Design of Protein Logic Gate System Operating on Lipid MembranesClick to copy article linkArticle link copied!
- Neža OmersaNeža OmersaDepartment of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova ulica 19, 1001 Ljubljana, SloveniaBiomedicine Doctoral Program, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, SloveniaMore by Neža Omersa
- Saša AdenSaša AdenDepartment of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova ulica 19, 1001 Ljubljana, SloveniaBiomedicine Doctoral Program, University of Ljubljana, Vrazov trg 2, 1000 Ljubljana, SloveniaMore by Saša Aden
- Matic KisovecMatic KisovecDepartment of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova ulica 19, 1001 Ljubljana, SloveniaMore by Matic Kisovec
- Marjetka PodobnikMarjetka PodobnikDepartment of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova ulica 19, 1001 Ljubljana, SloveniaMore by Marjetka Podobnik
- Gregor Anderluh*Gregor Anderluh*Email: [email protected]Department of Molecular Biology and Nanobiotechnology, National Institute of Chemistry, Hajdrihova ulica 19, 1001 Ljubljana, SloveniaMore by Gregor Anderluh
Abstract
Lipid membranes are becoming increasingly popular in synthetic biology due to their biophysical properties and crucial role in communication between different compartments. Several alluring protein–membrane sensors have already been developed, whereas protein logic gates designs on membrane-embedded proteins are very limited. Here we demonstrate the construction of a two-level protein–membrane logic gate with an OR-AND logic. The system consists of an engineered pH-dependent pore-forming protein listeriolysin O and its DARPin-based inhibitor, conjugated to a lipid vesicle membrane. The gate responds to low pH and removal of the inhibitor from the membrane either by switching to a reducing environment, protease cleavage, or any other signal depending on the conjugation chemistry used for inhibitor attachment to the membrane. This unique protein logic gate vesicle system advances generic sensing and actuator platforms used in synthetic biology and could be utilized in drug delivery.
Figure 1
Figure 1. Properties and permeabilizing activity of Y406A. (a) A ribbon model of LLO 3D structure (PDB ID: 4CDB) with each domain (D1–D4) labeled in different color. Position of residue 406 is denoted by an orange color and an arrow. (b) A model of pore formation by Y406A (monomer binding to the membrane, oligomerization, pore formation). Domains are colored as on panel a. (c) Calcein release from large unilamellar vesicles after 30 min is shown at different pH values for LLO (black) and Y406A (red). Data reproduced with permission from Kisovec et al. (40) (d) Relative rate of hemolysis by 2.3 nM CDCs at pH 5.7 in the presence of DARPins: 5 μM D22 (red bars), 5 μM D22M (blue bar), a variant that was used for immobilization to the lipid membrane (see below), and 5 μM D6 and D30 (green bars), DARPin clones that were selected with ribosome display as high affinity binders, but did not inhibit hemolytic activity of Y406A. Mean ± SD; n = 2–7.
Results
DARPin D22 Binds Specifically to Y406A in Solution and in the Membrane-Bound State
Figure 2
Figure 2. Binding of D22 to Y406A in solution. (a) Size exclusion chromatogram of LLO and Y406A in the absence or presence of D22. Triangles indicate positions of elution peaks for different proteins. Note that LLO travels aberrantly on the size exclusion column eluting with larger volumes of elution buffer than expected. (b) Binding of D22 to LLO (gray) or Y406A (black) in solution (22 mM MES, 150 mM NaCl and 5 mM 2-mercaptoethanol, pH 5.7), measured by isothermal titration calorimetry. Top panel represents raw data of injections of 54.9 μM D22 into a 5.9 μM solution of LLO or Y406A. Bottom panel shows normalized integrated enthalpies plotted against the molar ratio. Circles represent experimental points, and the solid line corresponds to the best fit obtained by one-site reaction model. (c) An overlay of experimental scattering data obtained by SAXS experiment of Y406A-D22 complex (red circles) with the calculated scattering curve from the representative DAMMIF model (χ2 = 1.125, black line). Inset, overlay of Y406A (blue ribbon) and D22 (red ribbon) refined by rigid body modeling (χ2 = 1.12), with the best SAXS bead model in surface representation and the average SAXS bead model in mesh representation.
Figure 3
Figure 3. Interaction of D22 with Y406A in the lipid membrane environment. (a) SPR measurements showing binding of 100 nM Y406A, 5 μM D22 and preincubated Y406A-D22 complex (with same concentrations of individual proteins as used for single proteins injections) to large unilamellar vesicles. (b) SPR sensorgrams of 5 μM D22 binding to 100 nM membrane-inserted Y406A or LLO. Membrane denotes control experiment with D22 binding to vesicles only. (c) Vesicle sedimentation assays with multilamellar vesicles after preincubation of LLO and D22, or Y406A and D22 in solution (“preincubated”) or when LLO or Y406A were first preincubated with vesicles (“membrane bound”). p, pellet; s, supernatant; w, additional washing step, which was included when assaying membrane bound LLO or Y406A in order to check for the completeness of binding. Band at app. 60 kDa corresponds to LLO or Y406A, while band at ca. 18 kDa corresponds to D22. D22 is present at 5× molar excess; therefore, a large portion of it is always unbound in supernatant. (d) Quantification of the SDS-PAGE data from (c) by densitometry. Full binding of D22 to Y406A (100%) was considered when one-fifth of the applied D22 was bound to Y406A. Mean ± SD; two sample t test, ***P < 0.001 (n = 4–6). Amount of bound D22 is reported when preincubated with LLO (1) or Y406A (2) in solution or when LLO (3) or Y406A (4) were first bound to vesicles.
D22 Provides Additional Control of Y406A Permeabilizing Activity
Figure 4
Figure 4. Modulation of Y406A permeabilization activity by D22 and pH. (a) Permeabilization of GUVs for FD10 at different conditions and induced by the 50 nM LLO. (b) Permeabilization induced by 50 nM Y406A. The graphs on the right in A and B show quantification of GUVs data from confocal microscopy images as represented on the left. Mean ± SD; n = 96–568. (c) Hemolysis induced by 18.2 nM LLO at different conditions. (d) Hemolysis induced by 18.2 nM Y406A. In C and D, mean ± SD is presented; n = 3. (e) 5 μM D22 by itself does not induce permeabilization of GUVs. Confocal images of GUVs on the left, and quantification is presented on the right. n is 256 and 458 for pH 6.5 and 8.0, respectively. (f) The truth table and schematic representation of a NOR logic gate for the Y406A-D22 system.
Figure 5
Figure 5. Modulation of Y406A activity in calcein release experiments. (a,b) Calcein release from SUVs composed of POPC:Chol, 3:2 (mol:mol) as a result of pore formation by 1 μM LLO (a) or Y406A (b), in presence or absence of 5 μM D22 and at pH values 6.5 and 8.0. (c) Calcein release from LUVs composed of POPC:Chol, 1:1 (mol:mol) monitored at different conditions. Vesicles were stirred in 10 mM HEPES, 150 mM NaCl, 1 mM EDTA, pH 8.0. Final concentration of 30 nM Y406A (black triangle), 1 μM D22 or 3 μL of 7% HCl (to reduce pH to approximately 6.5) were added, respectively, at times denoted by triangles. The scale bar is the same for all fluorescent traces.
Membrane-Based System Including Covalently Bound D22 Allows Employment of Various Signals for Activation of Y406A
Figure 6
Figure 6. Conjugation of D22M to vesicles and cleavage with different agents. Schematic diagrams of used systems and structures of employed lipids are shown. Y406A is presented with green color, D22 is shown in orange. Different parts of lipids used for conjugation are presented with different colors in the structural formulas and on the diagram. (a–c) A system employing DSPE-PEG2000Mal lipid, which attaches D22 to the lipid membrane and allows subsequent cleavage by MMP-9. (d–f) A system employing DSPE-PEG2000PDP lipid, which allows cleavage with reductant TCEP. The input of different proteins and reagents is shown above the sedimentation assay gels. The approximate positions of different proteins and lipids on the gels are indicated by arrows. i, 2 μg of D22M as an input; p and s denote pellet and supernatant after centrifugation of MLVs, respectively. (c) and (f) each represent one experiment where MLVs were first incubated with Y406A and then with MMP-9 or TCEP. 1, pellet after both incubations; 2, supernatant after centrifugation of MLVs after incubation with Y406A; 3, supernatant after centrifugation of MLVs with bound Y406A and incubation of MMP-9 or TCEP.
Membrane System with Logic Gate Based on Y406A and D22M
Figure 7
Figure 7. A tunable vesicle system. (a) Y406A activity is controlled by pH and the reversible inhibitor D22. Vesicles are at pH 8.0; therefore, two input signals are needed for system activation: lowering pH with HCl and elimination of D22 from the system by release from vesicles induced by MMP-9 or TCEP. (b) A system based on DSPE-PEG2000Mal lipid. Bars represent fluorescence increase due to release of calcein from MLVs. A logic truth table and schematic representation of an AND gate is presented below the graph. (c) A system based on DSPE-PEG2000PDP lipid, which gives more flexibility for the removal of D22 from the vesicle membrane. A logic truth table and schematic representation of an OR-AND gate is presented beside the graph. n = 3–6, mean ± SD *P < 0.05; **P < 0.01.
Discussion
Methods
Materials
D22 Design and Ribosome Display
Cloning, Expression, and Purification of LLO, Y406A, D22, and D22M
Enzyme-Linked Immunosorbent Assay (ELISA)
Size Exclusion Chromatography (SEC)
Isothermal Titration Calorimetry (ITC)
Small-Angle X-ray Scattering (SAXS) Data Collection, Ab Initio Shape Determination and Molecular Modeling
Lipid Vesicles Preparation
Surface Plasmon Resonance (SPR)
Vesicle Sedimentation Assay
Hemolytic Assay
Permeabilization Experiments
Calcein Release from SUVs and LUVs
Conjugation and Cleavage of D22M from MLVs
Calcein Release from MLVs with Conjugated D22M
Statistical Analysis
Acknowledgments
The work presented in this paper was supported by The Slovenian Research Agency Grants P1-0391 and J4-8225. We thank Andreas Plückthun for the pRDV vector. We thank Alexey Kikhney from EMBL c/o DESY Synchrotron, Germany, to assist us with the SAXS experiments, Proposal No. SAXS-429.
References
This article references 72 other publications.
- 1Evans, A. C., Thadani, N. N., and Suh, J. (2016) Biocomputing nanoplatforms as therapeutics and diagnostics. J. Controlled Release 240, 387– 393, DOI: 10.1016/j.jconrel.2016.01.045Google Scholar1Biocomputing nanoplatforms as therapeutics and diagnosticsEvans, A. C.; Thadani, N. N.; Suh, J.Journal of Controlled Release (2016), 240 (), 387-393CODEN: JCREEC; ISSN:0168-3659. (Elsevier B.V.)A review. Biocomputing nanoplatforms are designed to detect and integrate single or multiple inputs under defined algorithms, such as Boolean logic gates, and generate functionally useful outputs, such as delivery of therapeutics or release of optically detectable signals. Using sensing modules composed of small mols., polymers, nucleic acids, or proteins/peptides, nanoplatforms have been programmed to detect and process extrinsic stimuli, such as magnetic fields or light, or intrinsic stimuli, such as nucleic acids, enzymes, or pH. Stimulus detection can be transduced by the nanomaterial via three different mechanisms: system assembly, system disassembly, or system transformation. The increasingly sophisticated suite of biocomputing nanoplatforms may be invaluable for a multitude of applications, including medical diagnostics, biomedical imaging, environmental monitoring, and delivery of therapeutics to target cell populations.
- 2Khalil, A. S. and Collins, J. J. (2010) Synthetic biology: Applications come of age. Nat. Rev. Genet. 11, 367– 379, DOI: 10.1038/nrg2775Google Scholar2Synthetic biology: applications come of ageKhalil, Ahmad S.; Collins, James J.Nature Reviews Genetics (2010), 11 (5), 367-379CODEN: NRGAAM; ISSN:1471-0056. (Nature Publishing Group)Advances in the synthetic biol. field are allowing an expansion beyond small gene networks towards larger biol. programs that hold promise for a wide range of applications, including biosensing, therapeutics and the prodn. of biofuels, pharmaceuticals and biomaterials. Synthetic biol. is bringing together engineers and biologists to design and build novel biomol. components, networks and pathways, and to use these constructs to rewire and reprogram organisms. These re-engineered organisms will change our lives over the coming years, leading to cheaper drugs, 'green' means to fuel our cars and targeted therapies for attacking 'superbugs' and diseases, such as cancer. The de novo engineering of genetic circuits, biol. modules and synthetic pathways is beginning to address these crucial problems and is being used in related practical applications.
- 3Miyamoto, T., Razavi, S., Derose, R., and Inoue, T. (2013) Synthesizing biomolecule-based boolean logic gates. ACS Synth. Biol. 2, 72– 82, DOI: 10.1021/sb3001112Google Scholar3Synthesizing Biomolecule-Based Boolean Logic GatesMiyamoto, Takafumi; Razavi, Shiva; DeRose, Robert; Inoue, TakanariACS Synthetic Biology (2013), 2 (2), 72-82CODEN: ASBCD6; ISSN:2161-5063. (American Chemical Society)A review. One fascinating recent avenue of study in the field of synthetic biol. is the creation of biomol.-based computers. The main components of a computing device consist of an arithmetic logic unit, the control unit, memory, and the input and output devices. Boolean logic gates are at the core of the operational machinery of these parts, and hence to make biocomputers a reality, biomol. logic gates become a necessity. Indeed, with the advent of more sophisticated biol. tools, both nucleic acid- and protein-based logic systems have been generated. These devices function in the context of either test tubes or living cells and yield highly specific outputs given a set of inputs. In this review, we discuss various types of biomol. logic gates that have been synthesized, with particular emphasis on recent developments that promise increased complexity of logic gate circuitry, improved computational speed, and potential clin. applications.
- 4Gaber, R., Lebar, T., Majerle, A., Šter, B., Dobnikar, A., Benčina, M., and Jerala, R. (2014) Designable DNA-binding domains enable construction of logic circuits in mammalian cells. Nat. Chem. Biol. 10, 203– 208, DOI: 10.1038/nchembio.1433Google Scholar4Designable DNA-binding domains enable construction of logic circuits in mammalian cellsGaber, Rok; Lebar, Tina; Majerle, Andreja; Ster, Branko; Dobnikar, Andrej; Bencina, Mojca; Jerala, RomanNature Chemical Biology (2014), 10 (3), 203-208CODEN: NCBABT; ISSN:1552-4450. (Nature Publishing Group)Electronic computer circuits consisting of a large no. of connected logic gates of the same type, such as NOR, can be easily fabricated and can implement any logic function. In contrast, designed genetic circuits must employ orthogonal information mediators owing to free diffusion within the cell. Combinatorial diversity and orthogonality can be provided by designable DNA- binding domains. Here, we employed the transcription activator-like repressors to optimize the construction of orthogonal functionally complete NOR gates to construct logic circuits. We used transient transfection to implement all 16 two-input logic functions from combinations of the same type of NOR gates within mammalian cells. Addnl., we present a genetic logic circuit where one input is used to select between an AND and OR function to process the data input using the same circuit. This demonstrates the potential of designable modular transcription factors for the construction of complex biol. information-processing devices.
- 5Lebar, T., Bezeljak, U., Golob, A., Jerala, M., Kadunc, L., Pirš, B., Stražar, M., Vučko, D., Zupančič, U., Benčina, M., Forstnerič, V., Gaber, R., Lonzarić, J., Majerle, A., Oblak, A., Smole, A., and Jerala, R. (2014) A bistable genetic switch based on designable DNA-binding domains. Nat. Commun. 5, 5007, DOI: 10.1038/ncomms6007Google Scholar5A bistable genetic switch based on designable DNA-binding domainsLebar, Tina; Bezeljak, Urban; Golob, Anja; Jerala, Miha; Kadunc, Lucija; Pirs, Bostjan; Strazar, Martin; Vucko, Dusan; Zupancic, Uros; Bencina, Mojca; Forstneric, Vida; Gaber, Rok; Lonzaric, Jan; Majerle, Andreja; Oblak, Alja; Smole, Anze; Jerala, RomanNature Communications (2014), 5 (), 5007CODEN: NCAOBW; ISSN:2041-1723. (Nature Publishing Group)Bistable switches are fundamental regulatory elements of complex systems, ranging from electronics to living cells. Designed genetic toggle switches have been constructed from pairs of natural transcriptional repressors wired to inhibit one another. The complexity of the engineered regulatory circuits can be increased using orthogonal transcriptional regulators based on designed DNA-binding domains. However, a mutual repressor-based toggle switch comprising DNA-binding domains of transcription-activator-like effectors (TALEs) did not support bistability in mammalian cells. Here, the challenge of engineering a bistable switch based on monomeric DNA-binding domains is solved via the introduction of a pos. feedback loop composed of activators based on the same TALE domains as their opposing repressors and competition for the same DNA operator site. This design introduces nonlinearity and results in epigenetic bistability. This principle could be used to employ other monomeric DNA-binding domains such as CRISPR for applications ranging from reprogramming cells to building digital biol. memory.
