), 569 (7757), 514-518CODEN: NATUAS; ISSN:0028-0836. (Nature Research)Nature uses 64 codons to encode the synthesis of proteins from the genome, and chooses 1 sense codon-out of up to 6 synonyms-to encode each amino acid. Synonymous codon choice has diverse and important roles, and many synonymous substitutions are detrimental. Here we demonstrate that the no. of codons used to encode the canonical amino acids can be reduced, through the genome-wide substitution of target codons by defined synonyms. We create a variant of Escherichia coli with a four-megabase synthetic genome through a high-fidelity convergent total synthesis. Our synthetic genome implements a defined recoding and refactoring scheme-with simple corrections at just seven positions-to replace every known occurrence of two sense codons and a stop codon in the genome. Thus, we recode 18,214 codons to create an organism with a 61-codon genome; this organism uses 59 codons to encode the 20 amino acids, and enables the deletion of a previously essential tRNA.
- 152Zhao, Y.; Coelho, C.; Hughes, A. L.; Lazar-Stefanita, L.; Yang, S.; Brooks, A. N.; Walker, R. S. K.; Zhang, W.; Lauer, S.; Hernandez, C.; Cai, J.; Mitchell, L. A.; Agmon, N.; Shen, Y.; Sall, J.; Fanfani, V.; Jalan, A.; Rivera, J.; Liang, F.-X.; Bader, J. S.; Stracquadanio, G.; Steinmetz, L. M.; Cai, Y.; Boeke, J. D. Debugging and Consolidating Multiple Synthetic Chromosomes Reveals Combinatorial Genetic Interactions. Cell 2023, 186 (24), 5220– 5236, DOI: 10.1016/j.cell.2023.09.025There is no corresponding record for this reference.
- 153Woese, C. The Universal Ancestor. Proc. Natl. Acad. Sci. U. S. A. 1998, 95 (12), 6854– 6859, DOI: 10.1073/pnas.95.12.6854There is no corresponding record for this reference.
- 154Woese, C. R. On the Evolution of Cells. Proc. Natl. Acad. Sci. U. S. A. 2002, 99 (13), 8742– 8747, DOI: 10.1073/pnas.132266999There is no corresponding record for this reference.
- 155Sandberg, T. E.; Wise, K. S.; Dalldorf, C.; Szubin, R.; Feist, A. M.; Glass, J. I.; Palsson, B. O. Adaptive Evolution of a Minimal Organism with a Synthetic Genome. iScience 2023, 26 (9), 107500, DOI: 10.1016/j.isci.2023.107500There is no corresponding record for this reference.
- 156Moger-Reischer, R. Z.; Glass, J. I.; Wise, K. S.; Sun, L.; Bittencourt, D. M. C.; Lehmkuhl, B. K.; Schoolmaster, D. R., Jr; Lynch, M.; Lennon, J. T. Evolution of a Minimal Cell. Nature 2023, 620 (7972), 122– 127, DOI: 10.1038/s41586-023-06288-xThere is no corresponding record for this reference.
- 157Strotz, L. C.; Simões, M.; Girard, M. G.; Breitkreuz, L.; Kimmig, J.; Lieberman, B. S. Getting Somewhere with the Red Queen: Chasing a Biologically Modern Definition of the Hypothesis. Biol. Lett. 2018, 14 (5), 20170734, DOI: 10.1098/rsbl.2017.0734There is no corresponding record for this reference.
- 158Solé, R. Revisiting Leigh Van Valen’s “A New Evolutionary Law” (1973). Biol. Theory 2022, 17 (2), 120– 125, DOI: 10.1007/s13752-021-00391-wThere is no corresponding record for this reference.
- 159Hammerling, M. J.; Krüger, A.; Jewett, M. C. Strategies for in Vitro Engineering of the Translation Machinery. Nucleic Acids Res. 2020, 48 (3), 1068– 1083, DOI: 10.1093/nar/gkz1011159https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXhs1Ogur3M&md5=90749c3fcd81c379acb645f02d89a421Strategies for in vitro engineering of the translation machineryHammerling, Michael J.; Krueger, Antje; Jewett, Michael C.Nucleic Acids Research (2020), 48 (3), 1068-1083CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)A review. Engineering the process of mol. translation, or protein biosynthesis, has emerged as a major opportunity in synthetic and chem. biol. to generate novel biol. insights and enable new applications (e.g. designer protein therapeutics). Here, we review methods for engineering the process of translation in vitro. We discuss the advantages and drawbacks of the two major strategies-purified and ext.-based systems-and how they may be used to manipulate and study translation. Techniques to engineer each component of the translation machinery are covered in turn, including tRNAs, translation factors, and the ribosome. Finally, future directions and enabling technol. advances for the field are discussed.
- 160Williams, T. C.; Averesch, N. J. H.; Winter, G.; Plan, M. R.; Vickers, C. E.; Nielsen, L. K.; Krömer, J. O. Quorum-Sensing Linked RNA Interference for Dynamic Metabolic Pathway Control in Saccharomyces Cerevisiae. Metab. Eng. 2015, 29, 124– 134, DOI: 10.1016/j.ymben.2015.03.008160https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXkvF2ru7w%253D&md5=c58b47da7c9140d71340afecce19f5e7Quorum-sensing linked RNA interference for dynamic metabolic pathway control in Saccharomyces cerevisiaeWilliams, T. C.; Averesch, N. J. H.; Winter, G.; Plan, M. R.; Vickers, C. E.; Nielsen, L. K.; Kromer, J. O.Metabolic Engineering (2015), 29 (), 124-134CODEN: MEENFM; ISSN:1096-7176. (Elsevier B. V.)Some of the most productive metabolic engineering strategies involve genetic modifications that cause severe metabolic burden on the host cell. Growth-limiting genetic modifications can be more effective if they are 'switched on' after a population growth phase has been completed. To address this problem we have engineered dynamic regulation using a previously developed synthetic quorum sensing circuit in Saccharomyces cerevisiae. The circuit autonomously triggers gene expression at a high population d., and was linked with an RNA interference module to enable target gene silencing. As a demonstration the circuit was used to control flux through the shikimate pathway for the prodn. of para-hydroxybenzoic acid (PHBA). Dynamic RNA repression allowed gene knock-downs which were identified by elementary flux mode anal. as highly productive but with low biomass formation to be implemented after a population growth phase, resulting in the highest published PHBA titer in yeast (1.1 mM).
- 161Robinson, A. O.; Venero, O. M.; Adamala, K. P. Toward Synthetic Life: Biomimetic Synthetic Cell Communication. Curr. Opin. Chem. Biol. 2021, 64, 165– 173, DOI: 10.1016/j.cbpa.2021.08.008161https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXitFejsr%252FL&md5=850478c427d6eabda0a43b5fcfdb4423Toward synthetic life: Biomimetic synthetic cell communicationRobinson, Abbey O.; Venero, Orion M.; Adamala, Katarzyna P.Current Opinion in Chemical Biology (2021), 64 (), 165-173CODEN: COCBF4; ISSN:1367-5931. (Elsevier B.V.)A review. Engineering synthetic minimal cells provide a controllable chassis for studying the biochem. principles of natural life, increasing our understanding of complex biol. processes. Recently, synthetic cell engineering has enabled communication between both natural live cells and other synthetic cells. A system such as these enable studying interactions between populations of cells, both natural and artificial, and engineering small mol. cell communication protocols for a variety of basic research and practical applications. In this review, we summarize recent progress in engineering communication between synthetic and natural cells, and we speculate about the possible future directions of this work.
- 162Garamella, J.; Majumder, S.; Liu, A. P.; Noireaux, V. An Adaptive Synthetic Cell Based on Mechanosensing, Biosensing, and Inducible Gene Circuits. ACS Synth. Biol. 2019, 8 (8), 1913– 1920, DOI: 10.1021/acssynbio.9b00204162https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtl2jtLnP&md5=d0c92306403ebbacb296c43bf9f5e9d0An Adaptive Synthetic Cell Based on Mechanosensing, Biosensing, and Inducible Gene CircuitsGaramella, Jonathan; Majumder, Sagardip; Liu, Allen P.; Noireaux, VincentACS Synthetic Biology (2019), 8 (8), 1913-1920CODEN: ASBCD6; ISSN:2161-5063. (American Chemical Society)The bottom-up assembly of synthetic cell systems capable of recapitulating biol. functions has become a means to understand living matter by construction. The integration of biomol. components into active, cell-sized, genetically programmed compartments remains, however, a major bottleneck for building synthetic cells. A primary feature of real cells is their ability to actively interact with their surroundings, particularly in stressed conditions. Here, we construct a synthetic cell equipped with an inducible genetic circuit that responds to changes in osmotic pressure through the mechanosensitive channel MscL. Liposomes loaded with an E. coli cell-free transcription-translation (TXTL) system are induced with IPTG when exposed to hypo-osmotic soln., resulting in the expression of a bacterial cytoskeletal protein MreB. MreB assocs. with the membrane to generate a cortex-like structure. Our work provides the first example of mol. integration that couples mechanosensitivity, gene expression, and self-assembly at the inner membrane of synthetic cells.