- 6Lebar, T., and Jerala, R. (2018) Designed transcriptional regulation in mammalian cells based on TALE- and CRISPR/dCas9, in Synthetic Biology Methods and Protocols, Methods in Molecular Biology, (Braman, J. C., Ed.), pp 191– 203, Humana Press, New York, NY.Google ScholarThere is no corresponding record for this reference.
- 7Qian, L. and Winfree, E. (2011) Scaling up digital circuit computation with DNA strand displacement cascades. Science 332, 1196– 1201, DOI: 10.1126/science.1200520Google Scholar7Scaling Up Digital Circuit Computation with DNA Strand Displacement CascadesQian, Lulu; Winfree, ErikScience (Washington, DC, United States) (2011), 332 (6034), 1196-1201CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)To construct sophisticated biochem. circuits from scratch, one needs to understand how simple the building blocks can be and how robustly such circuits can scale up. Using a simple DNA reaction mechanism based on a reversible strand displacement process, the authors exptl. demonstrated several digital logic circuits, culminating in a 4-bit square-root circuit that comprises 130 DNA strands. These multilayer circuits include thresholding and catalysis within every logical operation to perform digital signal restoration, which enables fast and reliable function in large circuits with roughly const. switching time and linear signal propagation delays. The design naturally incorporates other crucial elements for large-scale circuitry, such as general debugging tools, parallel circuit prepn., and an abstraction hierarchy supported by an automated circuit compiler.
- 8Yasuga, H., Kawano, R., Takinoue, M., Tsuji, Y., Osaki, T., Kamiya, K., Miki, N., and Takeuchi, S. (2016) Logic gate operation by DNA translocation through biological nanopores. PLoS One 11, e0149667 DOI: 10.1371/journal.pone.0149667Google Scholar8Logic gate operation by DNA translocation through biological nanoporesYasuga, Hiroki; Kawano, Ryuji; Takinoue, Masahiro; Tsuji, Yutaro; Osaki, Toshihisa; Kamiya, Koki; Miki, Norihisa; Takeuchi, ShojiPLoS One (2016), 11 (2), e0149667/1-e0149667/13CODEN: POLNCL; ISSN:1932-6203. (Public Library of Science)Logical operations using biol. mols., such as DNA computing or programmable diagnosis using DNA, have recently received attention. Challenges remain with respect to the development of such systems, including label-free output detection and the rapidity of operation. Here, we propose integration of biol. nanopores with DNA mols. for development of a logical operating system. We configured outputs "1" and "0" as single-stranded DNA (ssDNA) that is or is not translocated through a nanopore; unlabeled DNA was detected elec. A neg.-AND (NAND) operation was successfully conducted within approx. 10 min, which is rapid compared with previous studies using unlabeled DNA. In addn., this operation was executed in a four-droplet network. DNA mols. and assocd. information were transferred among droplets via biol. nanopores. This system would facilitate linking of mols. and electronic interfaces. Thus, it could be applied to mol. robotics, genetic engineering, and even medical diagnosis and treatment.
- 9Faulhammer, D., Cukras, A. R., Lipton, R. J., and Landweber, L. F. (2000) Molecular computation: RNA solutions to chess problems. Proc. Natl. Acad. Sci. U. S. A. 97, 1385– 1389, DOI: 10.1073/pnas.97.4.1385Google Scholar9Molecular computation: RNA solutions to chess problemsFaulhammer, Dirk; Cukras, Anthony R.; Lipton, Richard J.; Landweber, Laura F.Proceedings of the National Academy of Sciences of the United States of America (2000), 97 (4), 1385-1389CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)We have expanded the field of "DNA computers" to RNA and present a general approach for the soln. of satisfiability problems. As an example, we consider a variant of the "Knight problem," which asks generally what configurations of knights can one place on an n × n chess board such that no knight is attacking any other knight on the board. Using specific RNase digestion to manipulate strands of a 10-bit binary RNA library, we developed a mol. algorithm and applied it to a 3×3 chessboard as a 9-bit instance of this problem. Here, the nine spaces on the board correspond to nine "bits" or placeholders in a combinatorial RNA library. We recovered a set of "winning" mols. that describe solns. to this problem.
- 10Choi, J. H. and Ostermeier, M. (2015) Rational design of a fusion protein to exhibit disulfide-mediated logic gate behavior. ACS Synth. Biol. 4, 400– 406, DOI: 10.1021/sb500254gGoogle Scholar10Rational Design of a Fusion Protein to Exhibit Disulfide-Mediated Logic Gate BehaviorChoi, Jay H.; Ostermeier, MarcACS Synthetic Biology (2015), 4 (4), 400-406CODEN: ASBCD6; ISSN:2161-5063. (American Chemical Society)Synthetic cellular logic gates are primarily built from gene circuits owing to their inherent modularity. Single proteins can also possess logic gate functions and offer the potential to be simpler, quicker, and less dependent on cellular resources than gene circuits. However, the design of protein logic gates that are modular and integrate with other cellular components is a considerable challenge. As a step toward addressing this challenge, we describe the design, construction, and characterization of AND, ORN, and YES logic gates built by introducing disulfide bonds into RG13, a fusion of maltose binding protein and TEM-1 β-lactamase for which maltose is an allosteric activator of enzyme activity. We rationally designed these disulfide bonds to manipulate RG13's allosteric regulation mechanism such that the gating had maltose and reducing agents as input signals, and the gates could be toggled between different gating functions using redox agents, although some gates performed suboptimally.
- 11Fink, T., Lonzarić, J., Praznik, A., Plaper, T., Merljak, E., Leben, K., Jerala, N., Lebar, T., Strmšek, Ž., Lapenta, F., Benčina, M., and Jerala, R. (2019) Design of fast proteolysis-based signaling and logic circuits in mammalian cells. Nat. Chem. Biol. 15, 115– 122, DOI: 10.1038/s41589-018-0181-6Google Scholar11Design of fast proteolysis-based signaling and logic circuits in mammalian cellsFink, Tina; Lonzaric, Jan; Praznik, Arne; Plaper, Tjasa; Merljak, Estera; Leben, Katja; Jerala, Nina; Lebar, Tina; Strmsek, Ziga; Lapenta, Fabio; Bencina, Mojca; Jerala, RomanNature Chemical Biology (2019), 15 (2), 115-122CODEN: NCBABT; ISSN:1552-4450. (Nature Research)Cellular signal transduction is predominantly based on protein interactions and their post-translational modifications, which enable a fast response to input signals. Owing to difficulties in designing new unique protein-protein interactions, designed cellular logic has focused on transcriptional regulation; however, that process has a substantially slower response, because it requires transcription and translation. Here, the authors present de novo design of modular, scalable signaling pathways based on proteolysis and designed coiled coils (CC) and implemented in mammalian cells. A set of split proteases with highly specific orthogonal cleavage motifs was constructed and combined with strategically positioned cleavage sites and designed orthogonal CC dimerizing domains with tunable affinity for competitive displacement after proteolytic cleavage. This framework enabled the implementation of Boolean logic functions and signaling cascades in mammalian cells. The designed split-protease-cleavable orthogonal-CC-based (SPOC) logic circuits enable response to chem. or biol. signals within minutes rather than hours and should be useful for diverse medical and nonmedical applications.
- 12Moseley, F., Halámek, J., Kramer, F., Poghossian, A., Schöning, M. J., and Katz, E. (2014) An enzyme-based reversible CNOT logic gate realized in a flow system. Analyst 139, 1839– 1842, DOI: 10.1039/c4an00133hGoogle Scholar12An enzyme-based reversible CNOT logic gate realized in a flow systemMoseley, Fiona; Halamek, Jan; Kramer, Friederike; Poghossian, Arshak; Schoening, Michael J.; Katz, EvgenyAnalyst (Cambridge, United Kingdom) (2014), 139 (8), 1839-1842CODEN: ANALAO; ISSN:0003-2654. (Royal Society of Chemistry)An enzyme system organized in a flow device was used to mimic a reversible Controlled NOT (CNOT) gate with two input and two output signals. Reversible conversion of NAD+ and NADH cofactors was used to perform a XOR logic operation, while biocatalytic hydrolysis of p-nitrophenyl phosphate resulted in an Identity operation working in parallel. The first biomol. realization of a CNOT gate is promising for integration into complex biomol. networks and future biosensor/biomedical applications.
- 13Stein, V. and Alexandrov, K. (2014) Protease-based synthetic sensing and signal amplification. Proc. Natl. Acad. Sci. U. S. A. 111, 15934– 15939, DOI: 10.1073/pnas.1405220111Google Scholar13Protease-based synthetic sensing and signal amplificationStein, Viktor; Alexandrov, KirillProceedings of the National Academy of Sciences of the United States of America (2014), 111 (45), 15934-15939CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The bottom-up design of protein-based signaling networks is a key goal of synthetic biol.; yet, it remains elusive due to our inability to tailor-make signal transducers and receptors that can be readily compiled into defined signaling networks. Here, we report a generic approach for the construction of protein-based mol. switches based on artficially autoinhibited proteases. Using structure-guided design and directed protein evolution, we created signal transducers based on artificially autoinhibited proteases that can be activated following site-specific proteolysis and also demonstrate the modular design of an allosterically regulated protease receptor following recombination with an affinity clamp peptide receptor. Notably, the receptor's mode of action can be varied from >5-fold switch-OFF to >30-fold switch-ON solely by changing the length of the connecting linkers, demonstrating a high functional plasticity not previously obsd. in naturally occurring receptor systems. We also create an integrated signaling circuit based on two orthogonal autoinhibited protease units that can propagate and amplify mol. queues generated by the protease receptor. Finally, we present a generic two-component receptor architecture based on proximity-based activation of two autoinhibited proteases. Overall, the approach allows the design of protease-based signaling networks that, in principle, can be connected to any biol. process.
- 14Zhou, J., Arugula, M. A., Halámek, J., Pita, M., and Katz, E. (2009) Enzyme-based NAND and NOR logic gates with modular design. J. Phys. Chem. B 113, 16065– 16070, DOI: 10.1021/jp9079052Google Scholar14Enzyme-Based NAND and NOR Logic Gates with Modular DesignZhou, Jian; Arugula, Mary A.; Halamek, Jan; Pita, Marcos; Katz, EvgenyJournal of Physical Chemistry B (2009), 113 (49), 16065-16070CODEN: JPCBFK; ISSN:1520-6106. (American Chemical Society)The logic gates NAND/NOR were mimicked by enzyme biocatalyzed reactions activated by sucrose, maltose and phosphate. The subunits performing AND/OR Boolean logic operations were designed using maltose phosphorylase and cooperative work of invertase/amyloglucosidase, resp. Glucose produced as the output signal from the AND/OR subunits was applied as the input signal for the INVERTER gate composed of alc. dehydrogenase, glucose oxidase, microperoxidase-11, ethanol and NAD+, which generated the final output in the form of NADH inverting the logic signal from 0 to 1 or from 1 to 0. The final output signal was amplified by a self-promoting biocatalytic system. In order to fulfill the Boolean properties of associativity and commutativity in logic networks, the final NADH output signal was converted to the initial signals of maltose and phosphate, thus allowing assembling of the same std. units in concatenated sequences. The designed modular approach, signal amplification and conversion processes open the way toward complex logic networks composed of std. elements resembling electronic integrated circuitries.
- 15Gao, X. J., Chong, L. S., Kim, M. S., and Elowitz, M. B. (2018) Programmable protein circuits in living cells. Science 361, 1252– 1258, DOI: 10.1126/science.aat5062Google Scholar15Programmable protein circuits in living cellsGao, Xiaojing J.; Chong, Lucy S.; Kim, Matthew S.; Elowitz, Michael B.Science (Washington, DC, United States) (2018), 361 (6408), 1252-1258CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Synthetic protein-level circuits could enable engineering of powerful new cellular behaviors. Rational protein circuit design would be facilitated by a composable protein-protein regulation system in which individual protein components can regulate one another to create a variety of different circuit architectures. In this study, we show that engineered viral proteases can function as composable protein components, which can together implement a broad variety of circuit-level functions in mammalian cells. In this system, termed CHOMP (circuits of hacked orthogonal modular proteases), input proteases dock with and cleave target proteases to inhibit their function. These components can be connected to generate regulatory cascades, binary logic gates, and dynamic analog signal-processing functions. To demonstrate the utility of this system, we rationally designed a circuit that induces cell death in response to upstream activators of the Ras oncogene. Because CHOMP circuits can perform complex functions yet be encoded as single transcripts and delivered without genomic integration, they offer a scalable platform to facilitate protein circuit engineering for biotechnol. applications.
- 16Hilburger, C. E., Jacobs, M. L., Lewis, K. R., Peruzzi, J. A., and Kamat, N. P. (2019) Controlling Secretion in Artificial Cells with a Membrane and Gate. ACS Synth. Biol. 8, 1224– 1230, DOI: 10.1021/acssynbio.8b00435Google Scholar16Controlling Secretion in Artificial Cells with a Membrane AND GateHilburger, Claire E.; Jacobs, Miranda L.; Lewis, Kamryn R.; Peruzzi, Justin A.; Kamat, Neha P.ACS Synthetic Biology (2019), 8 (6), 1224-1230CODEN: ASBCD6; ISSN:2161-5063. (American Chemical Society)The assembly of channel proteins into vesicle membranes is a useful strategy to control activities of vesicle-based systems. Here, the authors developed a membrane AND gate that responds to both a fatty acid and a pore-forming channel protein to induce the release of encapsulated cargo. The authors explored how membrane compn. affects the functional assembly of α-hemolysin into phospholipid vesicles as a function of oleic acid content and α-hemolysin concn. The authors then showed that the authors could induce α-hemolysin assembly when the authors added oleic acid micelles to a specific compn. of phospholipid vesicles. Finally, the authors demonstrated that the membrane AND gate could be coupled to a gene expression system. The study provides a new method to control the temporal dynamics of vesicle permeability by controlling when the functional assembly of a channel protein into synthetic vesicles occurs. Furthermore, a membrane AND gate that utilizes membrane-assocg. biomols. introduces a new way to implement Boolean logic that should complement genetic logic circuits and ultimately enhance the capabilities of artificial cellular systems.
- 17Adamala, K. P., Martin-Alarcon, D. A., Guthrie-Honea, K. R., and Boyden, E. S. (2017) Engineering genetic circuit interactions within and between synthetic minimal cells. Nat. Chem. 9, 431– 439, DOI: 10.1038/nchem.2644Google Scholar17Engineering 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.
- 18Einfalt, T., Goers, R., Dinu, I. A., Najer, A., Spulber, M., Onaca-Fischer, O., and Palivan, C. G. (2015) Stimuli-Triggered Activity of Nanoreactors by Biomimetic Engineering Polymer Membranes. Nano Lett. 15, 7596– 7603, DOI: 10.1021/acs.nanolett.5b03386Google Scholar18Stimuli-Triggered Activity of Nanoreactors by Biomimetic Engineering Polymer MembranesEinfalt, Tomaz; Goers, Roland; Dinu, Ionel Adrian; Najer, Adrian; Spulber, Mariana; Onaca-Fischer, Ozana; Palivan, Cornelia G.Nano Letters (2015), 15 (11), 7596-7603CODEN: NALEFD; ISSN:1530-6984. (American Chemical Society)The development of advanced stimuli-responsive systems for medicine, catalysis, or technol. requires compartmentalized reaction spaces with triggered activity. Only very few stimuli-responsive systems preserve the compartment architecture, and none allows a triggered activity in situ. We present here a biomimetic strategy to mol. transmembrane transport by engineering synthetic membranes equipped with channel proteins so that they are stimuli-responsive. Nanoreactors with triggered activity were designed by simultaneously encapsulating an enzyme inside polymer compartments, and inserting protein "gates" in the membrane. The outer membrane protein F (OmpF) porin was chem. modified with a pH-responsive mol. cap to serve as "gate" producing pH-driven mol. flow through the membrane and control the in situ enzymic activity. This strategy provides complex reaction spaces necessary in "smart" medicine and for biomimetic engineering of artificial cells.
- 19Einfalt, T., Witzigmann, D., Edlinger, C., Sieber, S., Goers, R., Najer, A., Spulber, M., Onaca-Fischer, O., Huwyler, J., and Palivan, C. G. (2018) Biomimetic artificial organelles with in vitro and in vivo activity triggered by reduction in microenvironment. Nat. Commun. 9, 1127, DOI: 10.1038/s41467-018-03560-xGoogle Scholar19Biomimetic artificial organelles with in vitro and in vivo activity triggered by reduction in microenvironmentEinfalt T; Edlinger C; Goers R; Najer A; Spulber M; Onaca-Fischer O; Palivan C G; Einfalt T; Witzigmann D; Sieber S; Huwyler J; Goers RNature communications (2018), 9 (1), 1127 ISSN:.Despite tremendous efforts to develop stimuli-responsive enzyme delivery systems, their efficacy has been mostly limited to in vitro applications. Here we introduce, by using an approach of combining biomolecules with artificial compartments, a biomimetic strategy to create artificial organelles (AOs) as cellular implants, with endogenous stimuli-triggered enzymatic activity. AOs are produced by inserting protein gates in the membrane of polymersomes containing horseradish peroxidase enzymes selected as a model for natures own enzymes involved in the redox homoeostasis. The inserted protein gates are engineered by attaching molecular caps to genetically modified channel porins in order to induce redox-responsive control of the molecular flow through the membrane. AOs preserve their structure and are activated by intracellular glutathione levels in vitro. Importantly, our biomimetic AOs are functional in vivo in zebrafish embryos, which demonstrates the feasibility of using AOs as cellular implants in living organisms. This opens new perspectives for patient-oriented protein therapy.
- 20van Meer, G., Voelker, D. R., and Feigenson, G. W. (2008) Membrane lipids: where they are and how they behave. Nat. Rev. Mol. Cell Biol. 9, 112– 124, DOI: 10.1038/nrm2330Google Scholar20Membrane lipids: where they are and how they behavevan Meer, Gerrit; Voelker, Dennis R.; Feigenson, Gerald W.Nature Reviews Molecular Cell Biology (2008), 9 (2), 112-124CODEN: NRMCBP; ISSN:1471-0072. (Nature Publishing Group)A review. Throughout the biol. world, a 30 Å hydrophobic film typically delimits the environments that serve as the margin between life and death for individual cells. Biochem. and biophys. findings have provided a detailed model of the compn. and structure of membranes, which includes levels of dynamic organization both across the lipid bilayer (lipid asymmetry) and in the lateral dimension (lipid domains) of membranes. How do cells apply anabolic and catabolic enzymes, translocases, and transporters, plus the intrinsic phys. phase behavior of lipids and their interactions with membrane proteins, to create the unique compns. and multiple functionalities of their individual membranes.
- 21Golynskiy, M. V., Koay, M. S., Vinkenborg, J. L., and Merkx, M. (2011) Engineering protein switches: sensors, regulators, and spare parts for biology and biotechnology. ChemBioChem 12, 353– 361, DOI: 10.1002/cbic.201000642Google Scholar21Engineering Protein Switches: Sensors, Regulators, and Spare Parts for Biology and BiotechnologyGolynskiy, Misha V.; Koay, Melissa S.; Vinkenborg, Jan L.; Merkx, MaartenChemBioChem (2011), 12 (3), 353-361CODEN: CBCHFX; ISSN:1439-4227. (Wiley-VCH Verlag GmbH & Co. KGaA)A review. The authors review the underlying engineering concepts for constructing protein switches, most of which are inspired by or reminiscent of natural examples. The authors focus on those strategies that have the potential to be generally applicable, and discuss them according to their mechanism of action in order of increasing modularity and decreasing domain integration. Finally, the authors identify the opportunities and challenges assocd. with these different engineering concepts and discuss future perspectives.
- 22Arosio, D., Ricci, F., Marchetti, L., Gualdani, R., Albertazzi, L., and Beltram, F. (2010) Simultaneous intracellular chloride and pH measurements using a GFP-based sensor. Nat. Methods 7, 516– 518, DOI: 10.1038/nmeth.1471Google Scholar22Simultaneous intracellular chloride and pH measurements using a GFP-based sensorArosio, Daniele; Ricci, Fernanda; Marchetti, Laura; Gualdani, Roberta; Albertazzi, Lorenzo; Beltram, FabioNature Methods (2010), 7 (7), 516-518CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)Chloride and protons perform important closely related roles in many cellular responses. Here we developed a ratiometric biosensor, ClopHensor, based on a highly chloride-sensitive Aequorea victoria GFP variant that is suited for the combined real-time optical detection of pH changes and chloride fluxes in live cells. We detected high chloride concn. in large dense-core exocytosis granules by targeting ClopHensor to these intracellular compartments.
- 23Dal Peraro, M. and van der Goot, F. G. (2016) Pore-forming toxins: ancient, but never really out of fashion. Nat. Rev. Microbiol. 14, 77– 92, DOI: 10.1038/nrmicro.2015.3Google Scholar23Pore-forming toxins: ancient, but never really out of fashionDal Peraro Matteo; van der Goot F GisouNature reviews. Microbiology (2016), 14 (2), 77-92 ISSN:.Pore-forming toxins (PFTs) are virulence factors produced by many pathogenic bacteria and have long fascinated structural biologists, microbiologists and immunologists. Interestingly, pore-forming proteins with remarkably similar structures to PFTs are found in vertebrates and constitute part of their immune system. Recently, structural studies of several PFTs have provided important mechanistic insights into the metamorphosis of PFTs from soluble inactive monomers to cytolytic transmembrane assemblies. In this Review, we discuss the diverse pore architectures and membrane insertion mechanisms that have been revealed by these studies, and we consider how these features contribute to binding specificity for different membrane targets. Finally, we explore the potential of these structural insights to enable the development of novel therapeutic strategies that would prevent both the establishment of bacterial resistance and an excessive immune response.
- 24Anderluh, G. and Lakey, J. H. (2008) Disparate proteins use similar architectures to damage membranes. Trends Biochem. Sci. 33, 482– 490, DOI: 10.1016/j.tibs.2008.07.004Google Scholar24Disparate proteins use similar architectures to damage membranesAnderluh, Gregor; Lakey, Jeremy H.Trends in Biochemical Sciences (2008), 33 (10), 482-490CODEN: TBSCDB; ISSN:0968-0004. (Elsevier B.V.)A review. Membrane disruption can efficiently alter cellular function; indeed, pore-forming toxins (PFTs) are well known as important bacterial virulence factors. However, recent data have revealed that structures similar to those found in PFTs are found in membrane active proteins across disparate phyla. Many similarities can be identified only at the 3D-structural level. Of note, domains found in membrane-attack complex proteins of complement and perforin (MACPF) resemble cholesterol-dependent cytolysins from Gram-pos. bacteria, and the Bcl family of apoptosis regulators share similar architectures with Escherichia coli pore-forming colicins. These and other correlations provide considerable help in understanding the structural requirements for membrane binding and pore formation.
- 25Laventie, B., Potrich, C., Atmanène, C., Saleh, M., Joubert, O., Viero, G., Bachmeyer, C., Antonini, V., Mancini, I., Cianferani-Sanglier, S., Keller, D., Colin, D. A., Bourcier, T., Anderluh, G., van Dorsselaer, A., Dalla Serra, M., and Prévost, G. (2013) p-Sulfonato-calix[n]arenes inhibit staphylococcal bicomponent leukotoxins by supramolecular interactions. Biochem. J. 450, 559– 571, DOI: 10.1042/BJ20121628Google Scholar25p-Sulfonato-calix[n]arenes inhibit staphylococcal bicomponent leukotoxins by supramolecular interactionsLaventie, Benoit-Joseph; Potrich, Cristina; Atmanene, Cedric; Saleh, Maher; Joubert, Olivier; Viero, Gabriella; Bachmeyer, Christoph; Antonini, Valeria; Mancini, Ines; Cianferani-Sanglier, Sarah; Keller, Daniel; Colin, Didier A.; Bourcier, Tristan; Anderluh, Gregor; van Dorsselaer, Alain; Dalla Serra, Mauro; Prevost, GillesBiochemical Journal (2013), 450 (3), 559-571CODEN: BIJOAK; ISSN:0264-6021. (Portland Press Ltd.)PVL (Panton-Valentine leukocidin) and other Staphylococcus aureus β-stranded pore-forming toxins are important virulence factors involved in various pathologies that are often necrotizing. The present study characterized leukotoxin inhibition by selected SCns (p-sulfonato-calix[n]arenes): SC4, SC6 and SC8. These chems. have no toxic effects on human erythrocytes or neutrophils, and some are able to inhibit both the activity of and the cell lysis by leukotoxins in a dose-dependent manner. Depending on the type of leukotoxins and SCns, flow cytometry revealed IC50 values of 6-22 μM for Ca2+ activation and of 2-50 μM for cell lysis. SCns were obsd. to affect membrane binding of class S proteins responsible for cell specificity. Electrospray MS and surface plasmon resonance established supramol. interactions (1:1 stoichiometry) between SCns and class S proteins in soln., but not class F proteins. The membrane-binding affinity of S proteins was Kd=0.07-6.2 nM. The binding ability was completely abolished by SCns at different concns. according to the no. of benzenes (30-300 μM; SC8>SC6»SC4). The inhibitory properties of SCns were also obsd. in vivo in a rabbit model of PVL-induced endophthalmitis. These calixarenes may represent new therapeutic avenues aimed at minimizing inflammatory reactions and necrosis due to certain virulence factors.
- 26Booth, M. J., Schild, V. R., Graham, A. D., Olof, S. N., and Bayley, H. (2016) Light-activated communication in synthetic tissues. Sci. Adv. 2, e1600056 DOI: 10.1126/sciadv.1600056Google Scholar26Light-activated communication in synthetic tissuesBooth, Michael J.; Schild, Vanessa Restrepo; Graham, Alexander D.; Olof, Sam N.; Bayley, HaganScience Advances (2016), 2 (4), e1600056/1-e1600056/11CODEN: SACDAF; ISSN:2375-2548. (American Association for the Advancement of Science)We have previously used three-dimensional (3D) printing to prep. tissue-like materials in which picoliter aq. compartments are sepd. by lipid bilayers. These printed droplets are elaborated into synthetic cells by using a tightly regulated in vitro transcription/translation system. A light-activated DNA promoter has been developed that can be used to turn on the expression of any gene within the synthetic cells. We used light activation to express protein pores in 3D-printed patterns within synthetic tissues. The pores are incorporated into specific bilayer interfaces and thereby mediate rapid, directional elec. communication between subsets of cells. Accordingly, we have developed a functional mimic of neuronal transmission that can be controlled in a precise way.
- 27Provoda, C. J., Stier, E. M., and Lee, K. D. (2003) Tumor cell killing enabled by listeriolysin O-liposome-mediated delivery of the protein toxin gelonin. J. Biol. Chem. 278, 35102– 35108, DOI: 10.1074/jbc.M305411200Google Scholar27Tumor cell killing enabled by Listeriolysin O-liposome-mediated delivery of the protein toxin geloninProvoda, Chester J.; Stier, Ethan M.; Lee, Kyung-DallJournal of Biological Chemistry (2003), 278 (37), 35102-35108CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)Gelonin is a type I plant toxin that has potential as an effective anti-tumor agent by virtue of its enzymic capacity to inactivate ribosomes and arrest protein synthesis, thereby effectively limiting the growth of cancer cells. Being a hydrophilic macromol., however, gelonin has limited access to its target subcellular compartment, the cytosol; it is effectively plasma membrane-impermeant and subject to rapid degrdn. within endosomes and lysosomes upon cellular uptake as it lacks the membrane-translocating capability that is typically provided by a disulfide-linked B polypeptide found in the type II toxins (e.g. ricin). These inherent characteristics generate the need for the development of a specialized cytosolic delivery strategy for gelonin as an effective anti-tumor therapeutic agent. Here we describe an efficient means of delivering gelonin to the cytosol of B16 melanoma cells. Gelonin was co-encapsulated inside pH-sensitive liposomes with listeriolysin O, the pore-forming protein that mediates escape of the intracellular pathogen Listeria monocytogenes from the endosome into the cytosol. In in vitro expts., co-encapsulated listeriolysin O enabled liposomal gelonin-mediated B16 cell killing with a gelonin IC50 of ∼0.1 nM with an extreme efficiency requiring an incubation time of only 1 h. By contrast, cells treated with equiv. concns. of unencapsulated gelonin or gelonin encapsulated alone in pH-sensitive liposomes exhibited no detectable cytotoxicity. Moreover, treatment by direct intratumor injection into s.c. solid tumors of B16 melanoma in a mouse model showed that pH-sensitive liposomes contg. both listeriolysin O and gelonin were more effective than control formulations in curtailing tumor growth rates.
- 28Robertson, J. W. F., Rodrigues, C. G., Stanford, V. M., Rubinson, K. A., Krasilnikov, O. V., and Kasianowicz, J. J. (2007) Single-molecule mass spectrometry in solution using a solitary nanopore. Proc. Natl. Acad. Sci. U. S. A. 104, 8207– 8211, DOI: 10.1073/pnas.0611085104Google Scholar28Single-molecule mass spectrometry in solution using a solitary nanoporeRobertson, Joseph W. F.; Rodrigues, Claudio G.; Stanford, Vincent M.; Rubinson, Kenneth A.; Krasilnikov, Oleg V.; Kasianowicz, John J.Proceedings of the National Academy of Sciences of the United States of America (2007), 104 (20), 8207-8211CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The authors introduce a 2-dimensional method for mass spectrometry in soln. that is based on the interaction between a nanometer-scale pore and analytes. As an example, poly(ethylene glycol) mols. that enter a single α-hemolysin pore cause distinct mass-dependent conductance states with characteristic mean residence times. The conductance-based mass spectrum clearly resolves the repeat unit of ethylene glycol, and the mean residence time increases monotonically with the poly(ethylene glycol) mass. This technique could prove useful for the real-time characterization of mols. in soln.
- 29Clarke, J., Wu, H., Jayasinghe, L., Patel, A., Reid, S., and Bayley, H. (2009) Continuous base identification for single-molecule nanopore DNA sequencing. Nat. Nanotechnol. 4, 265– 270, DOI: 10.1038/nnano.2009.12Google Scholar29Continuous base identification for single-molecule nanopore DNA sequencingClarke, James; Wu, Hai-Chen; Jayasinghe, Lakmal; Patel, Alpesh; Reid, Stuart; Bayley, HaganNature Nanotechnology (2009), 4 (4), 265-270CODEN: NNAABX; ISSN:1748-3387. (Nature Publishing Group)A single-mol. method for sequencing DNA that does not require fluorescent labeling could reduce costs and increase sequencing speeds. An exonuclease enzyme might be used to cleave individual nucleotide mols. from the DNA, and when coupled to an appropriate detection system, these nucleotides could be identified in the correct order. Here, the authors show that a protein nanopore with a covalently attached adapter mol. can continuously identify unlabeled nucleoside 5'-monophosphate mols. with accuracies averaging 99.8%. Methylated cytosine can also be distinguished from the four std. DNA bases: guanine, adenine, thymine and cytosine. The operating conditions are compatible with the exonuclease, and the kinetic data show that the nucleotides have a high probability of translocation through the nanopore and, therefore, of not being registered twice. This highly accurate tool is suitable for integration into a system for sequencing nucleic acids and for analyzing epigenetic modifications.
- 30Wang, Y., Montana, V., Grubišić, V., Stout, R. F., Parpura, V., and Gu, L. Q. (2015) Nanopore sensing of botulinum toxin type B by discriminating an enzymatically cleaved peptide from a synaptic protein synaptobrevin 2 derivative. ACS Appl. Mater. Interfaces 7, 184– 192, DOI: 10.1021/am5056596Google Scholar30Nanopore Sensing of Botulinum Toxin Type B by Discriminating an Enzymatically Cleaved Peptide from a Synaptic Protein Synaptobrevin 2 DerivativeWang, Yong; Montana, Vedrana; Grubisic, Vladimir; Stout, Randy F.; Parpura, Vladimir; Gu, Li-QunACS Applied Materials & Interfaces (2015), 7 (1), 184-192CODEN: AAMICK; ISSN:1944-8244. (American Chemical Society)Botulinum neurotoxins (BoNTs) are the most lethal toxin known to human. Biodefense requires early and rapid detection of BoNTs. Traditionally, BoNTs can be detected by looking for signs of botulism in mice that receive an injection of human material, serum or stool. While the living animal assay remains the most sensitive approach, it is costly, slow and assocd. with legal and ethical constrains. Various biochem., optical and mech. methods have been developed for BoNTs detection with improved speed, but with lesser sensitivity. Here, we report a novel nanopore-based BoNT type B (BoNT-B) sensor that monitors the toxin's enzymic activity on its substrate, a recombinant synaptic protein synaptobrevin 2 deriv. By analyzing the modulation of the pore current caused by the specific BoNT-B-digested peptide as a marker, the presence of BoNT-B at a subnanomolar concn. was identified within minutes. The nanopore detector would fill the niche for a much needed rapid and highly sensitive detection of neurotoxins, and provide an excellent system to explore biophys. mechanisms for biopolymer transportation.
- 31Wang, Y., Zheng, D., Tan, Q., Wang, M., and Gu, L. (2011) Nanopore-based detection of circulating microRNAs in lung cancer patients. Nat. Nanotechnol. 6, 668– 674, DOI: 10.1038/nnano.2011.147Google Scholar31Nanopore-based detection of circulating microRNAs in lung cancer patientsWang, Yong; Zheng, Da-Li; Tan, Qiu-Lin; Wang, Michael X.; Gu, Li-QunNature Nanotechnology (2011), 6 (10), 668-674CODEN: NNAABX; ISSN:1748-3387. (Nature Publishing Group)MicroRNAs are short RNA mols. that regulate gene expression, and have been investigated as potential biomarkers because their expression levels are correlated with various diseases. However, detecting microRNAs in the bloodstream remains difficult because current methods are not sufficiently selective or sensitive. Here, we show that a nanopore sensor based on the α-haemolysin protein can selectively detect microRNAs at the single mol. level in plasma samples from lung cancer patients without the need for labels or amplification of the microRNA. The sensor, which uses a programmable oligonucleotide probe to generate a target-specific signature signal, can quantify subpicomolar levels of cancer-assocd. microRNAs and can distinguish single-nucleotide differences between microRNA family members. This approach is potentially useful for quant. microRNA detection, the discovery of disease markers and non-invasive early diagnosis of cancer.