- 163Liu, H.; Yang, Q.; Peng, R.; Kuai, H.; Lyu, Y.; Pan, X.; Liu, Q.; Tan, W. Artificial Signal Feedback Network Mimicking Cellular Adaptivity. J. Am. Chem. Soc. 2019, 141 (16), 6458– 6461, DOI: 10.1021/jacs.8b13816163https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXms1Wqsr4%253D&md5=4f308379b93eae5483c35a93279cf605Artificial Signal Feedback Network Mimicking Cellular AdaptivityLiu, Hui; Yang, Qiuxia; Peng, Ruizi; Kuai, Hailan; Lyu, Yifan; Pan, Xiaoshu; Liu, Qiaoling; Tan, WeihongJournal of the American Chemical Society (2019), 141 (16), 6458-6461CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Inspired by this elegant system of cellular adaptivity, we herein report the rational design of a dynamic artificial adaptive system able to sense and respond to environmental stresses in a unique sense-and-respond mode. Utilizing DNA nanotechnol., we constructed an artificial signal feedback network and anchored it to surface membrane of a model giant membrane vesicle (GMV) protocell. Such system would need to both sense incoming stimuli and emit a feedback response to eliminate the stimuli. To accomplish this mechanistically, our DNA-based artificial signal system, hereinafter termed DASsys, was equipped with a DNA trig-ger-induced DNA polymer formation and dissocn. machinery. Thus, through a sequential cascade of stimulus-induced DNA strand displacement, DASsys could effectively sense and respond to incoming stimuli. Then, by eliminating the stimulus, the membrane surface would return to its initial state, realizing the formation of a cyclical feedback mechanism. Overall, our strategy opens up a route to the construction of artificial signaling system capable of maintaining homeostasis in the cellular micromilieu, and addresses important emerging challenges in bioinspired engineering.
- 164Gispert, I.; Hindley, J. W.; Pilkington, C. P.; Shree, H.; Barter, L. M. C.; Ces, O.; Elani, Y. Stimuli-Responsive Vesicles as Distributed Artificial Organelles for Bacterial Activation. Proc. Natl. Acad. Sci. U. S. A. 2022, 119 (42), e2206563119, DOI: 10.1073/pnas.2206563119There is no corresponding record for this reference.
- 165Aufinger, L.; Simmel, F. C. Establishing Communication Between Artificial Cells. Chemistry 2019, 25 (55), 12659– 12670, DOI: 10.1002/chem.201901726165https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhsFSrt7vJ&md5=c7040b1a0bbee738b5c20f4a23e8d205Establishing Communication Between Artificial CellsAufinger, Lukas; Simmel, Friedrich C.Chemistry - A European Journal (2019), 25 (55), 12659-12670CODEN: CEUJED; ISSN:0947-6539. (Wiley-VCH Verlag GmbH & Co. KGaA)A review. Communication between artificial cells is essential for the realization of complex dynamical behaviors at the multi-cell level. It is also an important prerequisite for modular systems design, because it dets. how spatially sepd. functional modules can coordinate their actions. Among others, mol. communication is required for artificial cell signaling, synchronization of cellular behaviors, computation, group-level decision-making processes and pattern formation in artificial tissues. In this review, an overview of various recent approaches to create communicating artificial cellular systems is provided. In this context, important physicochem. boundary conditions that have to be considered for the design of the communicating cells are also described, and a survey of the most striking emergent behaviors that may be achieved in such systems is given.
- 166Pereyre, S.; Sirand-Pugnet, P.; Beven, L.; Charron, A.; Renaudin, H.; Barré, A.; Avenaud, P.; Jacob, D.; Couloux, A.; Barbe, V.; de Daruvar, A.; Blanchard, A.; Bébéar, C. Life on Arginine for Mycoplasma Hominis: Clues from Its Minimal Genome and Comparison with Other Human Urogenital Mycoplasmas. PLoS Genet. 2009, 5 (10), e1000677, DOI: 10.1371/journal.pgen.1000677There is no corresponding record for this reference.
- 167Woese, C. R.; Maniloff, J.; Zablen, L. B. Phylogenetic Analysis of the Mycoplasmas. Proc. Natl. Acad. Sci. U. S. A. 1980, 77 (1), 494– 498, DOI: 10.1073/pnas.77.1.494There is no corresponding record for this reference.
- 168Li, S.; Guan, J.-L.; Chien, S. Biochemistry and Biomechanics of Cell Motility. Annu. Rev. Biomed. Eng. 2005, 7, 105– 150, DOI: 10.1146/annurev.bioeng.7.060804.100340168https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2MXps12ltL8%253D&md5=c2f77f8a164b1ab53ed970c85e6d7ffbBiochemistry and biomechanics of cell motilityLi, Song; Guan, Jun-Lin; Chien, ShuAnnual Review of Biomedical Engineering (2005), 7 (), 105-150, 2 platesCODEN: ARBEF7; ISSN:1523-9829. (Annual Reviews Inc.)A review. Cell motility is an essential cellular process for a variety of biol. events. The process of cell migration requires the integration and coordination of complex biochem. and biomech. signals. The protrusion force at the leading edge of a cell is generated by the cytoskeleton, and this force generation is controlled by multiple signaling cascades. The formation of new adhesions at the front and the release of adhesions at the rear involve the outside-in and inside-out signaling mediated by integrins and other adhesion receptors. The traction force generated by the cell on the extracellular matrix (ECM) regulates cell-ECM adhesions, and the counter force exerted by ECM on the cell drives the migration. The polarity of cell migration can be amplified and maintained by the feedback loop between the cytoskeleton and cell-ECM adhesions. Cell migration in 3-dimensional ECM has characteristics distinct from that on 2-dimensional ECM. The migration of cells is initiated and modulated by external chem. and mech. factors, such as chemoattractants and the mech. forces acting on the cells and ECM, as well as the surface d., distribution, topog., and rigidity of the ECM.
- 169Brunet, T.; Albert, M.; Roman, W.; Coyle, M. C.; Spitzer, D. C.; King, N. A Flagellate-to-Amoeboid Switch in the Closest Living Relatives of Animals. eLife 2021, DOI: 10.7554/eLife.61037There is no corresponding record for this reference.
- 170Siton-Mendelson, O.; Bernheim-Groswasser, A. Toward the Reconstitution of Synthetic Cell Motility. Cell Adh. Migr. 2016, 10 (5), 461– 474, DOI: 10.1080/19336918.2016.1170260170https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC28fkt1KitA%253D%253D&md5=614c8b2da2257939347704e78670e1f4Toward the reconstitution of synthetic cell motilitySiton-Mendelson Orit; Bernheim-Groswasser AnneCell adhesion & migration (2016), 10 (5), 461-474 ISSN:.Cellular motility is a fundamental process essential for embryonic development, wound healing, immune responses, and tissues development. Cells are mostly moving by crawling on external, or inside, substrates which can differ in their surface composition, geometry, and dimensionality. Cells can adopt different migration phenotypes, e.g., bleb-based and protrusion-based, depending on myosin contractility, surface adhesion, and cell confinement. In the few past decades, research on cell motility has focused on uncovering the major molecular players and their order of events. Despite major progresses, our ability to infer on the collective behavior from the molecular properties remains a major challenge, especially because cell migration integrates numerous chemical and mechanical processes that are coupled via feedbacks that span over large range of time and length scales. For this reason, reconstituted model systems were developed. These systems allow for full control of the molecular constituents and various system parameters, thereby providing insight into their individual roles and functions. In this review we describe the various reconstituted model systems that were developed in the past decades. Because of the multiple steps involved in cell motility and the complexity of the overall process, most of the model systems focus on very specific aspects of the individual steps of cell motility. Here we describe the main advancement in cell motility reconstitution and discuss the main challenges toward the realization of a synthetic motile cell.
- 171Blanken, D.; van Nies, P.; Danelon, C. Quantitative Imaging of Gene-Expressing Liposomes Reveals Rare Favorable Phenotypes. Phys. Biol. 2019, 16 (4), 045002, DOI: 10.1088/1478-3975/ab0c62171https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXitV2gs7Y%253D&md5=5cfe9787e67c5772d03f800f2a440dfcQuantitative imaging of gene-expressing liposomes reveals rare favorable phenotypesBlanken, Duco; van Nies, Pauline; Danelon, ChristophePhysical Biology (2019), 16 (4), 045002CODEN: PBHIAT; ISSN:1478-3975. (IOP Publishing Ltd.)We report on direct imaging of tens of thousands of gene-expressing liposomes per sample allowing us to assess sub-population features in a statistically relevant manner. Both the vesicle size(diam. <10μm) and lipid compn. (mixt. of phospholipids with zwitterionic and neg. charged headgroups, including cardiolipin) are compatible with the properties of bacterial cells. Therefore, our liposomes provide suitable chassis to host Escherichia coli-derived PURE translation machinery and other bacterial processes in future developments. The potential of high-content imaging to identify rare phenotypes is demonstrated by fact that a subset of the liposome population exhibits remarkably high yield of synthesized protein or prolonged expression lifespan that surpasses performance of ensemble liposome-averaged and bulk reactions. Among the three com. PURE systems tested, PUREfrex2.0 offers the most favorable phenotypes displaying both high yield and long protein synthesis lifespan. Moreover, probing membrane permeability reveals a large heterogeneity amongst liposomes. In situ expression and membrane embedding of the pore-forming connexin leads to a characteristic permeability time profile, while increasing the fraction of permeable liposomes in the population. We see diversity in gene expression dynamics and membrane permeability as an opportunity to complement a rational design approach aiming at further implementing biol. functions in liposome-based synthetic cells.
- 172Stano, P.; D’Aguanno, E.; Bolz, J.; Fahr, A.; Luisi, P. L. A Remarkable Self-Organization Process as the Origin of Primitive Functional Cells. Angew. Chem., Int. Ed. Engl. 2013, 52 (50), 13397– 13400, DOI: 10.1002/anie.201306613There is no corresponding record for this reference.
- 173Weitz, M.; Kim, J.; Kapsner, K.; Winfree, E.; Franco, E.; Simmel, F. C. Diversity in the Dynamical Behaviour of a Compartmentalized Programmable Biochemical Oscillator. Nat. Chem. 2014, 6 (4), 295– 302, DOI: 10.1038/nchem.1869173https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXisFOjtbs%253D&md5=bc290cb6326fadb156dcb85af3eb587fDiversity in the dynamical behaviour of a compartmentalized programmable biochemical oscillatorWeitz, Maximilian; Kim, Jongmin; Kapsner, Korbinian; Winfree, Erik; Franco, Elisa; Simmel, Friedrich C.Nature Chemistry (2014), 6 (4), 295-302CODEN: NCAHBB; ISSN:1755-4330. (Nature Publishing Group)In vitro compartmentalization of biochem. reaction networks is a crucial step towards engineering artificial cell-scale devices and systems. At this scale the dynamics of mol. systems becomes stochastic, which introduces several engineering challenges and opportunities. Here we study a programmable transcriptional oscillator system that is compartmentalized into microemulsion droplets with vols. between 33 fl and 16 pl. Simultaneous measurement of large populations of droplets reveals major variations in the amplitude, frequency and damping of the oscillations. Variability increases for smaller droplets and depends on the operating point of the oscillator. Rather than reflecting the stochastic kinetics of the chem. reaction network itself, the variability can be attributed to the statistical variation of reactant concns. created during their partitioning into droplets. We anticipate that robustness to partitioning variability will be a crit. challenge for engineering cell-scale systems, and that highly parallel time-series acquisition from microemulsion droplets will become a key tool for characterization of stochastic circuit function.
- 174Hansen, M. M. K.; Meijer, L. H. H.; Spruijt, E.; Maas, R. J. M.; Rosquelles, M. V.; Groen, J.; Heus, H. A.; Huck, W. T. S. Macromolecular Crowding Creates Heterogeneous Environments of Gene Expression in Picolitre Droplets. Nat. Nanotechnol. 2016, 11 (2), 191– 197, DOI: 10.1038/nnano.2015.243174https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhslantL%252FL&md5=d2f5c923076100fe2836c79f5bf0bc1bMacromolecular crowding creates heterogeneous environments of gene expression in picolitre dropletsHansen, Maike M. K.; Meijer, Lenny H. H.; Spruijt, Evan; Maas, Roel J. M.; Rosquelles, Marta Ventosa; Groen, Joost; Heus, Hans A.; Huck, Wilhelm T. S.Nature Nanotechnology (2016), 11 (2), 191-197CODEN: NNAABX; ISSN:1748-3387. (Nature Publishing Group)Understanding the dynamics of complex enzymic reactions in highly crowded small vols. is crucial for the development of synthetic minimal cells. Compartmentalized biochem. reactions in cell-sized containers exhibit a degree of randomness due to the small no. of mols. involved. However, it is unknown how the phys. environment contributes to the stochastic nature of multistep enzymic processes. Here, we present a robust method to quantify gene expression noise in vitro using droplet microfluidics. We study the changes in stochasticity in the cell-free gene expression of two genes compartmentalized within droplets as a function of DNA copy no. and macromol. crowding. We find that decreased diffusion caused by a crowded environment leads to the spontaneous formation of heterogeneous microenvironments of mRNA as local prodn. rates exceed the diffusion rates of macromols. This heterogeneity leads to a higher probability of the mol. machinery staying in the same microenvironment, directly increasing the system's stochasticity.
- 175Rao, C. V. Expanding the Synthetic Biology Toolbox: Engineering Orthogonal Regulators of Gene Expression. Curr. Opin. Biotechnol. 2012, 23 (5), 689– 694, DOI: 10.1016/j.copbio.2011.12.015There is no corresponding record for this reference.
- 176Yelleswarapu, M.; van der Linden, A. J.; van Sluijs, B.; Pieters, P. A.; Dubuc, E.; de Greef, T. F. A.; Huck, W. T. S. Sigma Factor-Mediated Tuning of Bacterial Cell-Free Synthetic Genetic Oscillators. ACS Synth. Biol. 2018, 7 (12), 2879– 2887, DOI: 10.1021/acssynbio.8b00300176https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXitFaqsbfK&md5=95b792df5ec10e6ce8bbdca9264977abSigma factor-mediated tuning of bacterial cell-free synthetic genetic oscillatorsYelleswarapu, Maaruthy; van der Linden, Ardjan J.; van Sluijs, Bob; Pieters, Pascal A.; Dubuc, Emilien; de Greef, Tom F. A.; Huck, Wilhelm T. S.ACS Synthetic Biology (2018), 7 (12), 2879-2887CODEN: ASBCD6; ISSN:2161-5063. (American Chemical Society)Cell-free transcription-translation provides a simplified prototyping environment to rapidly design and study synthetic networks. Despite the presence of a well-characterized toolbox of genetic elements, examples of genetic networks that exhibit complex temporal behavior are scarce. Here, we present a genetic oscillator implemented in an Escherichia coli-based cell-free system under steady-state conditions using microfluidic flow reactors. The oscillator had an activator-repressor motif that utilizes the native transcriptional machinery of E. coli: the RNA polymerase (RNAP) and its assocd. σ-factors. We optimized a kinetic model with exptl. data using an evolutionary algorithm to quantify the key regulatory model parameters. The functional modulation of RNAP was investigated by coupling 2 oscillators driven by competing σ-factors, allowing the modification of network properties by means of passive transcriptional regulation.
- 177Li, J.; Zhang, C.; Huang, P.; Kuru, E.; Forster-Benson, E. T. C.; Li, T.; Church, G. M. Dissecting Limiting Factors of the Protein Synthesis Using Recombinant Elements (PURE) System. Translation (Austin) 2017, 5 (1), e1327006, DOI: 10.1080/21690731.2017.1327006There is no corresponding record for this reference.
- 178Lavickova, B.; Maerkl, S. J. A Simple, Robust, and Low-Cost Method To Produce the PURE Cell-Free System. ACS Synth. Biol. 2019, 8 (2), 455– 462, DOI: 10.1021/acssynbio.8b00427178https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXnvF2gtA%253D%253D&md5=510c38e95769a3ea6ba79ea2238c7d87A Simple, Robust, and Low-Cost Method To Produce the PURE Cell-Free SystemLavickova, Barbora; Maerkl, Sebastian J.ACS Synthetic Biology (2019), 8 (2), 455-462CODEN: ASBCD6; ISSN:2161-5063. (American Chemical Society)We demonstrate a simple, robust, and low-cost method for producing the PURE cell-free transcription-translation system. Our OnePot PURE system achieved a protein synthesis yield of 156 μg/mL at a cost of 0.09 USD/μL, leading to a 14-fold improvement in cost normalized protein synthesis yield over existing PURE systems. The one-pot method makes the PURE system easy to generate and allows it to be readily optimized and modified.
- 179Li, J.; Gu, L.; Aach, J.; Church, G. M. Improved Cell-Free RNA and Protein Synthesis System. PLoS One 2014, 9 (9), e106232, DOI: 10.1371/journal.pone.0106232179https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhs12isr7F&md5=4eb9de2e59d20518523324e7ee54a406Improved cell-free RNA and protein synthesis systemLi, Jun; Gu, Liangcai; Aach, John; Church, George M.PLoS One (2014), 9 (9), e106232/1-e106232/11, 11 pp.CODEN: POLNCL; ISSN:1932-6203. (Public Library of Science)Cell-free RNA and protein synthesis (CFPS) is becoming increasingly used for protein prodn. as yields increase and costs decrease. Advances in reconstituted CFPS systems such as the Protein synthesis Using Recombinant Elements (PURE) system offer new opportunities to tailor the reactions for specialized applications including in vitro protein evolution, protein microarrays, isotopic labeling, and incorporating unnatural amino acids. In this study, using firefly luciferase synthesis as a reporter system, we improved PURE system productivity up to 5 fold by adding or adjusting a variety of factors that affect transcription and translation, including Elongation factors (EF-Ts, EF-Tu, EF-G and EF4), ribosome recycling factor (RRF), release factors (RF1, RF2, RF3), chaperones (GroEL/ES), BSA and tRNAs. The work provides a more efficient defined in vitro transcription and translation system and a deeper understanding of the factors that limit the whole system efficiency.