- 32Braha, O., Bayley, H., Gu, L. Q., Zhou, L., Lu, X., and Cheley, S. (2000) Simultaneous stochastic sensing of divalent metal ions. Nat. Biotechnol. 18, 1005– 1007, DOI: 10.1038/79275Google Scholar32Simultaneous stochastic sensing of divalent metal ionsBraha, Orit; Gu, Lin-Qun; Zhou, Li; Lu, Xiaofeng; Cheley, Stephen; Bayley, HaganNature Biotechnology (2000), 18 (9), 1005-1007CODEN: NABIF9; ISSN:1087-0156. (Nature America Inc.)Stochastic sensing is an emerging anal. technique that relies upon single-mol. detection. Transmembrane pores, into which binding sites for analytes have been placed by genetic engineering, have been developed as stochastic sensing elements. Reversible occupation of an engineered binding site modulates the ionic current passing through a pore in a transmembrane potential and thereby provides both the concn. of an analyte and, through a characteristic signature, its identity. Here, we show that the concns. of two or more divalent metal ions in soln. can be detd. simultaneously with a single sensor element. Further, the sensor element can be permanently calibrated without a detailed understanding of the kinetics of interaction of the metal ions with the engineered pore.
- 33Villar, G., Graham, A. D., and Bayley, H. (2013) A tissue-like printed material. Science 340, 48– 52, DOI: 10.1126/science.1229495Google Scholar33A Tissue-Like Printed MaterialVillar, Gabriel; Graham, Alexander D.; Bayley, HaganScience (Washington, DC, United States) (2013), 340 (6128), 48-52CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Living cells communicate and cooperate to produce the emergent properties of tissues. Synthetic mimics of cells, such as liposomes, are typically incapable of cooperation and therefore cannot readily display sophisticated collective behavior. We printed tens of thousands of picoliter aq. droplets that become joined by single lipid bilayers to form a cohesive material with cooperating compartments. Three-dimensional structures can be built with heterologous droplets in software-defined arrangements. The droplet networks can be functionalized with membrane proteins; for example, to allow rapid elec. communication along a specific path. The networks can also be programmed by osmolarity gradients to fold into otherwise unattainable designed structures. Printed droplet networks might be interfaced with tissues, used as tissue engineering substrates, or developed as mimics of living tissue.
- 34Elani, Y., Law, R. V., and Ces, O. (2014) Vesicle-based artificial cells as chemical microreactors with spatially segregated reaction pathways. Nat. Commun. 5, 5305, DOI: 10.1038/ncomms6305Google Scholar34Vesicle-based artificial cells as chemical microreactors with spatially segregated reaction pathwaysElani, Yuval; Law, Robert V.; Ces, OscarNature Communications (2014), 5 (), 5305CODEN: NCAOBW; ISSN:2041-1723. (Nature Publishing Group)In the discipline of bottom-up synthetic biol., vesicles define the boundaries of artificial cells and are increasingly being used as biochem. microreactors operating in physiol. environments. As the field matures, there is a need to compartmentalize processes in different spatial localities within vesicles, and for these processes to interact with one another. Here we address this by designing and constructing multi-compartment vesicles within which an engineered multi-step enzymic pathway is carried out. The individual steps are isolated in distinct compartments, and their products traverse into adjacent compartments with the aid of transmembrane protein pores, initiating subsequent steps. Thus, an engineered signalling cascade is recreated in an artificial cellular system. Importantly, by allowing different steps of a chem. pathway to be sepd. in space, this platform bridges the gap between table-top chem. and chem. that is performed within vesicles.
- 35Hamon, M. A., Ribet, D., Stavru, F., and Cossart, P. (2012) Listeriolysin O: the Swiss army knife of Listeria. Trends Microbiol. 20, 360– 368, DOI: 10.1016/j.tim.2012.04.006Google Scholar35Listeriolysin O: the Swiss army knife of ListeriaHamon, Melanie Anne; Ribet, David; Stavru, Fabrizia; Cossart, PascaleTrends in Microbiology (2012), 20 (8), 360-368CODEN: TRMIEA; ISSN:0966-842X. (Elsevier Ltd.)A review. Listeriolysin O (LLO) is a toxin produced by Listeria monocytogenes, an opportunistic bacterial pathogen responsible for the disease listeriosis. This disease starts with the ingestion of contaminated foods and mainly affects immunocompromised individuals, newborns, and pregnant women. In the lab., L. monocytogenes is used as a model organism to study processes such as cell invasion, intracellular survival, and cell-to-cell spreading, as this Gram-pos. bacterium has evolved elaborate mol. strategies to subvert host cell functions. LLO is a major virulence factor originally shown to be crucial for bacterial escape from the internalization vacuole after entry into cells. However, recent studies are revisiting the role of LLO during infection and are revealing new insights into the action of LLO, in particular before bacterial entry. These latest findings along with their impact on the infectious process will be discussed.
- 36Köster, S., van Pee, K., Hudel, M., Leustik, M., Rhinow, D., Kühlbrandt, W., Chakraborty, T., and Yildiz, O. (2014) Crystal structure of listeriolysin O reveals molecular details of oligomerization and pore formation. Nat. Commun. 5, 3690, DOI: 10.1038/ncomms4690Google Scholar36Crystal structure of listeriolysin O reveals molecular details of oligomerization and pore formationKoster Stefan; van Pee Katharina; Rhinow Daniel; Kuhlbrandt Werner; Yildiz Ozkan; Hudel Martina; Leustik Martin; Chakraborty TrinadNature communications (2014), 5 (), 3690 ISSN:.Listeriolysin O (LLO) is an essential virulence factor of Listeria monocytogenes that causes listeriosis. Listeria monocytogenes owes its ability to live within cells to the pH- and temperature-dependent pore-forming activity of LLO, which is unique among cholesterol-dependent cytolysins. LLO enables the bacteria to cross the phagosomal membrane and is also involved in activation of cellular processes, including the modulation of gene expression or intracellular Ca(2+) oscillations. Neither the pore-forming mechanism nor the mechanisms triggering the signalling processes in the host cell are known in detail. Here, we report the crystal structure of LLO, in which we identified regions important for oligomerization and pore formation. Mutants were characterized by determining their haemolytic and Ca(2+) uptake activity. We analysed the pore formation of LLO and its variants on erythrocyte ghosts by electron microscopy and show that pore formation requires precise interface interactions during toxin oligomerization on the membrane.
- 37Ruan, Y., Rezelj, S., Bedina Zavec, A., Anderluh, G., and Scheuring, S. (2016) Listeriolysin O membrane damaging activity involves arc formation and lineaction - implication for Listeria monocytogenes escape from phagocytic vacuole. PLoS Pathog. 12, e1005597 DOI: 10.1371/journal.ppat.1005597Google Scholar37Listeriolysin O membrane damaging activity involves arc formation and lineaction - implication for Listeria monocytogenes escape from phagocytic vacuoleRuan, Yi; Rezelj, Sasa; Zavec, Apolonija Bedina; Anderluh, Gregor; Scheuring, SimonPLoS Pathogens (2016), 12 (4), e1005597/1-e1005597/18CODEN: PPLACN; ISSN:1553-7374. (Public Library of Science)Listeriolysin-O (LLO) plays a crucial role during infection by Listeria monocytogenes. It enables escape of bacteria from phagocytic vacuole, which is the basis for its spread to other cells and tissues. It is not clear how LLO acts at phagosomal membranes to allow bacterial escape. The mechanism of action of LLO remains poorly understood, probably due to unavailability of suitable exptl. tools that could monitor LLO membrane disruptive activity in real time. Here, we used high-speed at. force microscopy (HS-AFM) featuring high spatio-temporal resoln. on model membranes and optical microscopy on giant unilamellar vesicles (GUVs) to investigate LLO activity. We analyze the assembly kinetics of toxin oligomers, the prepore-to-pore transition dynamics and the membrane disruption in real time. We reveal that LLO toxin efficiency and mode of action as a membrane-disrupting agent varies strongly depending on the membrane cholesterol concn. and the environmental pH. We discovered that LLO is able to form arc pores as well as damage lipid membranes as a lineactant, and this leads to large-scale membrane defects. These results altogether provide a mechanistic basis of how large-scale membrane disruption leads to release of Listeria from the phagocytic vacuole in the cellular context.
- 38Podobnik, M., Marchioretto, M., Zanetti, M., Bavdek, A., Kisovec, M., Cajnko, M. M., Lunelli, L., Serra, M. D., and Anderluh, G. (2015) Plasticity of lysteriolysin O pores and its regulation by pH and unique histidine. Sci. Rep. 5, 9623, DOI: 10.1038/srep09623Google Scholar38Plasticity of Listeriolysin O Pores and its Regulation by pH and Unique HistidinePodobnik, Marjetka; Marchioretto, Marta; Zanetti, Manuela; Bavdek, Andrej; Kisovec, Matic; Cajnko, Misa Mojca; Lunelli, Lorenzo; Dalla Serra, Mauro; Anderluh, GregorScientific Reports (2015), 5 (), 9623/1-9623/10CODEN: SRCEC3; ISSN:2045-2322. (Nature Publishing Group)Pore formation of cellular membranes is an ancient mechanism of bacterial pathogenesis that allows efficient damaging of target cells. Several mechanisms have been described, however, relatively little is known about the assembly and properties of pores. Listeriolysin O (LLO) is a pH-regulated cholesterol-dependent cytolysin from the intracellular pathogen Listeria monocytogenes, which forms transmembrane β-barrel pores. Here we report that the assembly of LLO pores is rapid and efficient irresp. of pH. While pore diams. at the membrane surface are comparable at either pH 5.5 or 7.4, the distribution of pore conductances is significantly pH-dependent. This is directed by the unique residue H311, which is also important for the conformational stability of the LLO monomer and the rate of pore formation. The functional pores exhibit variations in height profiles and can reconfigure significantly by merging to other full pores or arcs. Our results indicate significant plasticity of large β-barrel pores, controlled by environmental cues like pH.
- 39Mulvihill, E., Van Pee, K., Mari, S. A., Müller, D. J., and Yildiz, Ö. (2015) Directly observing the lipid-dependent self-assembly and pore-forming mechanism of the cytolytic toxin listeriolysin O. Nano Lett. 15, 6965– 6973, DOI: 10.1021/acs.nanolett.5b02963Google Scholar39Directly Observing the Lipid-Dependent Self-Assembly and Pore-Forming Mechanism of the Cytolytic Toxin Listeriolysin OMulvihill Estefania; Mari Stefania A; Muller Daniel J; van Pee Katharina; Yildiz OzkanNano letters (2015), 15 (10), 6965-73 ISSN:.Listeriolysin O (LLO) is the major virulence factor of Listeria monocytogenes and a member of the cholesterol-dependent cytolysin (CDC) family. Gram-positive pathogenic bacteria produce water-soluble CDC monomers that bind cholesterol-dependent to the lipid membrane of the attacked cell or of the phagosome, oligomerize into prepores, and insert into the membrane to form transmembrane pores. However, the mechanisms guiding LLO toward pore formation are poorly understood. Using electron microscopy and time-lapse atomic force microscopy, we show that wild-type LLO binds to membranes, depending on the presence of cholesterol and other lipids. LLO oligomerizes into arc- or slit-shaped assemblies, which merge into complete rings. All three oligomeric assemblies can form transmembrane pores, and their efficiency to form pores depends on the cholesterol and the phospholipid composition of the membrane. Furthermore, the dynamic fusion of arcs, slits, and rings into larger rings and their formation of transmembrane pores does not involve a height difference between prepore and pore. Our results reveal new insights into the pore-forming mechanism and introduce a dynamic model of pore formation by LLO and other CDC pore-forming toxins.
- 40Kisovec, M., Rezelj, S., Knap, P., Cajnko, M., Caserman, S., Flašker, A., Žnidaršič, N., Repič, M., Mavri, J., Ruan, Y., Scheuring, S., Podobnik, M., and Anderluh, G. (2017) Engineering a pH responsive pore forming protein. Sci. Rep. 7, 42231, DOI: 10.1038/srep42231Google Scholar40Engineering a pH responsive pore forming proteinKisovec, Matic; Rezelj, Sasa; Knap, Primoz; Cajnko, Misa Mojca; Caserman, Simon; Flasker, Ajda; Znidarsic, Nada; Repic, Matej; Mavri, Janez; Ruan, Yi; Scheuring, Simon; Podobnik, Marjetka; Anderluh, GregorScientific Reports (2017), 7 (), 42231CODEN: SRCEC3; ISSN:2045-2322. (Nature Publishing Group)Listeriolysin O (LLO) is a cytolysin capable of forming pores in cholesterol-rich lipid membranes of host cells. It is conveniently suited for engineering a pH-governed responsiveness, due to a pH sensor identified in its structure that was shown before to affect its stability. Here we introduced a new level of control of its hemolytic activity by making a variant with hemolytic activity that was pH-dependent. Based on detailed structural anal. coupled with mol. dynamics and mutational anal., we found that the bulky side chain of Tyr406 allosterically affects the pH sensor. Mol. dynamics simulation further suggested which other amino acid residues may also allosterically influence the pH-sensor. LLO was engineered to the point where it can, in a pH-regulated manner, perforate artificial and cellular membranes. The single mutant Tyr406Ala bound to membranes and oligomerized similarly to the wild-type LLO, however, the final membrane insertion step was pH-affected by the introduced mutation. We show that the mutant toxin can be activated at the surface of artificial membranes or living cells by a single wash with slightly acidic pH buffer. Y406A mutant has a high potential in development of novel nanobiotechnol. applications such as controlled release of substances or as a sensor of environmental pH.
- 41Dreier, B., and Plückthun, A. (2011) Ribosome Display: A Technology for Selecting and Evolving Proteins from Large Libraries, in PCR Protocols. Methods in Molecular Biology (Methods and Protocols), (Park, D. J., Ed.), pp 283– 306, Humana Press, Totowa, NJ.Google ScholarThere is no corresponding record for this reference.
- 42Binz, H. K., Amstutz, P., Kohl, A., Stumpp, M. T., Briand, C., Forrer, P., Grütter, M. G., and Plückthun, A. (2004) High-affinity binders selected from designed ankyrin repeat protein libraries. Nat. Biotechnol. 22, 575– 82, DOI: 10.1038/nbt962Google Scholar42High-affinity binders selected from designed ankyrin repeat protein librariesBinz, H. Kaspar; Amstutz, Patrick; Kohl, Andreas; Stumpp, Michael T.; Briand, Christophe; Forrer, Patrik; Gruetter, Markus G.; Plueckthun, AndreasNature Biotechnology (2004), 22 (5), 575-582CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)We report here the evolution of ankyrin repeat (AR) proteins in vitro for specific, high-affinity target binding. Using a consensus design strategy, we generated combinatorial libraries of AR proteins of varying repeat nos. with diversified binding surfaces. Libraries of two and three repeats, flanked by 'capping repeats,' were used in ribosome-display selections against maltose binding protein (MBP) and two eukaryotic kinases. We rapidly enriched target-specific binders with affinities in the low nanomolar range and detd. the crystal structure of one of the selected AR proteins in complex with MBP at 2.3 Å resoln. The interaction relies on the randomized positions of the designed AR protein and is comparable to natural, heterodimeric protein-protein interactions. Thus, our AR protein libraries are valuable sources for binding mols. and, because of the very favorable biophys. properties of the designed AR proteins, an attractive alternative to antibody libraries.
- 43Bavdek, A., Gekara, N. O., Priselac, D., Gutiérrez Aguirre, I., Darji, A., Chakraborty, T., Macek, P., Lakey, J. H., Weiss, S., and Anderluh, G. (2007) Sterol and pH interdependence in the binding, oligomerization, and pore formation of Listeriolysin O. Biochemistry 46, 4425– 4437, DOI: 10.1021/bi602497gGoogle Scholar43Sterol and pH Interdependence in the Binding, Oligomerization, and Pore Formation of Listeriolysin OBavdek, Andrej; Gekara, Nelson O.; Priselac, Dragan; Gutierrez Aguirre, Ion; Darji, Ayub; Chakraborty, Trinad; Macek, Peter; Lakey, Jeremy H.; Weiss, Siegfried; Anderluh, GregorBiochemistry (2007), 46 (14), 4425-4437CODEN: BICHAW; ISSN:0006-2960. (American Chemical Society)Listeriolysin O (LLO) is the most important virulence factor of the intracellular pathogen Listeria monocytogenes. Its main task is to enable escape of bacteria from the phagosomal vacuole into the cytoplasm. LLO belongs to the cholesterol-dependent cytolysin (CDC) family but differs from other members, as it exhibits optimal activity at low pH. Its pore forming ability at higher pH values has been largely disregarded in Listeria pathogenesis. Here we show that high cholesterol concns. in the membrane restore the low activity of LLO at high pH values. LLO binds to lipid membranes, at physiol. or even slightly basic pH values, in a cholesterol-dependent fashion. Binding, insertion into lipid monolayers, and permeabilization of calcein-loaded liposomes are maximal above approx. 35 mol % cholesterol, a concn. range typically found in lipid rafts. The narrow transition region of cholesterol concn. sepg. low and high activity indicates that cholesterol not only allows the binding of LLO to membranes but also affects other steps in pore formation. We were able to detect some of these by surface plasmon resonance-based assays. In particular, we show that LLO recognition of cholesterol is detd. by the most exposed 3β-hydroxy group of cholesterol. In addn., LLO binds and permeabilizes J774 cells and human erythrocytes in a cholesterol-dependent fashion at physiol. or slightly basic pH values. The results clearly show that LLO activity at physiol. pH cannot be neglected and that its action at sites distal to cell entry may have important physiol. consequences for Listeria pathogenesis.