- 180Jewett, M. C.; Calhoun, K. A.; Voloshin, A.; Wuu, J. J.; Swartz, J. R. An Integrated Cell-Free Metabolic Platform for Protein Production and Synthetic Biology. Mol. Syst. Biol. 2008, 4, 220, DOI: 10.1038/msb.2008.57180https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BD1cnmtVWluw%253D%253D&md5=59a196db8dc23aaf194e876ce7fdd36bAn integrated cell-free metabolic platform for protein production and synthetic biologyJewett Michael C; Calhoun Kara A; Voloshin Alexei; Wuu Jessica J; Swartz James RMolecular systems biology (2008), 4 (), 220 ISSN:.Cell-free systems offer a unique platform for expanding the capabilities of natural biological systems for useful purposes, i.e. synthetic biology. They reduce complexity, remove structural barriers, and do not require the maintenance of cell viability. Cell-free systems, however, have been limited by their inability to co-activate multiple biochemical networks in a single integrated platform. Here, we report the assessment of biochemical reactions in an Escherichia coli cell-free platform designed to activate natural metabolism, the Cytomim system. We reveal that central catabolism, oxidative phosphorylation, and protein synthesis can be co-activated in a single reaction system. Never before have these complex systems been shown to be simultaneously activated without living cells. The Cytomim system therefore promises to provide the metabolic foundation for diverse ab initio cell-free synthetic biology projects. In addition, we describe an improved Cytomim system with enhanced protein synthesis yields (up to 1200 mg/l in 2 h) and lower costs to facilitate production of protein therapeutics and biochemicals that are difficult to make in vivo because of their toxicity, complexity, or unusual cofactor requirements.
- 181Silverman, A. D.; Karim, A. S.; Jewett, M. C. Cell-Free Gene Expression: An Expanded Repertoire of Applications. Nat. Rev. Genet. 2020, 21 (3), 151– 170, DOI: 10.1038/s41576-019-0186-3181https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXit1OitbbO&md5=e227ddcf47b0b961b2b423d414fa3795Cell-free gene expression: an expanded repertoire of applicationsSilverman, Adam D.; Karim, Ashty S.; Jewett, Michael C.Nature Reviews Genetics (2020), 21 (3), 151-170CODEN: NRGAAM; ISSN:1471-0056. (Nature Research)Cell-free biol. is the activation of biol. processes without the use of intact living cells. It has been used for more than 50 years across the life sciences as a foundational research tool, but a recent tech. renaissance has facilitated high-yielding (grams of protein per L), cell-free gene expression systems from model bacteria, the development of cell-free platforms from non-model organisms and multiplexed strategies for rapidly assessing biol. design. These advances provide exciting opportunities to profoundly transform synthetic biol. by enabling new approaches to the model-driven design of synthetic gene networks, the fast and portable sensing of compds., on-demand biomanufg., building cells from the bottom up, and next-generation educational kits.
- 182Ouyang, X.; Zhou, X.; Lai, S. N.; Liu, Q.; Zheng, B. Immobilization of Proteins of Cell Extract to Hydrogel Networks Enhances the Longevity of Cell-Free Protein Synthesis and Supports Gene Networks. ACS Synth. Biol. 2021, 10 (4), 749– 755, DOI: 10.1021/acssynbio.0c00541There is no corresponding record for this reference.
- 183New England Biolabs. NEBExpress Cell-free ecoli Protein Synthesis System. https://www.neb.com/products/e5360-nebexpress-cell-free-ecoli-protein-synthesis-system (accessed on August 21, 2023).There is no corresponding record for this reference.
- 184Zimmerman, S. B.; Trach, S. O. Estimation of Macromolecule Concentrations and Excluded Volume Effects for the Cytoplasm of Escherichia Coli. J. Mol. Biol. 1991, 222 (3), 599– 620, DOI: 10.1016/0022-2836(91)90499-V184https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK38XlsFanuw%253D%253D&md5=df94a9250fe979cc422c967c1a7332caEstimation of macromolecule concentrations and excluded volume effects for the cytoplasm of Escherichia coliZimmerman, Steven B.; Trach, Stefan O.Journal of Molecular Biology (1991), 222 (3), 599-620CODEN: JMOBAK; ISSN:0022-2836.The very high concn. of macromols. within cells can potentially have an overwhelming effect on the thermodn. activity of cellular components because of excluded vol. effects. To est. the magnitudes of such effects, an exptl. study was made of the cytoplasm of Escherichia coli. Parameters from cells and cell exts. are used to calc. approx. activity coeffs. for cytoplasmic conditions. These calcns. require a representation of the sizes, concns. and effective sp. vol. of the macromols. in the exts. Macromol. size representations are obtained either by applying a two-phase distribution assay to define a related homogeneous soln. or by using the mol. mass distribution of macromols. from gel filtration. Macromol. concns. in cytoplasm are obtained from analyses of exts. by applying a correction for the diln. that occurs during extn. That factor is detd. from expts. based upon the known impermeability of the cytoplasmic vol. to sucrose in intact E. coli. Macromol. concns. in the cytoplasm of E. coli in either exponential or stationary growth phase are estd. to be ≈0.3 to 0.4 g/mL. Macromol. sp. vol. are inferred from the compn. of close-packed ppts. induced by polyethylene glycol. Several well-characterized proteins which bind to DNA (lac repressor, RNA polymerase) are extremely sensitive to changes in salt concn. in studies in vitro, but are insensitive in studies in vivo. Application of the activity coeffs. from the present work indicates that at least part of this discrepancy arises from the differences in excluded vols. in these studies. Applications of the activity coeffs. to soly. or to assocn. reactions are also discussed, as are changes assocd. with cell growth phase and osmotic or other effects. The use of solns. of purified macromols. that emulate the crowding conditions inferred for cytoplasm is discussed.
- 185Record, M. T., Jr; Courtenay, E. S.; Cayley, D. S.; Guttman, H. J. Responses of E. Coli to Osmotic Stress: Large Changes in Amounts of Cytoplasmic Solutes and Water. Trends Biochem. Sci. 1998, 23 (4), 143– 148, DOI: 10.1016/S0968-0004(98)01196-7185https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK1cXivFKmsLc%253D&md5=51c3002e687a30f01fccddaf77ca9ebbResponses of E. coli to osmotic stress: large changes in amounts of cytoplasmic solutes and waterRecord, M. Thomas, Jr.; Courtenay, Elizabeth S.; Cayley, D. Scott; Guttman, Harry J.Trends in Biochemical Sciences (1998), 23 (4), 143-148CODEN: TBSCDB; ISSN:0968-0004. (Elsevier Science Ltd.)A review with 37 refs. Escherichia coli is capable of growing in environments ranging from very dil. aq. solns. of essential nutrients to media contg. molar concns. of salts or nonelectrolyte solutes. Growth in environments with such a wide range (at least 100-fold) of osmolarities poses significant physiol. challenges for cells. To meet these challenges, E. coli adjusts a wide range of cytoplasmic soln. variables, including the cytoplasmic amts. both of water and of charged and uncharged solutes.
- 186Cayley, S.; Lewis, B. A.; Guttman, H. J.; Record, M. T. Jr. Characterization of the Cytoplasm of Escherichia Coli K-12 as a Function of External Osmolarity. Implications for Protein-DNA Interactions in Vivo. J. Mol. Biol. 1991, 222 (2), 281– 300, DOI: 10.1016/0022-2836(91)90212-OThere is no corresponding record for this reference.
- 187Li, J.; Haas, W.; Jackson, K.; Kuru, E.; Jewett, M. C.; Fan, Z. H.; Gygi, S.; Church, G. M. Cogenerating Synthetic Parts toward a Self-Replicating System. ACS Synth. Biol. 2017, 6 (7), 1327– 1336, DOI: 10.1021/acssynbio.6b00342187https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXks12htrg%253D&md5=6ecae4340f4b4616f4b3bd1f41d5a359Co-generating Synthetic Parts toward a Self-Replicating SystemLi, Jun; Haas, Wilhelm; Jackson, Kirsten; Kuru, Erkin; Jewett, Michael C.; Fan, Z. Hugh; Gygi, Steven; Church, George M.ACS Synthetic Biology (2017), 6 (7), 1327-1336CODEN: ASBCD6; ISSN:2161-5063. (American Chemical Society)To build replicating systems with new functions, the engineering of existing biol. machineries requires a sensible strategy. Protein synthesis Using Recombinant Elements (PURE) system consists of the desired components for transcription, translation, aminoacylation and energy regeneration. PURE might be the basis for a radically alterable, lifelike system after optimization. Here, the authors regenerated 54 E. coli ribosomal (r-) proteins individually from DNA templates in the PURE system. The authors show that using stable isotope labeling with amino acids, mass spectrometry based quant. proteomics could detect 26 of the 33 50S and 20 of the 21 30S subunit r-proteins when coexpressed in batch format PURE system. By optimizing DNA template concns. and adapting a miniaturized Fluid Array Device with optimized feeding soln., the authors were able to cogenerate and detect at least 29 of the 33 50S and all of the 21 30S subunit r-proteins in one pot. The boost on yield of a single r-protein in coexpression pool varied from ∼1.5 to 5-fold compared to the batch mode, with up to ∼2.4 μM yield for a single r-protein. Reconstituted ribosomes under physiol. condition from PURE system synthesized 30S r-proteins and native 16S rRNA showed ∼13% activity of native 70S ribosomes, which increased to 21% when supplemented with GroEL/ES. This work also points to what is still needed to obtain self-replicating synthetic ribosomes in situ in the PURE system.
- 188Murase, Y.; Nakanishi, H.; Tsuji, G.; Sunami, T.; Ichihashi, N. In Vitro Evolution of Unmodified 16S rRNA for Simple Ribosome Reconstitution. ACS Synth. Biol. 2018, 7 (2), 576– 583, DOI: 10.1021/acssynbio.7b00333There is no corresponding record for this reference.
- 189Jewett, M. C.; Fritz, B. R.; Timmerman, L. E.; Church, G. M. In Vitro Integration of Ribosomal RNA Synthesis, Ribosome Assembly, and Translation. Mol. Syst. Biol. 2013, 9, 678, DOI: 10.1038/msb.2013.31189https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhtFSqsLbM&md5=65e884f5f49acc8f173da9b949a81237In vitro integration of ribosomal RNA synthesis, ribosome assembly, and translationJewett, Michael C.; Fritz, Brian R.; Timmerman, Laura E.; Church, George M.Molecular Systems Biology (2013), 9 (), 678CODEN: MSBOC3; ISSN:1744-4292. (Nature Publishing Group)Purely in vitro ribosome synthesis could provide a crit. step towards unraveling the systems biol. of ribosome biogenesis, constructing minimal cells from defined components, and engineering ribosomes with new functions. Here, as an initial step towards this goal, we report a method for constructing Escherichia coli ribosomes in crude S150 E. coli exts. While conventional methods for E. coli ribosome reconstitution are non-physiol., our approach attempts to mimic chem. conditions in the cytoplasm, thus permitting several biol. processes to occur simultaneously. Specifically, our integrated synthesis, assembly, and translation (iSAT) technol. enables one-step co-activation of rRNA transcription, assembly of transcribed rRNA with native ribosomal proteins into functional ribosomes, and synthesis of active protein by these ribosomes in the same compartment. We show that iSAT makes possible the in vitro construction of modified ribosomes by introducing a 23S rRNA mutation that mediates resistance against clindamycin. We anticipate that iSAT will aid studies of ribosome assembly and open new avenues for making ribosomes with altered properties.
- 190Fritz, B. R.; Jamil, O. K.; Jewett, M. C. Implications of Macromolecular Crowding and Reducing Conditions for in Vitro Ribosome Construction. Nucleic Acids Res. 2015, 43 (9), 4774– 4784, DOI: 10.1093/nar/gkv329190https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhsFaisbzI&md5=3183327da923d2c7d665b3b7a33c4299Implications of macromolecular crowding and reducing conditions for in vitro ribosome constructionFritz, Brian R.; Jamil, Osman K.; Jewett, Michael C.Nucleic Acids Research (2015), 43 (9), 4774-4784CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)The in vitro construction of Escherichia coli ribosomes could elucidate a deeper understanding of these complex mol. machines and make possible the prodn. of synthetic variants with new functions. Toward this goal, the authors recently developed an integrated synthesis, assembly, and translation (iSAT) system that allows for co-activation of rRNA transcription and ribosome assembly, mRNA transcription, and protein translation without intact cells. Here, the authors discovered that macromol. crowding and reducing agents increased overall iSAT protein synthesis; the combination of 6% Ficoll 400 and 2 mM DTBA yielded an approx. 5-fold increase in overall iSAT protein synthesis activity. By utilizing a fluorescent RNA aptamer, fluorescent reporter proteins, and ribosome sedimentation anal., the authors showed that crowding agents increased iSAT yields by enhancing translation while reducing agents increased rRNA transcription and ribosome assembly. Finally, the authors showed that iSAT ribosomes possessed ∼70% of the protein synthesis activity of in vivo-assembled E. coli ribosomes. This work improved iSAT protein synthesis through the addn. of crowding and reducing agents, provides a thorough understanding of the effect of these additives within the iSAT system and demonstrated how iSAT allows for manipulation and anal. of ribosome biogenesis in the context of an in vitro transcription-translation system.
- 191Hammerling, M. J.; Fritz, B. R.; Yoesep, D. J.; Kim, D. S.; Carlson, E. D.; Jewett, M. C. In Vitro Ribosome Synthesis and Evolution through Ribosome Display. Nat. Commun. 2020, 11 (1), 1108, DOI: 10.1038/s41467-020-14705-2191https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3cXkvFaisbY%253D&md5=7a7004c5d0c04e4727b54c6d3c5723ccIn-vitro ribosome synthesis and evolution through ribosome displayHammerling, Michael J.; Fritz, Brian R.; Yoesep, Danielle J.; Kim, Do Soon; Carlson, Erik D.; Jewett, Michael C.Nature Communications (2020), 11 (1), 1108CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)Directed evolution of the ribosome for expanded substrate incorporation and novel functions is challenging because the requirement of cell viability limits the mutations that can be made. Here we address this challenge by combining cell-free synthesis and assembly of translationally competent ribosomes with ribosome display to develop a fully in-vitro methodol. for ribosome synthesis and evolution (called RISE). We validate the RISE method by selecting active genotypes from a ∼1.7 X 107 member library of rRNA (rRNA) variants, as well as identifying mutant ribosomes resistant to the antibiotic clindamycin from a library of ∼4 X 103 rRNA variants. We further demonstrate the prevalence of pos. epistasis in resistant genotypes, highlighting the importance of such interactions in selecting for new function. We anticipate that RISE will facilitate understanding of mol. translation and enable selection of ribosomes with altered properties.
- 192Wang, T.; Lu, Y. Toward Minimal Transcription-Translation Machinery. ACS Synth. Biol. 2023, 12 (11), 3312– 3327, DOI: 10.1021/acssynbio.3c00324There is no corresponding record for this reference.
- 193Oberholzer, T.; Albrizio, M.; Luisi, P. L. Polymerase Chain Reaction in Liposomes. Chem. Biol. 1995, 2 (10), 677– 682, DOI: 10.1016/1074-5521(95)90031-4193https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK2MXptFygtbc%253D&md5=297f9e6efcf5d03c3ab4810317037320Polymerase chain reaction in liposomesOberholzer, Thomas; Albrizio, Maria; Luisi, Pier LuigiChemistry & Biology (1995), 2 (10), 677-82CODEN: CBOLE2; ISSN:1074-5521. (Current Biology)Background: Compartmentalization of biochem. reactions within a spherically closed bilayer is an important step in the mol. evolution of cells. Liposomes are the most suitable structures to model this kind of chem. We have used the polymerase chain reaction (PCR) to demonstrate that complex biochem. reactions such as DNA replication can be carried out inside these compartments. Results: We describe the first example of DNA amplification by the PCR occurring inside liposomes composed of 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC), or of a mixt. of POPC and phosphatidylserine. We show that these liposomes are stable even under the high temp. conditions used for PCR. Although only a very small fraction of liposomes contains all eight different reagents together, a significant amt. of DNA is produced which can be obsd. by polyacrylamide gel electrophoresis. Conclusions: This work shows that it is possible to carry out complex biochem. reactions within liposomes, which may be germane to the question of the origin of living cells. We have established the parameters and conditions that are crit. for carrying out this complex reaction within the liposome compartment.
- 194Kawska, A.; Carvalho, K.; Manzi, J.; Boujemaa-Paterski, R.; Blanchoin, L.; Martiel, J.-L.; Sykes, C. How Actin Network Dynamics Control the Onset of Actin-Based Motility. Proc. Natl. Acad. Sci. U. S. A. 2012, 109 (36), 14440– 14445, DOI: 10.1073/pnas.1117096109There is no corresponding record for this reference.
- 195Hürtgen, D.; Härtel, T.; Murray, S. M.; Sourjik, V.; Schwille, P. Functional Modules of Minimal Cell Division for Synthetic Biology. Adv. Biosyst. 2019, 3 (6), e1800315, DOI: 10.1002/adbi.201800315There is no corresponding record for this reference.
- 196Pelletier, J. F.; Sun, L.; Wise, K. S.; Assad-Garcia, N.; Karas, B. J.; Deerinck, T. J.; Ellisman, M. H.; Mershin, A.; Gershenfeld, N.; Chuang, R.-Y.; Glass, J. I.; Strychalski, E. A. Genetic Requirements for Cell Division in a Genomically Minimal Cell. Cell 2021, 184 (9), 2430– 2440, DOI: 10.1016/j.cell.2021.03.008196https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXns1Cisrs%253D&md5=f2e8dc01302d6eac0e75d1d4d1597dc2Genetic requirements for cell division in a genomically minimal cellPelletier, James F.; Sun, Lijie; Wise, Kim S.; Assad-Garcia, Nacyra; Karas, Bogumil J.; Deerinck, Thomas J.; Ellisman, Mark H.; Mershin, Andreas; Gershenfeld, Neil; Chuang, Ray-Yuan; Glass, John I.; Strychalski, Elizabeth A.Cell (Cambridge, MA, United States) (2021), 184 (9), 2430-2440.e16CODEN: CELLB5; ISSN:0092-8674. (Cell Press)Genomically minimal cells, such as JCVI-syn3.0, offer a platform to clarify genes underlying core physiol. processes. Although this minimal cell includes genes essential for population growth, the physiol. of its single cells remained uncharacterized. To investigate striking morphol. variation in JCVI-syn3.0 cells, we present an approach to characterize cell propagation and det. genes affecting cell morphol. Microfluidic chemostats allowed observation of intrinsic cell dynamics that result in irregular morphologies. A genome with 19 genes not retained in JCVI-syn3.0 generated JCVI-syn3A, which presents morphol. similar to that of JCVI-syn1.0. We further identified seven of these 19 genes, including two known cell division genes, ftsZ and sepF, a hydrolase of unknown substrate, and four genes that encode membrane-assocd. proteins of unknown function, which are required together to restore a phenotype similar to that of JCVI-syn1.0. This result emphasizes the polygenic nature of cell division and morphol. in a genomically minimal cell. The fully annotated genome sequence of JCVI-syn3A is deposited in NCBI (GenBank: CP016816.2).