- 44Sallee, N. A., Yeh, B. J., and Lim, W. A. (2007) Engineering modular protein interaction switches by sequence overlap. J. Am. Chem. Soc. 129, 4606– 4611, DOI: 10.1021/ja0672728Google Scholar44Engineering Modular Protein Interaction Switches by Sequence OverlapSallee, Nathan A.; Yeh, Brian J.; Lim, Wendell A.Journal of the American Chemical Society (2007), 129 (15), 4606-4611CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Many cellular signaling pathways contain proteins whose interactions change in response to upstream inputs, allowing for conditional activation or repression of the interaction based on the presence of the input mol. The ability to engineer similar regulation into protein interaction elements would provide us with powerful tools for controlling cell signaling. Here we describe an approach for engineering diverse synthetic protein interaction switches. Specifically, by overlapping the sequences of pairs of protein interaction domains and peptides, we have been able to generate mutually exclusive regulation over their interactions. Thus, the hybrid protein (which is composed of the two overlapped interaction modules) can bind to either of the two resp. ligands for those modules, but not to both simultaneously. We show that these synthetic switch proteins can be used to regulate specific protein-protein interactions in vivo. These switches allow us to disrupt an interaction with the addn. or activation of a protein input that has no natural connection to the interaction in question. Therefore, they give us the ability to make novel connections between normally unrelated signaling pathways and to rewire the input/output relationships of cellular behaviors. Our expts. also suggest a possible mechanism by which complex regulatory proteins might have evolved from simpler components.
- 45Karginov, A. V., Ding, F., Kota, P., Dokholyan, N. V., and Hahn, K. M. (2010) Engineered allosteric activation of kinases in living cells. Nat. Biotechnol. 28, 743– 747, DOI: 10.1038/nbt.1639Google Scholar45Engineered allosteric activation of kinases in living cellsKarginov, Andrei V.; Ding, Feng; Kota, Pradeep; Dokholyan, Nikolay V.; Hahn, Klaus M.Nature Biotechnology (2010), 28 (7), 743-747CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)Studies of cellular and tissue dynamics benefit greatly from tools that can control protein activity with specificity and precise timing in living systems. Here we describe an approach to confer allosteric regulation specifically on the catalytic activity of protein kinases. A highly conserved portion of the kinase catalytic domain is modified with a small protein insert that inactivates catalytic activity but does not affect other protein functions. Catalytic activity is restored by addn. of rapamycin or non-immunosuppresive rapamycin analogs. Mol. modeling and mutagenesis indicate that the protein insert reduces activity by increasing the flexibility of the catalytic domain. Drug binding restores activity by increasing rigidity. We demonstrate the approach by specifically activating focal adhesion kinase (FAK) within minutes in living cells and show that FAK is involved in the regulation of membrane dynamics. Successful regulation of Src and p38 by insertion of the rapamycin-responsive element at the same conserved site used in FAK suggests that our strategy will be applicable to other kinases.
- 46Edwards, W. R., Busse, K., Allemann, R. K., and Jones, D. D. (2008) Linking the functions of unrelated proteins using a novel directed evolution domain insertion method. Nucleic Acids Res. 36, e78 DOI: 10.1093/nar/gkn363Google Scholar46Linking the functions of unrelated proteins using a novel directed evolution domain insertion methodEdwards, Wayne R.; Busse, Kathy; Allemann, Rudolf K.; Jones, D. DafyddNucleic Acids Research (2008), 36 (13), e78/1-e78/9CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)We have successfully developed a new directed evolution method for generating integral protein fusions comprising of one domain inserted within another. Creating two connections between the insert and accepting parent domain can result in the inter-dependence of the sep. protein activities, thus providing a general strategy for constructing mol. switches. Using an engineered transposon termed MuDel, contiguous trinucleotide sequences were removed at random positions from the bla gene encoding TEM-1 β-lactamase. The deleted trinucleotide sequence was then replaced by a DNA cassette encoding cytochrome b562 with differing linking sequences at each terminus and sampling all three reading frames. The result was a variety of chimeric genes encoding novel integral fusion proteins that retained TEM-1 activity. While most of the tolerated insertions were obsd. in loops, several also occurred close to the termini of α-helixes and β-strands. Several variants conferred a switching phenotype on Escherichia coli, with bacterial tolerance to ampicillin being dependent on the presence of haem in the growth medium. The magnitude of the switching phenotype ranged from 4- to 128-fold depending on the insertion position within TEM-1 and the linker sequences that join the two domains.
- 47Ha, J. H., Butler, J. S., Mitrea, D. M., and Loh, S. N. (2006) Modular enzyme design: Regulation by mutually exclusive protein folding. J. Mol. Biol. 357, 1058– 1062, DOI: 10.1016/j.jmb.2006.01.073Google Scholar47Modular Enzyme Design: Regulation by Mutually Exclusive Protein FoldingHa, Jeung-Hoi; Butler, James S.; Mitrea, Diana M.; Loh, Stewart N.Journal of Molecular Biology (2006), 357 (4), 1058-1062CODEN: JMOBAK; ISSN:0022-2836. (Elsevier B.V.)A regulatory mechanism is introduced whereupon the catalytic activity of a given enzyme is controlled by ligand binding to a receptor domain of choice. A small enzyme (barnase) and a ligand-binding polypeptide (GCN4) are fused so that a simple topol. constraint prevents them from existing simultaneously in their folded states. The two domains consequently engage in a thermodn. tug-of-war in which the more stable domain forces the less stable domain to unfold. In the absence of ligand, the barnase domain is more stable and is therefore folded and active; the GCN4 domain is substantially unstructured. DNA binding induces folding of GCN4, forcibly unfolding and inactivating the barnase domain. Barnase-GCN4 is thus a "natively unfolded" protein that uses ligand binding to switch between partially folded forms. The key characteristics of each parent protein (catalytic efficiency of barnase, DNA binding affinity and sequence specificity of GCN4) are retained in the chimera. Barnase-GCN4 thus defines a modular approach for assembling enzymes with novel sensor capabilities from a variety of catalytic and ligand binding domains.
- 48Tweten, R. K., Hotze, E. M., and Wade, K. R. (2015) The unique molecular choreography of giant pore formation by the cholesterol-dependent cytolysins of Gram-positive bacteria. Annu. Rev. Microbiol. 69, 323– 340, DOI: 10.1146/annurev-micro-091014-104233Google Scholar48The Unique Molecular Choreography of Giant Pore Formation by the Cholesterol-Dependent Cytolysins of Gram-Positive BacteriaTweten, Rodney K.; Hotze, Eileen M.; Wade, Kristin R.Annual Review of Microbiology (2015), 69 (), 323-340CODEN: ARMIAZ; ISSN:0066-4227. (Annual Reviews)A review. The mechanism by which the cholesterol-dependent cytolysins (CDCs) assemble their giant β-barrel pore in cholesterol-rich membranes has been the subject of intense study in the past two decades. A combination of structural, biophys., and biochem. analyses revealed deep insights into the series of complex and highly choreographed secondary and tertiary structural transitions that the CDCs undergo to assemble their β-barrel pore in eukaryotic membranes. The authors' knowledge of the mol. details of these dramatic structural changes in CDCs has transformed the understanding of how giant pore complexes are assembled and has been crit. to the understanding of the mechanisms of other important classes of pore-forming toxins and proteins across the kingdoms of life. Finally, there are tantalizing hints that the CDC pore-forming mechanism is more sophisticated than previously imagined and that some CDCs were employed in pore-independent processes.
- 49Song, L., Hobaugh, M. R., Shustak, C., Cheley, S., Bayley, H., and Gouaux, J. E. (1996) Structure of staphylococcal alpha-hemolysin, a heptameric transmembrane pore. Science 274, 1859– 1866, DOI: 10.1126/science.274.5294.1859Google Scholar49Structure of staphylococcal α-hemolysin, a heptameric transmembrane poreSong, Langzhou; Hobaugh, Michael R.; Shustak, Christopher; Cheley, Stephen; Bayley, Hagan; Gouaux, J. EricScience (Washington, D. C.) (1996), 274 (5294), 1859-1866CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)The structure of the Staphylococcus aureus α-hemolysin pore has been detd. to 1.9 Å resoln. Contained within the mushroom-shaped homo-oligomeric heptamer is a solvent-filled channel, 100 Å in length, that runs along the sevenfold axis and ranges from 14 Å to 46 Å in diam. The lytic, transmembrane domain comprises the lower half of a 14-strand antiparallel β barrel, to which each promoter contributes two β strands, each 65 Å long. The interior of the β barrel is primarily hydrophilic, and the exterior has a hydrophobic belt 28 Å wide. The structure proves the heptameric subunit stoichiometry of the α-hemolysin oligomer, shows that a glycine-rich and solvent-exposed region of a water-sol. protein can self-assemble to form a transmembrane pore of defined structure, and provides insight into the principles of membrane interaction and transport activity of β barrel pore-forming toxins.
- 50Podobnik, M., Savory, P., Rojko, N., Kisovec, M., Wood, N., Hambley, R., Pugh, J., Wallace, E. J., McNeill, L., Bruce, M., Liko, I., Allison, T. M., Mehmood, S., Yilmaz, N., Kobayashi, T., Gilbert, R. J. C., Robinson, C. V., Jayasinghe, L., and Anderluh, G. (2016) Crystal structure of an invertebrate cytolysin pore reveals unique properties and mechanism of assembly. Nat. Commun. 7, 11598, DOI: 10.1038/ncomms11598Google Scholar50Crystal structure of an invertebrate cytolysin pore reveals unique properties and mechanism of assemblyPodobnik, Marjetka; Savory, Peter; Rojko, Nejc; Kisovec, Matic; Wood, Neil; Hambley, Richard; Pugh, Jonathan; Wallace, E. Jayne; McNeill, Luke; Bruce, Mark; Liko, Idlir; Allison, Timothy M.; Mehmood, Shahid; Yilmaz, Neval; Kobayashi, Toshihide; Gilbert, Robert J. C.; Robinson, Carol V.; Jayasinghe, Lakmal; Anderluh, GregorNature Communications (2016), 7 (), 11598CODEN: NCAOBW; ISSN:2041-1723. (Nature Publishing Group)The invertebrate cytolysin lysenin is a member of the aerolysin family of pore-forming toxins that includes many representatives from pathogenic bacteria. Here we report the crystal structure of the lysenin pore and provide insights into its assembly mechanism. The lysenin pore is assembled from nine monomers via dramatic reorganization of almost half of the monomeric subunit structure leading to a β-barrel pore ∼10 nm long and 1.6-2.5 nm wide. The lysenin pore is devoid of addnl. luminal compartments as commonly found in other toxin pores. Mutagenic anal. and at. force microscopy imaging, together with these structural insights, suggest a mechanism for pore assembly for lysenin. These insights are relevant to the understanding of pore formation by other aerolysin-like pore-forming toxins, which often represent crucial virulence factors in bacteria.
- 51Tsuchikama, K. and An, Z. (2018) Antibody-drug conjugates: recent advances in conjugation and linker chemistries. Protein Cell 9, 33– 46, DOI: 10.1007/s13238-016-0323-0Google Scholar51Antibody-drug conjugates: recent advances in conjugation and linker chemistriesTsuchikama, Kyoji; An, ZhiqiangProtein & Cell (2018), 9 (1), 33-46CODEN: PCREFB; ISSN:1674-800X. (Higher Education Press)The antibody-drug conjugate (ADC), a humanized or human monoclonal antibody conjugated with highly cytotoxic small mols. (payloads) through chem. linkers, is a novel therapeutic format and has great potential to make a paradigm shift in cancer chemotherapy. This new antibody-based mol. platform enables selective delivery of a potent cytotoxic payload to target cancer cells, resulting in improved efficacy, reduced systemic toxicity, and preferable pharmacokinetics (PK)/pharmacodynamics (PD) and biodistribution compared to traditional chemotherapy. Boosted by the successes of FDA-approved Adcetris and Kadcyla, this drug class has been rapidly growing along with about 60 ADCs currently in clin. trials. In this article, we briefly review mol. aspects of each component (the antibody, payload, and linker) of ADCs, and then mainly discuss traditional and new technologies of the conjugation and linker chemistries for successful construction of clin. effective ADCs. Current efforts in the conjugation and linker chemistries will provide greater insights into mol. design and strategies for clin. effective ADCs from medicinal chem. and pharmacol. standpoints. The development of site-specific conjugation methodologies for constructing homogeneous ADCs is an esp. promising path to improving ADC design, which will open the way for novel cancer therapeutics.
- 52Leriche, G., Chisholm, L., and Wagner, A. (2012) Cleavable linkers in chemical biology. Bioorg. Med. Chem. 20, 571– 582, DOI: 10.1016/j.bmc.2011.07.048Google Scholar52Cleavable linkers in chemical biologyLeriche, Geoffray; Chisholm, Louise; Wagner, AlainBioorganic & Medicinal Chemistry (2012), 20 (2), 571-582CODEN: BMECEP; ISSN:0968-0896. (Elsevier B.V.)A review. Interest in cleavable linkers is growing due to the rapid development and expansion of chem. biol. The chem. constrains imposed by the biol. conditions cause significant challenges for org. chemists. In this review we will present an overview of the cleavable linkers used in chem. biol. classified according to their cleavage conditions by enzymes, nucleophilic/basic reagents, reducing agents, photo-irradn., electrophilic/acidic reagents, organometallic and metal reagents, oxidizing reagents.
- 53Cajnko, M. M., Marušić, M., Kisovec, M., Rojko, N., Benčina, M., Caserman, S., and Anderluh, G. (2015) Listeriolysin O affects the permeability of Caco-2 monolayer in a pore-dependent and Ca2+-independent manner. PLoS One 10, e0130471 DOI: 10.1371/journal.pone.0130471Google Scholar53Listeriolysin O affects the permeability of Caco-2 monolayer in a pore-dependent and Ca2+-independent mannerCajnko Misa, Mojca; Marusic, Maja; Kisovec, Matic; Rojko, Nejc; Bencina, Mojca; Caserman, Simon; Anderluh, GregorPLoS One (2015), 10 (6), e0130471/1-e0130471/21CODEN: POLNCL; ISSN:1932-6203. (Public Library of Science)Listeria monocytogenes is a food and soil-borne pathogen that secretes a pore-forming toxin listeriolysin O (LLO) as its major virulence factor. We tested the effects of LLO on an intestinal epithelial cell line Caco-2 and compared them to an unrelated pore-forming toxin equinatoxin II (EqtII). Apical application of both toxins causes a significant drop in transepithelial elec. resistance (TEER), with higher LLO concns. or prolonged exposure time needed to achieve the same magnitude of response than with EqtII. The drop in TEER was due to pore formation and coincided with rearrangement of claudin-1 within tight junctions and assocd. actin cytoskeleton; however, no significant increase in permeability to fluorescein or 3 kDa FITC-dextran was obsd. Influx of calcium after pore formation affected the magnitude of the drop in TEER. Both toxins exhibit similar effects on epithelium morphol. and physiol. Importantly, LLO action upon the membrane is much slower and results in compromised epithelium on a longer time scale at lower concns. than EqtII. This could favor listerial invasion in hosts resistant to E-cadherin related infection.
- 54Gerweck, L. E. and Seetharaman, K. (1996) Cellular pH gradient in tumor versus normal tissue: Potential exploitation for the treatment of cancer. Cancer Res. 56, 1194– 1198Google Scholar54Cellular pH gradient in tumor versus normal tissue: potential exploitation for the treatment of cancerGerweck, Leo E.; Seetharaman, KalaCancer Research (1996), 56 (6), 1194-8CODEN: CNREA8; ISSN:0008-5472. (American Association for Cancer Research)Although limited data exist, electrode-measured pH values of human tumors and adjacent normal tissues, which are concurrently obtained by the same investigator in the same patient, consistently show that the electrode pH (believed to primarily represent tissue extracellular pH) is substantially and consistently lower in tumor than in normal tissue. In contrast, the 31P-magnetic resonance spectroscopy estd. that intracellular pH is essentially identical or slightly more basic in tumor compared to normal tissue. As a consequence, the cellular pH gradient is substantially reduced or reversed in tumor compared to normal tissue: in normal tissue the extracellular pH is relatively basic, and in tumor tissue the magnitude of the pH gradient is reduced or reversed. This difference provides an exploitable avenue for the treatment of cancer. The extent to which drugs exhibiting weakly acid or basic properties are ionized is strongly dependent on the pH of their milieu. Weakly acidic drugs which are relatively lipid sol. in their nonionized state may diffuse freely across the cell membrane and, upon entering a relatively basic intracellular compartment, become trapped and accumulate within a cell, leading to substantial differences in the intracellular/extracellular drug distribution between tumor and normal tissue for drugs exhibiting appropriate pKas.