- 197Zhang, S.; Contini, C.; Hindley, J. W.; Bolognesi, G.; Elani, Y.; Ces, O. Engineering Motile Aqueous Phase-Separated Droplets via Liposome Stabilisation. Nat. Commun. 2021, 12 (1), 1673, DOI: 10.1038/s41467-021-21832-x197https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXmslKhtbc%253D&md5=d911a2717fe36d0872045e44ef9edf56Engineering motile aqueous phase-separated droplets via liposome stabilisationZhang, Shaobin; Contini, Claudia; Hindley, James W.; Bolognesi, Guido; Elani, Yuval; Ces, OscarNature Communications (2021), 12 (1), 1673CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)There are increasing efforts to engineer functional compartments that mimic cellular behaviors from the bottom-up. One behavior that is receiving particular attention is motility, due to its biotechnol. potential and ubiquity in living systems. Many existing platforms make use of the Marangoni effect to achieve motion in water/oil (w/o) droplet systems. However, most of these systems are unsuitable for biol. applications due to biocompatibility issues caused by the presence of oil phases. Here we report a biocompatible all aq. (wt./wt.) PEG/dextran Pickering-like emulsion system consisting of liposome-stabilized cell-sized droplets, where the stability can be easily tuned by adjusting liposome compn. and concn. We demonstrate that the compartments are capable of neg. chemotaxis: these droplets can respond to a PEG/dextran polymer gradient through directional motion down to the gradient. The biocompatibility, motility and partitioning abilities of this droplet system offers new directions to pursue research in motion-related biol. processes.
- 198Li, M.; Brinkmann, M.; Pagonabarraga, I.; Seemann, R.; Fleury, J.-B. Spatiotemporal Control of Cargo Delivery Performed by Programmable Self-Propelled Janus Droplets. Commun. Phys. 2018, 1 (1), 1– 8, DOI: 10.1038/s42005-018-0025-4198https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXpvV2lurk%253D&md5=25c2517f251115af25f3cfced1c1ab32Breakdown of diffusivity-entropy scaling in colloidal glass-forming liquidsLi, Bo; Xiao, Xiuming; Lou, Kai; Wang, Shuxia; Wen, Weijia; Wang, ZirenCommunications Physics (2018), 1 (1), 1-9CODEN: CPOHDJ; ISSN:2399-3650. (Nature Research)Glass is a liq. that has lost its ability to flow. Why this particular substance undergoes such a dramatic kinetic slowdown yet remains barely distinguishable in structure from its fluid state upon cooling constitutes the central question of glass transition physics. Here, we investigate the pathway of kinetic slowdown in glass-forming liqs. that consist of monolayers of ellipsoidal or binary spherical colloids. In contrast to rotational motion, the dynamics of the translational motion begin to violently slow down at considerably low area fractions (.vphi.T). At .vphi.T, anomalous translation-rotation coupling is enhanced and the topog. of the free energy landscape become rugged. Based on the pos. correlation between .vphi.T and fragility, the measurement of .vphi.T offers a novel method for predicting glassy dynamics, circumventing the prohibitive increase in equil. times required in high-d. regions. Our results highlight the role that thermodynamical entropy plays in glass transitions.
- 199Litschel, T.; Kelley, C. F.; Holz, D.; Adeli Koudehi, M.; Vogel, S. K.; Burbaum, L.; Mizuno, N.; Vavylonis, D.; Schwille, P. Reconstitution of Contractile Actomyosin Rings in Vesicles. Nat. Commun. 2021, 12 (1), 2254, DOI: 10.1038/s41467-021-22422-7199https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXptFOqsL8%253D&md5=5f2aaa315f2c39163e5b7e59c7b2aacbReconstitution of contractile actomyosin rings in vesiclesLitschel, Thomas; Kelley, Charlotte F.; Holz, Danielle; Adeli Koudehi, Maral; Vogel, Sven K.; Burbaum, Laura; Mizuno, Naoko; Vavylonis, Dimitrios; Schwille, PetraNature Communications (2021), 12 (1), 2254CODEN: NCAOBW; ISSN:2041-1723. (Nature Research)One of the grand challenges of bottom-up synthetic biol. is the development of minimal machineries for cell division. The mech. transformation of large-scale compartments, such as Giant Unilamellar Vesicles (GUVs), requires the geometry-specific coordination of active elements, several orders of magnitude larger than the mol. scale. Of all cytoskeletal structures, large-scale actomyosin rings appear to be the most promising cellular elements to accomplish this task. Here, we have adopted advanced encapsulation methods to study bundled actin filaments in GUVs and compare our results with theor. modeling. By changing few key parameters, actin polymn. can be differentiated to resemble various types of networks in living cells. Importantly, we find membrane binding to be crucial for the robust condensation into a single actin ring in spherical vesicles, as predicted by theor. considerations. Upon force generation by ATP-driven myosin motors, these ring-like actin structures contract and locally constrict the vesicle, forming furrow-like deformations. On the other hand, cortex-like actin networks are shown to induce and stabilize deformations from spherical shapes.
- 200Bashirzadeh, Y.; Moghimianavval, H.; Liu, A. P. Encapsulated Actomyosin Patterns Drive Cell-like Membrane Shape Changes. iScience 2022, 25 (5), 104236, DOI: 10.1016/j.isci.2022.104236200https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XhslGksbjK&md5=ed9418fdac1a79597adabe19c16c5de2Encapsulated actomyosin patterns drive cell-like membrane shape changesBashirzadeh, Yashar; Moghimianavval, Hossein; Liu, Allen P.iScience (2022), 25 (5), 104236CODEN: ISCICE; ISSN:2589-0042. (Elsevier B.V.)Cell shape changes from locomotion to cytokinesis are, to a large extent, driven by myosin-driven remodeling of cortical actin patterns. Passive crosslinkers such as α-actinin and fascin as well as actin nucleator Arp2/3 complex largely det. actin network architecture and, consequently, membrane shape changes. Here we reconstitute actomyosin networks inside cell-sized lipid bilayer vesicles and show that depending on vesicle size and concns. of α-actinin and fascin actomyosin networks assemble into ring and aster-like patterns. Anchoring actin to the membrane does not change actin network architecture yet exerts forces and deforms the membrane when assembled in the form of a contractile ring. In the presence of α-actinin and fascin, an Arp2/3 complex-mediated actomyosin cortex is shown to assemble a ring-like pattern at the equatorial cortex followed by myosin-driven clustering and consequently blebbing. An active gel theory unifies a model for the obsd. membrane shape changes induced by the contractile cortex.
- 201Gardner, P. M.; Winzer, K.; Davis, B. G. Sugar Synthesis in a Protocellular Model Leads to a Cell Signalling Response in Bacteria. Nat. Chem. 2009, 1 (5), 377– 383, DOI: 10.1038/nchem.296201https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXptVSjsrY%253D&md5=ad496ab10f8ec2775e43fbae9d4b25e8Sugar synthesis in a protocellular model leads to a cell signalling response in bacteriaGardner, Paul M.; Winzer, Klaus; Davis, Benjamin G.Nature Chemistry (2009), 1 (5), 377-383, S377/1-S377/50CODEN: NCAHBB; ISSN:1755-4330. (Nature Publishing Group)The design of systems with life-like properties from simple chem. components may offer insights into biol. processes, with the ultimate goal of creating an artificial chem. cell that would be considered to be alive. Most efforts to create artificial cells have concd. on systems based on complex natural mols. such as DNA and RNA. Here we have constructed a lipid-bound protometabolism that synthesizes complex carbohydrates from simple feedstocks, which are capable of engaging the natural quorum sensing mechanism of the marine bacterium Vibrio harveyi and stimulating a proportional bioluminescent response. This encapsulated system may represent the first step towards the realization of a cellular mimic' and a starting point for bottom-up' designs of other chem. cells, which could perhaps display complex behaviors such as communication with natural cells.
- 202Lentini, R.; Santero, S. P.; Chizzolini, F.; Cecchi, D.; Fontana, J.; Marchioretto, M.; Del Bianco, C.; Terrell, J. L.; Spencer, A. C.; Martini, L.; Forlin, M.; Assfalg, M.; Dalla Serra, M.; Bentley, W. E.; Mansy, S. S. Integrating Artificial with Natural Cells to Translate Chemical Messages That Direct E. Coli Behaviour. Nat. Commun. 2014, 5, 4012, DOI: 10.1038/ncomms5012202https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhvF2mur3K&md5=d80e7b3bbd5f02d32806c289530c7bc0Integrating artificial with natural cells to translate chemical messages that direct E. coli behaviourLentini, Roberta; Santero, Silvia Perez; Chizzolini, Fabio; Cecchi, Dario; Fontana, Jason; Marchioretto, Marta; Del Bianco, Cristina; Terrell, Jessica L.; Spencer, Amy C.; Martini, Laura; Forlin, Michele; Assfalg, Michael; Dalla Serra, Mauro; Bentley, William E.; Mansy, Sheref S.Nature Communications (2014), 5 (), 4012CODEN: NCAOBW; ISSN:2041-1723. (Nature Publishing Group)Previous efforts to control cellular behavior have largely relied upon various forms of genetic engineering. Once the genetic content of a living cell is modified, the behavior of that cell typically changes as well. However, other methods of cellular control are possible. All cells sense and respond to their environment. Therefore, artificial, non-living cellular mimics could be engineered to activate or repress already existing natural sensory pathways of living cells through chem. communication. Here we describe the construction of such a system. The artificial cells expand the senses of Escherichia coli by translating a chem. message that E. coli cannot sense on its own to a mol. that activates a natural cellular response. This methodol. could open new opportunities in engineering cellular behavior without exploiting genetically modified organisms.