- 55Cerutti, P. A. (1985) Prooxidant states and tumor promotion. Science 227, 375– 381, DOI: 10.1126/science.2981433Google Scholar55Prooxidant states and tumor promotionCerutti, Peter A.Science (Washington, DC, United States) (1985), 227 (4685), 375-81CODEN: SCIEAS; ISSN:0036-8075.There is convincing evidence that cellular prooxidant states-i.e., increased concns. of active O and org. peroxides and radicals-can promote initiated cells to neoplastic growth. Prooxidant states can be caused by different classes of agents, including hyperbaric O, radiation, xenobiotic metabolites and Fenton-type reagents, modulators of the cytochrome P 450 electron-transport chain, peroxisome proliferators, inhibitors of the antioxidant defense, and membrane-active agents. Many of these agents are promoters or complete carcinogens. They cause chromosomal damage by indirect action, but the role of this damage in carcinogenesis remains unclear. Prooxidant states can be prevented or suppressed by the enzymes of the cellular antioxidant defense and low mol. wt. scavenger mols., and many antioxidants are antipromoters and anticarcinogens. Finally, prooxidant states may modulate the expression of a family of prooxidant genes, which are related to cell growth and differentiation, by inducing alterations in DNA structure or by epigenetic mechanisms, for example, by polyadenosine diphosphate-ribosylation of chromosomal proteins.
- 56Bergers, G., Brekken, R., McMahon, G., Vu, T. H., Itoh, T., Tamaki, K., Tanzawa, K., Thorpe, P., Itohara, S., Werb, Z., and Hanahan, D. (2000) Matrix metalloproteinase-9 triggers the angiogenic switch during carcinogenesis. Nat. Cell Biol. 2, 737– 744, DOI: 10.1038/35036374Google Scholar56Matrix metalloproteinase-9 triggers the angiogenic switch during carcinogenesisBergers, Gabriele; Brekken, Rolf; McMahon, Gerald; Vu, Thiennu H.; Itoh, Takeshi; Tamaki, Kazuhiko; Tanzawa, Kazuhiko; Thorpe, Philip; Itohara, Shigeyoshi; Werb, Zena; Hanahan, DouglasNature Cell Biology (2000), 2 (10), 737-744CODEN: NCBIFN; ISSN:1465-7392. (Nature Publishing Group)During carcinogenesis of pancreatic islets in transgenic mice, an angiogenic switch activates the quiescent vasculature. Paradoxically, vascular endothelial growth factor (VEGF) and its receptors are expressed constitutively. Nevertheless, a synthetic inhibitor (SU5416) of VEGF signalling impairs angiogenic switching and tumor growth. Two metalloproteinases, MMP-2/gelatinase-A and MMP-9/gelatinase-B, are upregulated in angiogenic lesions. MMP-9 can render normal islets angiogenic, releasing VEGF. MMP inhibitors reduce angiogenic switching, and tumor no. and growth, as does genetic ablation of MMP-9. Absence of MMP-2 does not impair induction of angiogenesis, but retards tumor growth, whereas lack of urokinase has no effect. Our results show that MMP-9 is a component of the angiogenic switch.
- 57Karimi, M., Ghasemi, A., Sahandi Zangabad, P., Rahighi, R., Moosavi Basri, S. M., Mirshekari, H., Amiri, M., Shafaei Pishabad, Z., Aslani, A., Bozorgomid, M., Ghosh, D., Beyzavi, A., Vaseghi, A., Aref, A. R., Haghani, L., Bahrami, S., and Hamblin, M. R. (2016) Smart micro/nanoparticles in stimulus-responsive drug/gene delivery systems. Chem. Soc. Rev. 45, 1457– 1501, DOI: 10.1039/C5CS00798DGoogle Scholar57Smart micro/nanoparticles in stimulus-responsive drug/gene delivery systemsKarimi, Mahdi; Ghasemi, Amir; Sahandi Zangabad, Parham; Rahighi, Reza; Moosavi Basri, S. Masoud; Mirshekari, H.; Amiri, M.; Shafaei Pishabad, Z.; Aslani, A.; Bozorgomid, M.; Ghosh, D.; Beyzavi, A.; Vaseghi, A.; Aref, A. R.; Haghani, L.; Bahrami, S.; Hamblin, Michael R.Chemical Society Reviews (2016), 45 (5), 1457-1501CODEN: CSRVBR; ISSN:0306-0012. (Royal Society of Chemistry)New achievements in the realm of nanoscience and innovative techniques of nanomedicine have moved micro/nanoparticles (MNPs) to the point of becoming actually useful for practical applications in the near future. Various differences between the extracellular and intracellular environments of cancerous and normal cells and the particular characteristics of tumors such as physicochem. properties, neovasculature, elasticity, surface elec. charge, and pH have motivated the design and fabrication of inventive "smart" MNPs for stimulus-responsive controlled drug release. These novel MNPs can be tailored to be responsive to pH variations, redox potential, enzymic activation, thermal gradients, magnetic fields, light, and ultrasound (US), or can even be responsive to dual or multi-combinations of different stimuli. This unparalleled capability has increased their importance as site-specific controlled drug delivery systems (DDSs) and has encouraged their rapid development in recent years. An in-depth understanding of the underlying mechanisms of these DDS approaches is expected to further contribute to this groundbreaking field of nanomedicine. Smart nanocarriers in the form of MNPs that can be triggered by internal or external stimulus are summarized and discussed in the present review, including pH-sensitive peptides and polymers, redox-responsive micelles and nanogels, thermo- or magnetic-responsive nanoparticles (NPs), mech.- or elec.-responsive MNPs, light or ultrasound-sensitive particles, and multi-responsive MNPs including dual stimuli-sensitive nanosheets of graphene. This review highlights the recent advances of smart MNPs categorized according to their activation stimulus (phys., chem., or biol.) and looks forward to future pharmaceutical applications.
- 58Zahnd, C., Amstutz, P., and Plückthun, A. (2007) Ribosome display: selecting and evolving proteins in vitro that specifically bind to a target. Nat. Methods 4, 269– 279, DOI: 10.1038/nmeth1003Google Scholar58Ribosome display: selecting and evolving proteins in vitro that specifically bind to a targetZahnd, Christian; Amstutz, Patrick; Plueckthun, AndreasNature Methods (2007), 4 (3), 269-279CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)Ribosome display is an in vitro selection and evolution technol. for proteins and peptides from large libraries. As it is performed entirely in vitro, there are two main advantages over other selection technologies. First, the diversity of the library is not limited by the transformation efficiency of bacterial cells, but only by the no. of ribosomes and different mRNA mols. present in the test tube. Second, random mutations can be introduced easily after each selection round, as no library must be transformed after any diversification step. This allows facile directed evolution of binding proteins over several generations (Box 1). A prerequisite for the selection of proteins from libraries is the coupling of genotype (RNA, DNA) and phenotype (protein). In ribosome display, this link is accomplished during in vitro translation by stabilizing the complex consisting of the ribosome, the mRNA and the nascent, correctly folded polypeptide (Fig. 1). The DNA library coding for a particular library of binding proteins is genetically fused to a spacer sequence lacking a stop codon. This spacer sequence, when translated, is still attached to the peptidyl tRNA and occupies the ribosomal tunnel, and thus allows the protein of interest to protrude out of the ribosome and fold. The ribosomal complexes are allowed to bind to surface-immobilized target. Whereas non-bound complexes are washed away, mRNA of the complexes displaying a binding polypeptide can be recovered, and thus, the genetic information of the binding polypeptides is available for anal. Here we describe a step-by-step procedure to perform ribosome display selection using an Escherichia coli S30 ext. for in vitro translation, based on the work originally described and further refined in our lab. A protocol that makes use of eukaryotic in vitro translation systems for ribosome display is also included in this issue.
- 59Zahnd, C., Wyler, E., Schwenk, J. M., Steiner, D., Lawrence, M. C., McKern, N. M., Pecorari, F., Ward, C. W., Joos, T. O., and Plückthun, A. (2007) A designed ankyrin repeat protein evolved to picomolar affinity to Her2. J. Mol. Biol. 369, 1015– 28, DOI: 10.1016/j.jmb.2007.03.028Google Scholar59A Designed Ankyrin Repeat Protein Evolved to Picomolar Affinity to Her2Zahnd, Christian; Wyler, Emanuel; Schwenk, Jochen M.; Steiner, Daniel; Lawrence, Michael C.; McKern, Neil M.; Pecorari, Frederic; Ward, Colin W.; Joos, Thomas O.; Plueckthun, AndreasJournal of Molecular Biology (2007), 369 (4), 1015-1028CODEN: JMOBAK; ISSN:0022-2836. (Elsevier Ltd.)Designed ankyrin repeat proteins (DARPins) are a novel class of binding mols., which can be selected to recognize specifically a wide variety of target proteins. DARPins were previously selected against human epidermal growth factor receptor 2 (Her2) with low nanomolar affinities. We describe here their affinity maturation by error-prone PCR and ribosome display yielding clones with zero to seven (av. 2.5) amino acid substitutions in framework positions. The DARPin with highest affinity (90 pM) carried four mutations at framework positions, leading to a 3000-fold affinity increase compared to the consensus framework variant, mainly coming from a 500-fold increase of the on-rate. This DARPin was found to be highly sensitive in detecting Her2 in human carcinoma exts. We have detd. the crystal structure of this DARPin at 1.7 Å, and found that a His to Tyr mutation at the framework position 52 alters the inter-repeat H-bonding pattern and causes a significant conformational change in the relative disposition of the repeat subdomains. These changes are thought to be the reason for the enhanced on-rate of the mutated DARPin. The DARPin not bearing the residue 52 mutation has an unusually slow on-rate, suggesting that binding occurred via conformational selection of a relatively rare state, which was stabilized by this His52Tyr mutation, increasing the on-rate again to typical values. An anal. of the structural location of the framework mutations suggests that randomization of some framework residues either by error-prone PCR or by design in a future library could increase affinities and the target binding spectrum.
- 60Liu, S., Netzel-Arnett, S., Birkedal-Hansen, H., and Leppla, S. H. (2000) Tumor cell-selective cytotoxicity of matrix metalloproteinase-activated anthrax toxin. Cancer Res. 60, 6061– 6067Google Scholar60Tumor cell-selective cytotoxicity of matrix metalloproteinase-activated anthrax toxinLiu, Shihui; Netzel-Arnett, Sarah; Birkedal-Hansen, Henning; Leppla, Stephen H.Cancer Research (2000), 60 (21), 6061-6067CODEN: CNREA8; ISSN:0008-5472. (American Association for Cancer Research)Matrix metalloproteinases (MMPs) are overexpressed in a variety of tumor tissues and cell lines, and their expression is highly correlated to tumor invasion and metastasis. To exploit these characteristics in the design of tumor cell-selective cytotoxins, we constructed two mutated anthrax toxin protective antigen (PA) proteins in which the furin protease cleavage site is replaced by sequences selectively cleaved by MMPs. These MMP-targeted PA proteins were activated rapidly and selectively on the surface of MMP-overexpressing tumor cells. The activated PA proteins caused internalization of a recombinant cytotoxin, FP59, consisting of anthrax toxin lethal factor residues 1-254 fused to the ADP-ribosylation domain of Pseudomonas exotoxin A. The toxicity of the mutated PA proteins for MMP-overexpressing cells was blocked by hydroxamate inhibitors of MMPs, including BB94, and by a tissue inhibitor of matrix metalloproteinases (TIMP-2). The mutated PA proteins killed MMP-overexpressing tumor cells while sparing nontumorigenic normal cells when these were grown together in a coculture model, indicating that PA activation occurred on the tumor cell surface and not in the supernatant. This method of achieving cell-type specificity is conceptually distinct from, and potentially synergistic with, the more common strategy of retargeting a protein toxin by fusion to a growth factor, cytokine, or antibody.
- 61Franke, D., Petoukhov, M. V., Konarev, P. V., Panjkovich, A., Tuukkanen, A., Mertens, H. D. T., Kikhney, A. G., Hajizadeh, N. R., Franklin, J. M., Jeffries, C. M., and Svergun, D. I. (2017) ATSAS 2.8: a comprehensive data analysis suite for small-angle scattering from macromolecular solutions. J. Appl. Crystallogr. 50, 1212– 1225, DOI: 10.1107/S1600576717007786Google Scholar61ATSAS 2.8: a comprehensive data analysis suite for small-angle scattering from macromolecular solutionsFranke, D.; Petoukhov, M. V.; Konarev, P. V.; Panjkovich, A.; Tuukkanen, A.; Mertens, H. D. T.; Kikhney, A. G.; Hajizadeh, N. R.; Franklin, J. M.; Jeffries, C. M.; Svergun, D. I.Journal of Applied Crystallography (2017), 50 (4), 1212-1225CODEN: JACGAR; ISSN:1600-5767. (International Union of Crystallography)A review. ATSAS is a comprehensive software suite for the anal. of small-angle scattering data from dil. solns. of biol. macromols. or nanoparticles. It contains applications for primary data processing and assessment, ab initio bead modeling, and model validation, as well as methods for the anal. of flexibility and mixts. In addn., approaches are supported that utilize information from X-ray crystallog., NMR spectroscopy or atomistic homol. modeling to construct hybrid models based on the scattering data. This article summarizes the progress made during the 2.5-2.8 ATSAS release series and highlights the latest developments. These include AMBIMETER, an assessment of the reconstruction ambiguity of exptl. data; DATCLASS, a multiclass shape classification based on exptl. data; SASRES, for estg. the resoln. of ab initio model reconstructions; CHROMIXS, a convenient interface to analyze in-line size exclusion chromatog. data; SHANUM, to evaluate the useful angular range in measured data; SREFLEX, to refine available high-resoln. models using normal mode anal.; SUPALM for a rapid superposition of low- and high-resoln. models; and SASPy, the ATSAS plugin for interactive modeling in PyMOL. All these features and other improvements are included in the ATSAS release 2.8, freely available for academic users from https://www.embl-hamburg.de/biosaxs/software.html.</a></a></a></a></a></a></a></a></a>.
- 62Franke, D. and Svergun, D. I. (2009) DAMMIF, a program for rapid ab-initio shape determination in small-angle scattering. J. Appl. Crystallogr. 42, 342– 346, DOI: 10.1107/S0021889809000338Google Scholar62DAMMIF, a program for rapid ab-initio shape determination in small-angle scatteringFranke, Daniel; Svergun, Dmitri I.Journal of Applied Crystallography (2009), 42 (2), 342-346CODEN: JACGAR; ISSN:0021-8898. (International Union of Crystallography)DAMMIF, a revised implementation of the ab-initio shape-detn. program DAMMIN for small-angle scattering data, is presented. The program was fully rewritten, and its algorithm was optimized for speed of execution and modified to avoid limitations due to the finite search vol. Symmetry and anisometry constraints can be imposed on the particle shape, similar to DAMMIN. In equivalent conditions, DAMMIF is 25-40 times faster than DAMMIN on a single CPU. The possibility to utilize multiple CPUs is added to DAMMIF. The application is available in binary form for major platforms.
- 63Volkov, V. V. and Svergun, D. I. (2003) Uniqueness of ab initio shape determination in small-angle scattering. J. Appl. Crystallogr. 36, 860– 864, DOI: 10.1107/S0021889803000268Google Scholar63Uniqueness of ab initio shape determination in small-angle scatteringVolkov, Vladimir V.; Svergun, Dmitri I.Journal of Applied Crystallography (2003), 36 (3, Pt. 1), 860-864CODEN: JACGAR; ISSN:0021-8898. (Blackwell Munksgaard)Scattering patterns from geometrical bodies with different shapes and anisometry (solid and hollow spheres, cylinders, prisms) are computed and the shapes are reconstructed ab initio using envelope function and bead modeling methods. A procedure is described to analyze multiple solns. provided by bead modeling methods and to est. stability and reliability of the shape reconstruction. It is demonstrated that flat shapes are more difficult to restore than elongated ones and types of shapes are indicated, which require addnl. information for reliable shape reconstruction from the scattering data.
- 64The PyMOL Molecular Graphics System, Version 2.0, Schrödinger, LLC.Google ScholarThere is no corresponding record for this reference.
- 65Webb, B., and Sali, A. (2017) Protein Structure Modeling with MODELLER, in Functional Genomics: Methods and Protocols, Methods in Molecular Biology, (Kaufmann, M., Klinger, C., and Savelsbergh, A., Eds.), pp 39– 54, Humana Press, New York, NY.Google ScholarThere is no corresponding record for this reference.
- 66Waterhouse, A., Bertoni, M., Bienert, S., Studer, G., Tauriello, G., Gumienny, R., Heer, F. T., De Beer, T. A. P., Rempfer, C., Bordoli, L., Lepore, R., and Schwede, T. (2018) SWISS-MODEL: Homology modelling of protein structures and complexes. Nucleic Acids Res. 46, W296– W303, DOI: 10.1093/nar/gky427Google Scholar66SWISS-MODEL: homology modelling of protein structures and complexesWaterhouse, Andrew; Bertoni, Martino; Bienert, Stefan; Studer, Gabriel; Tauriello, Gerardo; Gumienny, Rafal; Heer, Florian T.; de Beer, Tjaart A. P.; Rempfer, Christine; Bordoli, Lorenza; Lepore, Rosalba; Schwede, TorstenNucleic Acids Research (2018), 46 (W1), W296-W303CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)Homol. modeling has matured into an important technique in structural biol., significantly contributing to narrowing the gap between known protein sequences and exptl. detd. structures. Fully automated workflows and servers simplify and streamline the homol. modeling process, also allowing users without a specific computational expertise to generate reliable protein models and have easy access to modeling results, their visualization and interpretation. Here, we present an update to the SWISS-MODEL server, which pioneered the field of automated modeling 25 years ago and been continuously further developed. Recently, its functionality has been extended to the modeling of homo- and heteromeric complexes. Starting from the amino acid sequences of the interacting proteins, both the stoichiometry and the overall structure of the complex are inferred by homol. modeling. Other major improvements include the implementation of a new modeling engine, ProMod3 and the introduction a new local model quality estn. method, QMEANDisCo.
- 67Schweizer, A., Roschitzki-Voser, H., Amstutz, P., Briand, C., Gulotti-Georgieva, M., Prenosil, E., Binz, H. K., Capitani, G., Baici, A., Plückthun, A., and Grütter, M. G. (2007) Inhibition of caspase-2 by a designed ankyrin repeat protein: specificity, structure, and inhibition mechanism. Structure 15, 625– 36, DOI: 10.1016/j.str.2007.03.014Google Scholar67Inhibition of Caspase-2 by a Designed Ankyrin Repeat Protein: Specificity, Structure, and Inhibition MechanismSchweizer, Andreas; Roschitzki-Voser, Heidi; Amstutz, Patrick; Briand, Christophe; Gulotti-Georgieva, Maya; Prenosil, Eva; Binz, H. Kaspar; Capitani, Guido; Baici, Antonio; Plueckthun, Andreas; Gruetter, Markus G.Structure (Cambridge, MA, United States) (2007), 15 (5), 625-636CODEN: STRUE6; ISSN:0969-2126. (Cell Press)Summary: Specific and potent caspase inhibitors are indispensable for the dissection of the intricate pathways leading to apoptosis. We selected a designed ankyrin repeat protein (DARPin) from a combinatorial library that inhibits caspase-2 in vitro with a subnanomolar inhibition const. and, in contrast to the peptidic caspase inhibitors, with very high specificity for this particular caspase. The crystal structure of this inhibitor (AR_F8) in complex with caspase-2 reveals the mol. basis for the specificity and, together with kinetic analyses, the allosteric mechanism of inhibition. The structure also shows a conformation of the active site that can be exploited for the design of inhibitory compds. AR_F8 is a specific inhibitor of an initiator caspase and has the potential to help identify the function of caspase-2 in the complex biol. apoptotic signaling network.
- 68Petoukhov, M. V. and Svergun, D. I. (2005) Global rigid body modeling of macromolecular complexes against small-angle scattering data. Biophys. J. 89, 1237– 1250, DOI: 10.1529/biophysj.105.064154Google Scholar68Global rigid body modeling of macromolecular complexes against small-angle scattering dataPetoukhov, Maxim V.; Svergun, Dmitri I.Biophysical Journal (2005), 89 (2), 1237-1250CODEN: BIOJAU; ISSN:0006-3495. (Biophysical Society)New methods to automatically build models of macromol. complexes from high-resoln. structures or homol. models of their subunits or domains against x-ray or neutron small-angle scattering data are presented. Depending on the complexity of the object, different approaches are employed for the global search of the optimum configuration of subunits fitting the exptl. data. An exhaustive grid search is used for hetero- and homodimeric particles and for sym. oligomers formed by identical subunits. For the assemblies or multidomain proteins contg. more then one subunit/domain per asym. unit, heuristic algorithms based on simulated annealing are used. Fast computational algorithms based on spherical harmonics representation of scattering amplitudes are employed. The methods allow one to construct interconnected models without steric clashes, to account for the particle symmetry and to incorporate information from other methods, on distances between specific residues or nucleotides. For multidomain proteins, addn. of missing linkers between the domains is possible. Simultaneous fitting of multiple scattering patterns from subcomplexes or deletion mutants is incorporated. The efficiency of the methods is illustrated by their application to complexes of different types in several simulated and practical examples. Limitations and possible ambiguity of rigid body modeling are discussed and simplified docking criteria are provided to rank multiple models. The methods described are implemented in publicly available computer programs running on major hardware platforms.
- 69Panjkovich, A. and Svergun, D. I. (2016) SASpy: A PyMOL plugin for manipulation and refinement of hybrid models against small angle X-ray scattering data. Bioinformatics 32, 2062– 2064, DOI: 10.1093/bioinformatics/btw071Google Scholar69SASpy: a PyMOL plugin for manipulation and refinement of hybrid models against small angle X-ray scattering dataPanjkovich, Alejandro; Svergun, Dmitri I.Bioinformatics (2016), 32 (13), 2062-2064CODEN: BOINFP; ISSN:1367-4803. (Oxford University Press)Complex formation and conformational transitions of biol. macromols. in soln. can be effectively studied using the information about overall shape and size provided by small angle X-ray scattering (SAXS). Hybrid modeling is often applied to integrate high-resoln. models into SAXS data anal. To facilitate this task, we present SASpy, a PyMOL plugin that provides an easy-to-use graphical interface for SAXS-based hybrid modeling. Through a few mouse clicks in SASpy, low-resoln. models can be superimposed to high-resoln. structures, theor. scattering profiles and fits can be calcd. and displayed on-the-fly. Mouse-based manual rearrangements of complexes are conveniently applied to rapidly check and interactively refine tentative models. Interfaces to automated rigid-body and flexible refinement of macromol. models against the exptl. SAXS data are provided.
- 70Hodnik, V., and Anderluh, G. (2013) Surface Plasmon Resonance for Measuring Interactions of Proteins with Lipid Membranes, in Lipid-Protein Interactions: Methods and Protocols, Methods in Molecular Biology, (Kleinschmidt, J. H., Ed.), pp 23– 36, Humana Press, Totowa, NJ.Google ScholarThere is no corresponding record for this reference.
- 71Anderluh, G., Beseničar, M., Kladnik, A., Lakey, J. H., and Maček, P. (2005) Properties of nonfused liposomes immobilized on an L1 Biacore chip and their permeabilization by a eukaryotic pore-forming toxin. Anal. Biochem. 344, 43– 52, DOI: 10.1016/j.ab.2005.06.013Google Scholar71Properties of nonfused liposomes immobilized on an L1 Biacore chip and their permeabilization by a eukaryotic pore-forming toxinAnderluh, Gregor; Besenicar, Mojca; Kladnik, Ales; Lakey, Jeremy H.; Macek, PeterAnalytical Biochemistry (2005), 344 (1), 43-52CODEN: ANBCA2; ISSN:0003-2697. (Elsevier)The L1 chip is used intensively for protein-membrane interaction studies in Biacore surface plasmon resonance systems. The exact form of captured lipid membranes on the chip is, however, not precisely known. Evidence exists that the vesicles both remain intact after the binding to the chip and fuse to form a large single-bilayer membrane. In this study, the authors were able to bind up to approx. 11,500 resonance units of zwitterionic liposomes (100 nm in diam.) at a low flow rate. The authors show by fluorescence microscopy that the entire surface of the flow cell is covered homogeneously by liposomes. Neg. charged vesicles (i.e., those composed of phosphatidylcholine/phosphatidylglycerol [1:1]) always deposited less densely, but the authors were able to increase the d. slightly with the use of calcium chloride that promotes fusion of the vesicles. Finally, the authors used zwitterionic liposomes loaded with fluorescent probe calcein to show that they remain intact after the capture on the L1 chip. The fluorescence was lost only after the authors used equinatoxin, a well-studied pore-forming toxin, to perform on-chip permeabilization of vesicles. The characteristics of permeabilization process for chip-immobilized liposomes are similar to those of liposomes free in soln. All results collectively suggest that liposomes do not fuse to form a single bilayer on the surface of the chip.
- 72Schneider, C. A., Rasband, W. S., and Eliceiri, K. W. (2012) NIH Image to ImageJ: 25 years of image analysis. Nat. Methods 9, 671– 675, DOI: 10.1038/nmeth.2089Google Scholar72NIH Image to ImageJ: 25 years of image analysisSchneider, Caroline A.; Rasband, Wayne S.; Eliceiri, Kevin W.Nature Methods (2012), 9 (7_part1), 671-675CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)For the past 25 years NIH Image and ImageJ software have been pioneers as open tools for the anal. of scientific images. We discuss the origins, challenges and solns. of these two programs, and how their history can serve to advise and inform other software projects.
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 12 publications.
- Jin Xu. Protein Computing. 2025, 301-329. https://doi.org/10.1007/978-981-96-3870-3_12
- Carolina Monck, Yuval Elani, Francesca Ceroni. Genetically programmed synthetic cells for thermo-responsive protein synthesis and cargo release. Nature Chemical Biology 2024, 20
(10)
, 1380-1386. https://doi.org/10.1038/s41589-024-01673-7
- Debopriya Bose, Gun Anit Kaur, Sapna Balayan, Subhrangsu Chatterjee, Ashutosh Tiwari. The logic devices for biomolecular computing: Progress, strategies, and future directions. Nano Today 2024, 57 , 102320. https://doi.org/10.1016/j.nantod.2024.102320
- Stephan L. Watkins. Current Trends and Changes in Use of Membrane Molecular Dynamics Simulations within Academia and the Pharmaceutical Industry. Membranes 2023, 13
(2)
, 148. https://doi.org/10.3390/membranes13020148
- Dennis S. Winston, David D. Boehr. Catalyst-Based Biomolecular Logic Gates. Catalysts 2022, 12
(7)
, 712. https://doi.org/10.3390/catal12070712
- Xiaopeng Gao, Ke Xu, Nadeem Ahmad, Lei Qin, Chun Li. Recent advances in engineering of microbial cell factories for intelligent pH regulation and tolerance. Biotechnology Journal 2021, 16
(9)
https://doi.org/10.1002/biot.202100151
- Nejc Petrišič, Mirijam Kozorog, Saša Aden, Marjetka Podobnik, Gregor Anderluh. The molecular mechanisms of listeriolysin O-induced lipid membrane damage. Biochimica et Biophysica Acta (BBA) - Biomembranes 2021, 1863
(7)
, 183604. https://doi.org/10.1016/j.bbamem.2021.183604
- Robert Strutt, James W. Hindley, Jordan Gregg, Paula J. Booth, John D. Harling, Robert V. Law, Mark S. Friddin, Oscar Ces. Activating mechanosensitive channels embedded in droplet interface bilayers using membrane asymmetry. Chemical Science 2021, 12
(6)
, 2138-2145. https://doi.org/10.1039/D0SC03889J
- Jiafeng Pan, Ying He, Zhi Liu, Junhua Chen. Dual recognition element-controlled logic DNA circuit for COVID-19 detection based on exonuclease III and DNAzyme. Chemical Communications 2021, 57
(9)
, 1125-1128. https://doi.org/10.1039/D0CC06799G
- Saša Aden, Tina Snoj, Gregor Anderluh. The use of giant unilamellar vesicles to study functional properties of pore-forming toxins. 2021, 219-251. https://doi.org/10.1016/bs.mie.2021.01.016
- Aleksandra Šakanović, Nace Kranjc, Neža Omersa, Marjetka Podobnik, Gregor Anderluh. More than one way to bind to cholesterol: atypical variants of membrane-binding domain of perfringolysin O selected by ribosome display. RSC Advances 2020, 10
(63)
, 38678-38682. https://doi.org/10.1039/D0RA06976K
- Eran A. Barnoy, Rachela Popovtzer, Dror Fixler. Fluorescence for biological logic gates. Journal of Biophotonics 2020, 13
(9)
https://doi.org/10.1002/jbio.202000158
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. Properties and permeabilizing activity of Y406A. (a) A ribbon model of LLO 3D structure (PDB ID: 4CDB) with each domain (D1–D4) labeled in different color. Position of residue 406 is denoted by an orange color and an arrow. (b) A model of pore formation by Y406A (monomer binding to the membrane, oligomerization, pore formation). Domains are colored as on panel a. (c) Calcein release from large unilamellar vesicles after 30 min is shown at different pH values for LLO (black) and Y406A (red). Data reproduced with permission from Kisovec et al. (40) (d) Relative rate of hemolysis by 2.3 nM CDCs at pH 5.7 in the presence of DARPins: 5 μM D22 (red bars), 5 μM D22M (blue bar), a variant that was used for immobilization to the lipid membrane (see below), and 5 μM D6 and D30 (green bars), DARPin clones that were selected with ribosome display as high affinity binders, but did not inhibit hemolytic activity of Y406A. Mean ± SD; n = 2–7.
Figure 2
Figure 2. Binding of D22 to Y406A in solution. (a) Size exclusion chromatogram of LLO and Y406A in the absence or presence of D22. Triangles indicate positions of elution peaks for different proteins. Note that LLO travels aberrantly on the size exclusion column eluting with larger volumes of elution buffer than expected. (b) Binding of D22 to LLO (gray) or Y406A (black) in solution (22 mM MES, 150 mM NaCl and 5 mM 2-mercaptoethanol, pH 5.7), measured by isothermal titration calorimetry. Top panel represents raw data of injections of 54.9 μM D22 into a 5.9 μM solution of LLO or Y406A. Bottom panel shows normalized integrated enthalpies plotted against the molar ratio. Circles represent experimental points, and the solid line corresponds to the best fit obtained by one-site reaction model. (c) An overlay of experimental scattering data obtained by SAXS experiment of Y406A-D22 complex (red circles) with the calculated scattering curve from the representative DAMMIF model (χ2 = 1.125, black line). Inset, overlay of Y406A (blue ribbon) and D22 (red ribbon) refined by rigid body modeling (χ2 = 1.12), with the best SAXS bead model in surface representation and the average SAXS bead model in mesh representation.
Figure 3
Figure 3. Interaction of D22 with Y406A in the lipid membrane environment. (a) SPR measurements showing binding of 100 nM Y406A, 5 μM D22 and preincubated Y406A-D22 complex (with same concentrations of individual proteins as used for single proteins injections) to large unilamellar vesicles. (b) SPR sensorgrams of 5 μM D22 binding to 100 nM membrane-inserted Y406A or LLO. Membrane denotes control experiment with D22 binding to vesicles only. (c) Vesicle sedimentation assays with multilamellar vesicles after preincubation of LLO and D22, or Y406A and D22 in solution (“preincubated”) or when LLO or Y406A were first preincubated with vesicles (“membrane bound”). p, pellet; s, supernatant; w, additional washing step, which was included when assaying membrane bound LLO or Y406A in order to check for the completeness of binding. Band at app. 60 kDa corresponds to LLO or Y406A, while band at ca. 18 kDa corresponds to D22. D22 is present at 5× molar excess; therefore, a large portion of it is always unbound in supernatant. (d) Quantification of the SDS-PAGE data from (c) by densitometry. Full binding of D22 to Y406A (100%) was considered when one-fifth of the applied D22 was bound to Y406A. Mean ± SD; two sample t test, ***P < 0.001 (n = 4–6). Amount of bound D22 is reported when preincubated with LLO (1) or Y406A (2) in solution or when LLO (3) or Y406A (4) were first bound to vesicles.
Figure 4
Figure 4. Modulation of Y406A permeabilization activity by D22 and pH. (a) Permeabilization of GUVs for FD10 at different conditions and induced by the 50 nM LLO. (b) Permeabilization induced by 50 nM Y406A. The graphs on the right in A and B show quantification of GUVs data from confocal microscopy images as represented on the left. Mean ± SD; n = 96–568. (c) Hemolysis induced by 18.2 nM LLO at different conditions. (d) Hemolysis induced by 18.2 nM Y406A. In C and D, mean ± SD is presented; n = 3. (e) 5 μM D22 by itself does not induce permeabilization of GUVs. Confocal images of GUVs on the left, and quantification is presented on the right. n is 256 and 458 for pH 6.5 and 8.0, respectively. (f) The truth table and schematic representation of a NOR logic gate for the Y406A-D22 system.
Figure 5
Figure 5. Modulation of Y406A activity in calcein release experiments. (a,b) Calcein release from SUVs composed of POPC:Chol, 3:2 (mol:mol) as a result of pore formation by 1 μM LLO (a) or Y406A (b), in presence or absence of 5 μM D22 and at pH values 6.5 and 8.0. (c) Calcein release from LUVs composed of POPC:Chol, 1:1 (mol:mol) monitored at different conditions. Vesicles were stirred in 10 mM HEPES, 150 mM NaCl, 1 mM EDTA, pH 8.0. Final concentration of 30 nM Y406A (black triangle), 1 μM D22 or 3 μL of 7% HCl (to reduce pH to approximately 6.5) were added, respectively, at times denoted by triangles. The scale bar is the same for all fluorescent traces.