- 203Ding, Y.; Contreras-Llano, L. E.; Morris, E.; Mao, M.; Tan, C. Minimizing Context Dependency of Gene Networks Using Artificial Cells. ACS Appl. Mater. Interfaces 2018, 10 (36), 30137– 30146, DOI: 10.1021/acsami.8b10029There is no corresponding record for this reference.
- 204Toparlak, Ö. D.; Zasso, J.; Bridi, S.; Serra, M. D.; Macchi, P.; Conti, L.; Baudet, M.-L.; Mansy, S. S. Artificial Cells Drive Neural Differentiation. Sci. Adv. 2020, DOI: 10.1126/sciadv.abb4920There is no corresponding record for this reference.
- 205Fanalista, F.; Birnie, A.; Maan, R.; Burla, F.; Charles, K.; Pawlik, G.; Deshpande, S.; Koenderink, G. H.; Dogterom, M.; Dekker, C. Shape and Size Control of Artificial Cells for Bottom-Up Biology. ACS Nano 2019, 13 (5), 5439– 5450, DOI: 10.1021/acsnano.9b00220205https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXptlKrtLw%253D&md5=10854f36682b1ae84c7c9bea92f127edShape and Size Control of Artificial Cells for Bottom-Up BiologyFanalista, Federico; Birnie, Anthony; Maan, Renu; Burla, Federica; Charles, Kevin; Pawlik, Grzegorz; Deshpande, Siddharth; Koenderink, Gijsje H.; Dogterom, Marileen; Dekker, CeesACS Nano (2019), 13 (5), 5439-5450CODEN: ANCAC3; ISSN:1936-0851. (American Chemical Society)Bottom-up biol. is an expanding research field that aims to understand the mechanisms underlying biol. processes via in vitro assembly of their essential components in synthetic cells. As encapsulation and controlled manipulation of these elements is a crucial step in the recreation of such cell-like objects, microfluidics is increasingly used for the prodn. of minimal artificial containers such as single-emulsion droplets, double-emulsion droplets, and liposomes. Despite the importance of cell morphol. on cellular dynamics, current synthetic-cell studies mainly use spherical containers, and methods to actively shape manipulate these have been lacking. In this paper, the authors describe a microfluidic platform to deform the shape of artificial cells into a variety of shapes (rods and disks) with adjustable cell-like dimensions below 5 μm, thereby mimicking realistic cell morphologies. To illustrate the potential of the method, the authors reconstitute three biol. relevant protein systems (FtsZ, microtubules, collagen) inside rod-shaped containers and study the arrangement of the protein networks inside these synthetic containers with physiol. relevant morphologies resembling those found in living cells.
- 206Van de Cauter, L.; Fanalista, F.; van Buren, L.; De Franceschi, N.; Godino, E.; Bouw, S.; Danelon, C.; Dekker, C.; Koenderink, G. H.; Ganzinger, K. A. Optimized cDICE for Efficient Reconstitution of Biological Systems in Giant Unilamellar Vesicles. ACS Synth. Biol. 2021, 10 (7), 1690– 1702, DOI: 10.1021/acssynbio.1c00068206https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXhsVWrt7zP&md5=b34708aa4daf3694358fa8fa0b4dfa3cOptimized cDICE for Efficient Reconstitution of Biological Systems in Giant Unilamellar VesiclesVan de Cauter, Lori; Fanalista, Federico; van Buren, Lennard; De Franceschi, Nicola; Godino, Elisa; Bouw, Sharon; Danelon, Christophe; Dekker, Cees; Koenderink, Gijsje H.; Ganzinger, Kristina A.ACS Synthetic Biology (2021), 10 (7), 1690-1702CODEN: ASBCD6; ISSN:2161-5063. (American Chemical Society)Giant unilamellar vesicles (GUVs) are often used to mimic biol. membranes in reconstitution expts. They are also widely used in research on synthetic cells, as they provide a mech. responsive reaction compartment that allows for controlled exchange of reactants with the environment. However, while many methods exist to encapsulate functional biomols. in GUVs, there is no one-size-fits-all soln. and reliable GUV fabrication still remains a major exptl. hurdle in the field. Here, we show that defect-free GUVs contg. complex biochem. systems can be generated by optimizing a double-emulsion method for GUV formation called continuous droplet interface crossing encapsulation (cDICE). By tightly controlling environmental conditions and tuning the lipid-in-oil dispersion, we show that it is possible to significantly improve the reproducibility of high-quality GUV formation as well as the encapsulation efficiency. We demonstrate efficient encapsulation for a range of biol. systems including a minimal actin cytoskeleton, membrane-anchored DNA nanostructures, and a functional PURE (protein synthesis using recombinant elements) system. Our optimized cDICE method displays promising potential to become a std. method in biophysics and bottom-up synthetic biol.
- 207Ganar, K. A.; Leijten, L.; Deshpande, S. Actinosomes: Condensate-Templated Containers for Engineering Synthetic Cells. ACS Synth. Biol. 2022, 11 (8), 2869– 2879, DOI: 10.1021/acssynbio.2c00290207https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XitVyjsLvM&md5=9c7b4edc9a9e23d40018ea0eef031085Actinosomes: Condensate-Templated Containers for Engineering Synthetic CellsGanar, Ketan A.; Leijten, Liza; Deshpande, SiddharthACS Synthetic Biology (2022), 11 (8), 2869-2879CODEN: ASBCD6; ISSN:2161-5063. (American Chemical Society)Engineering synthetic cells has a broad appeal, from understanding living cells to designing novel biomaterials for therapeutics, biosensing, and hybrid interfaces. A key prerequisite to creating synthetic cells is a three-dimensional container capable of orchestrating biochem. reactions. In this study, we present an easy and effective technique to make cell-sized porous containers, coined actinosomes, using the interactions between biomol. condensates and the actin cytoskeleton. This approach uses polypeptide/nucleoside triphosphate condensates and localizes actin monomers on their surface. By triggering actin polymn. and using osmotic gradients, the condensates are transformed into containers, with the boundary made up of actin filaments and polylysine polymers. We show that the guanosine triphosphate (GTP)-to-ATP (ATP) ratio is a crucial parameter for forming actinosomes: insufficient ATP prevents condensate dissoln., while excess ATP leads to undesired crumpling. Permeability studies reveal the porous surface of actinosomes, allowing small mols. to pass through while restricting bigger macromols. within the interior. We show the functionality of actinosomes as bioreactors by carrying out in vitro protein translation within them. Actinosomes are a handy addn. to the synthetic cell platform, with appealing properties like ease of prodn., inherent encapsulation capacity, and a potentially active surface to trigger signaling cascades and form multicellular assemblies, conceivably useful for biotechnol. applications.
- 208Frischmon, C.; Sorenson, C.; Winikoff, M.; Adamala, K. P. Build-a-Cell: Engineering a Synthetic Cell Community. Life 2021, 11 (11), 1176, DOI: 10.3390/life11111176There is no corresponding record for this reference.
- 209Schwille, P.; Spatz, J.; Landfester, K.; Bodenschatz, E.; Herminghaus, S.; Sourjik, V.; Erb, T. J.; Bastiaens, P.; Lipowsky, R.; Hyman, A.; Dabrock, P.; Baret, J.-C.; Vidakovic-Koch, T.; Bieling, P.; Dimova, R.; Mutschler, H.; Robinson, T.; Tang, T.-Y. D.; Wegner, S.; Sundmacher, K. MaxSynBio: Avenues Towards Creating Cells from the Bottom Up. Angew. Chem., Int. Ed. Engl. 2018, 57 (41), 13382– 13392, DOI: 10.1002/anie.201802288209https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1MfitFWnsA%253D%253D&md5=869c39a9f651f1fcbb33665f2d5c97a9MaxSynBio: Avenues Towards Creating Cells from the Bottom UpSchwille Petra; Mutschler Hannes; Spatz Joachim; Landfester Katharina; Wegner Seraphine; Bodenschatz Eberhard; Herminghaus Stephan; Sourjik Victor; Erb Tobias J; Bastiaens Philippe; Bieling Peter; Lipowsky Reinhard; Dimova Rumiana; Robinson Tom; Hyman Anthony; Tang T-Y Dora; Dabrock Peter; Baret Jean-Christophe; Vidakovic-Koch Tanja; Sundmacher KaiAngewandte Chemie (International ed. in English) (2018), 57 (41), 13382-13392 ISSN:.A large German research consortium mainly within the Max Planck Society ("MaxSynBio") was formed to investigate living systems from a fundamental perspective. The research program of MaxSynBio relies solely on the bottom-up approach to synthetic biology. MaxSynBio focuses on the detailed analysis and understanding of essential processes of life through modular reconstitution in minimal synthetic systems. The ultimate goal is to construct a basic living unit entirely from non-living components. The fundamental insights gained from the activities in MaxSynBio could eventually be utilized for establishing a new generation of biotechnological processes, which would be based on synthetic cell constructs that replace the natural cells currently used in conventional biotechnology.
- 210Habets, M. G. J. L.; Zwart, H. A. E.; van Est, R. Why the Synthetic Cell Needs Democratic Governance. Trends Biotechnol. 2021, 39 (6), 539– 541, DOI: 10.1016/j.tibtech.2020.11.006There is no corresponding record for this reference.
- 211Rasmussen, S. Protocells: Bridging Nonliving and Living Matter; MIT Press, 2008.There is no corresponding record for this reference.
- 212Schwille, P. Bottom-up Synthetic Biology: Engineering in a Tinkerer’s World. Science 2011, 333 (6047), 1252– 1254, DOI: 10.1126/science.1211701212https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXhtV2jt7vF&md5=f390108f5f3ffacc15a14093ef4277c3Bottom-Up Synthetic Biology: Engineering in a Tinkerer's WorldSchwille, PetraScience (Washington, DC, United States) (2011), 333 (6047), 1252-1254CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)A review. How synthetic can "synthetic biol." be A literal interpretation of the name of this new life science discipline invokes expectations of the systematic construction of biol. systems with cells being built module by module-from the bottom up. But can this possibly be achieved, taking into account the enormous complexity and redundancy of living systems, which distinguish them quite remarkably from design features that characterize human inventions There are several recent developments in biol., in tight conjunction with quant. disciplines, that may bring this literal perspective into the realm of the possible. However, such bottom-up engineering requires tools that were originally designed by nature's greatest tinkerer: evolution.
- 213Abil, Z.; Danelon, C. Roadmap to Building a Cell: An Evolutionary Approach. Front. Bioeng. Biotechnol. 2020, 8, 927, DOI: 10.3389/fbioe.2020.00927213https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3s%252Fjt1Ontg%253D%253D&md5=1453ab51194499edbf69bf236a3e6c64Roadmap to Building a Cell: An Evolutionary ApproachAbil Zhanar; Danelon ChristopheFrontiers in bioengineering and biotechnology (2020), 8 (), 927 ISSN:2296-4185.Laboratory synthesis of an elementary biological cell from isolated components may aid in understanding of the fundamental principles of life and will provide a platform for a range of bioengineering and medical applications. In essence, building a cell consists in the integration of cellular modules into system's level functionalities satisfying a definition of life. To achieve this goal, we propose in this perspective to undertake a semi-rational, system's level evolutionary approach. The strategy would require iterative cycles of genetic integration of functional modules, diversification of hereditary information, compartmentalized gene expression, selection/screening, and possibly, assistance from open-ended evolution. We explore the underlying challenges to each of these steps and discuss possible solutions toward the bottom-up construction of an artificial living cell.
- 214Yarmuth; [d-Ky-3], J. A. Inflation Reduction Act of 2022 ; 2022. http://www.congress.gov/ (accessed on April 30, 2023).There is no corresponding record for this reference.
- 215The White House. Executive order on advancing biotechnology and biomanufacturing innovation for a sustainable, safe, and secure American bioeconomy. The White House. https://www.whitehouse.gov/briefing-room/presidential-actions/2022/09/12/executive-order-on-advancing-biotechnology-and-biomanufacturing-innovation-for-a-sustainable-safe-and-secure-american-bioeconomy/ (accessed on April 30, 2023).There is no corresponding record for this reference.
- 216BioMADE. https://www.biomade.org/ (accessed on April 30, 2023).There is no corresponding record for this reference.
- 217Huang, A.; Nguyen, P. Q.; Stark, J. C.; Takahashi, M. K.; Donghia, N.; Ferrante, T.; Dy, A. J.; Hsu, K. J.; Dubner, R. S.; Pardee, K.; Jewett, M. C.; Collins, J. J. BioBitsTM Explorer: A Modular Synthetic Biology Education Kit. Sci. Adv. 2018, 4 (8), eaat5105, DOI: 10.1126/sciadv.aat5105There is no corresponding record for this reference.
- 218Stark, J. C.; Huang, A.; Nguyen, P. Q.; Dubner, R. S.; Hsu, K. J.; Ferrante, T. C.; Anderson, M.; Kanapskyte, A.; Mucha, Q.; Packett, J. S.; Patel, P.; Patel, R.; Qaq, D.; Zondor, T.; Burke, J.; Martinez, T.; Miller-Berry, A.; Puppala, A.; Reichert, K.; Schmid, M.; Brand, L.; Hill, L. R.; Chellaswamy, J. F.; Faheem, N.; Fetherling, S.; Gong, E.; Gonzalzles, E. M.; Granito, T.; Koritsaris, J.; Nguyen, B.; Ottman, S.; Palffy, C.; Patel, A.; Skweres, S.; Slaton, A.; Woods, T.; Donghia, N.; Pardee, K.; Collins, J. J.; Jewett, M. C. BioBitsTM Bright: A Fluorescent Synthetic Biology Education Kit. Sci. Adv. 2018, 4 (8), eaat5107, DOI: 10.1126/sciadv.aat5107There is no corresponding record for this reference.
- 219Stark, J. C.; Huang, A.; Hsu, K. J.; Dubner, R. S.; Forbrook, J.; Marshalla, S.; Rodriguez, F.; Washington, M.; Rybnicky, G. A.; Nguyen, P. Q.; Hasselbacher, B.; Jabri, R.; Kamran, R.; Koralewski, V.; Wightkin, W.; Martinez, T.; Jewett, M. C. BioBits Health: Classroom Activities Exploring Engineering, Biology, and Human Health with Fluorescent Readouts. ACS Synth. Biol. 2019, 8 (5), 1001– 1009, DOI: 10.1021/acssynbio.8b00381219https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXmt1Olu70%253D&md5=99551c2017559b0b1275e1d168dfb5a4BioBits Health: Classroom Activities Exploring Engineering, Biology, and Human Health with Fluorescent ReadoutsStark, Jessica C.; Huang, Ally; Hsu, Karen J.; Dubner, Rachel S.; Forbrook, Jason; Marshalla, Suzanne; Rodriguez, Faith; Washington, Mechelle; Rybnicky, Grant A.; Nguyen, Peter Q.; Hasselbacher, Brenna; Jabri, Ramah; Kamran, Rijha; Koralewski, Veronica; Wightkin, Will; Martinez, Thomas; Jewett, Michael C.ACS Synthetic Biology (2019), 8 (5), 1001-1009CODEN: ASBCD6; ISSN:2161-5063. (American Chemical Society)Recent advances in synthetic biol. have resulted in biol. technologies with the potential to reshape the way we understand and treat human disease. Educating students about the biol. and ethics underpinning these technologies is crit. to empower them to make informed future policy decisions regarding their use and to inspire the next generation of synthetic biologists. However, hands-on, educational activities that convey emerging synthetic biol. topics can be difficult to implement due to the expensive equipment and expertise required to grow living cells. We present BioBits Health, an educational kit contg. lab activities and supporting curricula for teaching antibiotic resistance mechanisms and CRISPR-Cas9 gene editing in high school classrooms. This kit links complex biol. concepts to visual, fluorescent readouts in user-friendly freeze-dried cell-free reactions. BioBits Health represents a set of educational resources that promises to encourage teaching of cutting-edge, health-related synthetic biol. topics in classrooms and other nonlab. settings.
- 220Rybnicky, G. A.; Dixon, R. A.; Kuhn, R. M.; Karim, A. S.; Jewett, M. C. Development of a Freeze-Dried CRISPR-Cas12 Sensor for Detecting Wolbachia in the Secondary Science Classroom. ACS Synth. Biol. 2022, 11 (2), 835– 842, DOI: 10.1021/acssynbio.1c00503There is no corresponding record for this reference.
- 221Jung, K. J.; Rasor, B. J.; Rybnicky, G. A.; Silverman, A. D.; Standeven, J.; Kuhn, R.; Granito, T.; Ekas, H. M.; Wang, B. M.; Karim, A. S.; Lucks, J. B.; Jewett, M. C. At-Home, Cell-Free Synthetic Biology Education Modules for Transcriptional Regulation and Environmental Water Quality Monitoring. bioRxiv , January 9, 2023. DOI: 10.1101/2023.01.09.523248 .There is no corresponding record for this reference.
- 222Perry, E.; Weber, J.; Pataranutaporn, P.; Volf, V.; Gonzalez, L. M.; Nejad, S.; Angleton, C.; Chen, J.-E.; Gabo, A.; Jammalamadaka, M. S. S.; Kuru, E.; Fortuna, P.; Rico, A.; Sulich, K.; Wawrzyniak, D.; Jacobson, J.; Church, G.; Kong, D. How to Grow (almost) Anything: A Hybrid Distance Learning Model for Global Laboratory-Based Synthetic Biology Education. Nat. Biotechnol. 2022, 40 (12), 1874– 1879, DOI: 10.1038/s41587-022-01601-x222https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB38XjtFSlsLzF&md5=5a8f9f979f93a31e57c86f0bca9f1a05How to grow (almost) anything: a hybrid distance learning model for global laboratory-based synthetic biology educationPerry, Eyal; Weber, Jessica; Pataranutaporn, Pat; Volf, Verena; Gonzalez, Laura Maria; Nejad, Sara; Angleton, Carolyn; Chen, Jia-En; Gabo, Ananda; Jammalamadaka, Mani Sai Suryateja; Kuru, Erkin; Fortuna, Patrick; Rico, Andres; Sulich, Karolina; Wawrzyniak, Dominika; Jacobson, Joseph; Church, George; Kong, DavidNature Biotechnology (2022), 40 (12), 1874-1879CODEN: NABIF9; ISSN:1087-0156. (Nature Portfolio)A pilot program for synthetic biol. education via a scalable distributed network model of distance-based lab. learning can be accessible globally across disciplines and backgrounds.