Figure 6
Figure 6. Conjugation of D22M to vesicles and cleavage with different agents. Schematic diagrams of used systems and structures of employed lipids are shown. Y406A is presented with green color, D22 is shown in orange. Different parts of lipids used for conjugation are presented with different colors in the structural formulas and on the diagram. (a–c) A system employing DSPE-PEG2000Mal lipid, which attaches D22 to the lipid membrane and allows subsequent cleavage by MMP-9. (d–f) A system employing DSPE-PEG2000PDP lipid, which allows cleavage with reductant TCEP. The input of different proteins and reagents is shown above the sedimentation assay gels. The approximate positions of different proteins and lipids on the gels are indicated by arrows. i, 2 μg of D22M as an input; p and s denote pellet and supernatant after centrifugation of MLVs, respectively. (c) and (f) each represent one experiment where MLVs were first incubated with Y406A and then with MMP-9 or TCEP. 1, pellet after both incubations; 2, supernatant after centrifugation of MLVs after incubation with Y406A; 3, supernatant after centrifugation of MLVs with bound Y406A and incubation of MMP-9 or TCEP.
Figure 7
Figure 7. A tunable vesicle system. (a) Y406A activity is controlled by pH and the reversible inhibitor D22. Vesicles are at pH 8.0; therefore, two input signals are needed for system activation: lowering pH with HCl and elimination of D22 from the system by release from vesicles induced by MMP-9 or TCEP. (b) A system based on DSPE-PEG2000Mal lipid. Bars represent fluorescence increase due to release of calcein from MLVs. A logic truth table and schematic representation of an AND gate is presented below the graph. (c) A system based on DSPE-PEG2000PDP lipid, which gives more flexibility for the removal of D22 from the vesicle membrane. A logic truth table and schematic representation of an OR-AND gate is presented beside the graph. n = 3–6, mean ± SD *P < 0.05; **P < 0.01.
References
This article references 72 other publications.
- 1Evans, A. C., Thadani, N. N., and Suh, J. (2016) Biocomputing nanoplatforms as therapeutics and diagnostics. J. Controlled Release 240, 387– 393, DOI: 10.1016/j.jconrel.2016.01.0451Biocomputing nanoplatforms as therapeutics and diagnosticsEvans, A. C.; Thadani, N. N.; Suh, J.Journal of Controlled Release (2016), 240 (), 387-393CODEN: JCREEC; ISSN:0168-3659. (Elsevier B.V.)A review. Biocomputing nanoplatforms are designed to detect and integrate single or multiple inputs under defined algorithms, such as Boolean logic gates, and generate functionally useful outputs, such as delivery of therapeutics or release of optically detectable signals. Using sensing modules composed of small mols., polymers, nucleic acids, or proteins/peptides, nanoplatforms have been programmed to detect and process extrinsic stimuli, such as magnetic fields or light, or intrinsic stimuli, such as nucleic acids, enzymes, or pH. Stimulus detection can be transduced by the nanomaterial via three different mechanisms: system assembly, system disassembly, or system transformation. The increasingly sophisticated suite of biocomputing nanoplatforms may be invaluable for a multitude of applications, including medical diagnostics, biomedical imaging, environmental monitoring, and delivery of therapeutics to target cell populations.
- 2Khalil, A. S. and Collins, J. J. (2010) Synthetic biology: Applications come of age. Nat. Rev. Genet. 11, 367– 379, DOI: 10.1038/nrg27752Synthetic biology: applications come of ageKhalil, Ahmad S.; Collins, James J.Nature Reviews Genetics (2010), 11 (5), 367-379CODEN: NRGAAM; ISSN:1471-0056. (Nature Publishing Group)Advances in the synthetic biol. field are allowing an expansion beyond small gene networks towards larger biol. programs that hold promise for a wide range of applications, including biosensing, therapeutics and the prodn. of biofuels, pharmaceuticals and biomaterials. Synthetic biol. is bringing together engineers and biologists to design and build novel biomol. components, networks and pathways, and to use these constructs to rewire and reprogram organisms. These re-engineered organisms will change our lives over the coming years, leading to cheaper drugs, 'green' means to fuel our cars and targeted therapies for attacking 'superbugs' and diseases, such as cancer. The de novo engineering of genetic circuits, biol. modules and synthetic pathways is beginning to address these crucial problems and is being used in related practical applications.
- 3Miyamoto, T., Razavi, S., Derose, R., and Inoue, T. (2013) Synthesizing biomolecule-based boolean logic gates. ACS Synth. Biol. 2, 72– 82, DOI: 10.1021/sb30011123Synthesizing Biomolecule-Based Boolean Logic GatesMiyamoto, Takafumi; Razavi, Shiva; DeRose, Robert; Inoue, TakanariACS Synthetic Biology (2013), 2 (2), 72-82CODEN: ASBCD6; ISSN:2161-5063. (American Chemical Society)A review. One fascinating recent avenue of study in the field of synthetic biol. is the creation of biomol.-based computers. The main components of a computing device consist of an arithmetic logic unit, the control unit, memory, and the input and output devices. Boolean logic gates are at the core of the operational machinery of these parts, and hence to make biocomputers a reality, biomol. logic gates become a necessity. Indeed, with the advent of more sophisticated biol. tools, both nucleic acid- and protein-based logic systems have been generated. These devices function in the context of either test tubes or living cells and yield highly specific outputs given a set of inputs. In this review, we discuss various types of biomol. logic gates that have been synthesized, with particular emphasis on recent developments that promise increased complexity of logic gate circuitry, improved computational speed, and potential clin. applications.
- 4Gaber, R., Lebar, T., Majerle, A., Šter, B., Dobnikar, A., Benčina, M., and Jerala, R. (2014) Designable DNA-binding domains enable construction of logic circuits in mammalian cells. Nat. Chem. Biol. 10, 203– 208, DOI: 10.1038/nchembio.14334Designable DNA-binding domains enable construction of logic circuits in mammalian cellsGaber, Rok; Lebar, Tina; Majerle, Andreja; Ster, Branko; Dobnikar, Andrej; Bencina, Mojca; Jerala, RomanNature Chemical Biology (2014), 10 (3), 203-208CODEN: NCBABT; ISSN:1552-4450. (Nature Publishing Group)Electronic computer circuits consisting of a large no. of connected logic gates of the same type, such as NOR, can be easily fabricated and can implement any logic function. In contrast, designed genetic circuits must employ orthogonal information mediators owing to free diffusion within the cell. Combinatorial diversity and orthogonality can be provided by designable DNA- binding domains. Here, we employed the transcription activator-like repressors to optimize the construction of orthogonal functionally complete NOR gates to construct logic circuits. We used transient transfection to implement all 16 two-input logic functions from combinations of the same type of NOR gates within mammalian cells. Addnl., we present a genetic logic circuit where one input is used to select between an AND and OR function to process the data input using the same circuit. This demonstrates the potential of designable modular transcription factors for the construction of complex biol. information-processing devices.
- 5Lebar, T., Bezeljak, U., Golob, A., Jerala, M., Kadunc, L., Pirš, B., Stražar, M., Vučko, D., Zupančič, U., Benčina, M., Forstnerič, V., Gaber, R., Lonzarić, J., Majerle, A., Oblak, A., Smole, A., and Jerala, R. (2014) A bistable genetic switch based on designable DNA-binding domains. Nat. Commun. 5, 5007, DOI: 10.1038/ncomms60075A bistable genetic switch based on designable DNA-binding domainsLebar, Tina; Bezeljak, Urban; Golob, Anja; Jerala, Miha; Kadunc, Lucija; Pirs, Bostjan; Strazar, Martin; Vucko, Dusan; Zupancic, Uros; Bencina, Mojca; Forstneric, Vida; Gaber, Rok; Lonzaric, Jan; Majerle, Andreja; Oblak, Alja; Smole, Anze; Jerala, RomanNature Communications (2014), 5 (), 5007CODEN: NCAOBW; ISSN:2041-1723. (Nature Publishing Group)Bistable switches are fundamental regulatory elements of complex systems, ranging from electronics to living cells. Designed genetic toggle switches have been constructed from pairs of natural transcriptional repressors wired to inhibit one another. The complexity of the engineered regulatory circuits can be increased using orthogonal transcriptional regulators based on designed DNA-binding domains. However, a mutual repressor-based toggle switch comprising DNA-binding domains of transcription-activator-like effectors (TALEs) did not support bistability in mammalian cells. Here, the challenge of engineering a bistable switch based on monomeric DNA-binding domains is solved via the introduction of a pos. feedback loop composed of activators based on the same TALE domains as their opposing repressors and competition for the same DNA operator site. This design introduces nonlinearity and results in epigenetic bistability. This principle could be used to employ other monomeric DNA-binding domains such as CRISPR for applications ranging from reprogramming cells to building digital biol. memory.
- 6Lebar, T., and Jerala, R. (2018) Designed transcriptional regulation in mammalian cells based on TALE- and CRISPR/dCas9, in Synthetic Biology Methods and Protocols, Methods in Molecular Biology, (Braman, J. C., Ed.), pp 191– 203, Humana Press, New York, NY.There is no corresponding record for this reference.
- 7Qian, L. and Winfree, E. (2011) Scaling up digital circuit computation with DNA strand displacement cascades. Science 332, 1196– 1201, DOI: 10.1126/science.12005207Scaling Up Digital Circuit Computation with DNA Strand Displacement CascadesQian, Lulu; Winfree, ErikScience (Washington, DC, United States) (2011), 332 (6034), 1196-1201CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)To construct sophisticated biochem. circuits from scratch, one needs to understand how simple the building blocks can be and how robustly such circuits can scale up. Using a simple DNA reaction mechanism based on a reversible strand displacement process, the authors exptl. demonstrated several digital logic circuits, culminating in a 4-bit square-root circuit that comprises 130 DNA strands. These multilayer circuits include thresholding and catalysis within every logical operation to perform digital signal restoration, which enables fast and reliable function in large circuits with roughly const. switching time and linear signal propagation delays. The design naturally incorporates other crucial elements for large-scale circuitry, such as general debugging tools, parallel circuit prepn., and an abstraction hierarchy supported by an automated circuit compiler.
- 8Yasuga, H., Kawano, R., Takinoue, M., Tsuji, Y., Osaki, T., Kamiya, K., Miki, N., and Takeuchi, S. (2016) Logic gate operation by DNA translocation through biological nanopores. PLoS One 11, e0149667 DOI: 10.1371/journal.pone.01496678Logic gate operation by DNA translocation through biological nanoporesYasuga, Hiroki; Kawano, Ryuji; Takinoue, Masahiro; Tsuji, Yutaro; Osaki, Toshihisa; Kamiya, Koki; Miki, Norihisa; Takeuchi, ShojiPLoS One (2016), 11 (2), e0149667/1-e0149667/13CODEN: POLNCL; ISSN:1932-6203. (Public Library of Science)Logical operations using biol. mols., such as DNA computing or programmable diagnosis using DNA, have recently received attention. Challenges remain with respect to the development of such systems, including label-free output detection and the rapidity of operation. Here, we propose integration of biol. nanopores with DNA mols. for development of a logical operating system. We configured outputs "1" and "0" as single-stranded DNA (ssDNA) that is or is not translocated through a nanopore; unlabeled DNA was detected elec. A neg.-AND (NAND) operation was successfully conducted within approx. 10 min, which is rapid compared with previous studies using unlabeled DNA. In addn., this operation was executed in a four-droplet network. DNA mols. and assocd. information were transferred among droplets via biol. nanopores. This system would facilitate linking of mols. and electronic interfaces. Thus, it could be applied to mol. robotics, genetic engineering, and even medical diagnosis and treatment.
- 9Faulhammer, D., Cukras, A. R., Lipton, R. J., and Landweber, L. F. (2000) Molecular computation: RNA solutions to chess problems. Proc. Natl. Acad. Sci. U. S. A. 97, 1385– 1389, DOI: 10.1073/pnas.97.4.13859Molecular computation: RNA solutions to chess problemsFaulhammer, Dirk; Cukras, Anthony R.; Lipton, Richard J.; Landweber, Laura F.Proceedings of the National Academy of Sciences of the United States of America (2000), 97 (4), 1385-1389CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)We have expanded the field of "DNA computers" to RNA and present a general approach for the soln. of satisfiability problems. As an example, we consider a variant of the "Knight problem," which asks generally what configurations of knights can one place on an n × n chess board such that no knight is attacking any other knight on the board. Using specific RNase digestion to manipulate strands of a 10-bit binary RNA library, we developed a mol. algorithm and applied it to a 3×3 chessboard as a 9-bit instance of this problem. Here, the nine spaces on the board correspond to nine "bits" or placeholders in a combinatorial RNA library. We recovered a set of "winning" mols. that describe solns. to this problem.
- 10Choi, J. H. and Ostermeier, M. (2015) Rational design of a fusion protein to exhibit disulfide-mediated logic gate behavior. ACS Synth. Biol. 4, 400– 406, DOI: 10.1021/sb500254g10Rational Design of a Fusion Protein to Exhibit Disulfide-Mediated Logic Gate BehaviorChoi, Jay H.; Ostermeier, MarcACS Synthetic Biology (2015), 4 (4), 400-406CODEN: ASBCD6; ISSN:2161-5063. (American Chemical Society)Synthetic cellular logic gates are primarily built from gene circuits owing to their inherent modularity. Single proteins can also possess logic gate functions and offer the potential to be simpler, quicker, and less dependent on cellular resources than gene circuits. However, the design of protein logic gates that are modular and integrate with other cellular components is a considerable challenge. As a step toward addressing this challenge, we describe the design, construction, and characterization of AND, ORN, and YES logic gates built by introducing disulfide bonds into RG13, a fusion of maltose binding protein and TEM-1 β-lactamase for which maltose is an allosteric activator of enzyme activity. We rationally designed these disulfide bonds to manipulate RG13's allosteric regulation mechanism such that the gating had maltose and reducing agents as input signals, and the gates could be toggled between different gating functions using redox agents, although some gates performed suboptimally.
- 11Fink, T., Lonzarić, J., Praznik, A., Plaper, T., Merljak, E., Leben, K., Jerala, N., Lebar, T., Strmšek, Ž., Lapenta, F., Benčina, M., and Jerala, R. (2019) Design of fast proteolysis-based signaling and logic circuits in mammalian cells. Nat. Chem. Biol. 15, 115– 122, DOI: 10.1038/s41589-018-0181-611Design of fast proteolysis-based signaling and logic circuits in mammalian cellsFink, Tina; Lonzaric, Jan; Praznik, Arne; Plaper, Tjasa; Merljak, Estera; Leben, Katja; Jerala, Nina; Lebar, Tina; Strmsek, Ziga; Lapenta, Fabio; Bencina, Mojca; Jerala, RomanNature Chemical Biology (2019), 15 (2), 115-122CODEN: NCBABT; ISSN:1552-4450. (Nature Research)Cellular signal transduction is predominantly based on protein interactions and their post-translational modifications, which enable a fast response to input signals. Owing to difficulties in designing new unique protein-protein interactions, designed cellular logic has focused on transcriptional regulation; however, that process has a substantially slower response, because it requires transcription and translation. Here, the authors present de novo design of modular, scalable signaling pathways based on proteolysis and designed coiled coils (CC) and implemented in mammalian cells. A set of split proteases with highly specific orthogonal cleavage motifs was constructed and combined with strategically positioned cleavage sites and designed orthogonal CC dimerizing domains with tunable affinity for competitive displacement after proteolytic cleavage. This framework enabled the implementation of Boolean logic functions and signaling cascades in mammalian cells. The designed split-protease-cleavable orthogonal-CC-based (SPOC) logic circuits enable response to chem. or biol. signals within minutes rather than hours and should be useful for diverse medical and nonmedical applications.
- 12Moseley, F., Halámek, J., Kramer, F., Poghossian, A., Schöning, M. J., and Katz, E. (2014) An enzyme-based reversible CNOT logic gate realized in a flow system. Analyst 139, 1839– 1842, DOI: 10.1039/c4an00133h12An enzyme-based reversible CNOT logic gate realized in a flow systemMoseley, Fiona; Halamek, Jan; Kramer, Friederike; Poghossian, Arshak; Schoening, Michael J.; Katz, EvgenyAnalyst (Cambridge, United Kingdom) (2014), 139 (8), 1839-1842CODEN: ANALAO; ISSN:0003-2654. (Royal Society of Chemistry)An enzyme system organized in a flow device was used to mimic a reversible Controlled NOT (CNOT) gate with two input and two output signals. Reversible conversion of NAD+ and NADH cofactors was used to perform a XOR logic operation, while biocatalytic hydrolysis of p-nitrophenyl phosphate resulted in an Identity operation working in parallel. The first biomol. realization of a CNOT gate is promising for integration into complex biomol. networks and future biosensor/biomedical applications.
- 13Stein, V. and Alexandrov, K. (2014) Protease-based synthetic sensing and signal amplification. Proc. Natl. Acad. Sci. U. S. A. 111, 15934– 15939, DOI: 10.1073/pnas.140522011113Protease-based synthetic sensing and signal amplificationStein, Viktor; Alexandrov, KirillProceedings of the National Academy of Sciences of the United States of America (2014), 111 (45), 15934-15939CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The bottom-up design of protein-based signaling networks is a key goal of synthetic biol.; yet, it remains elusive due to our inability to tailor-make signal transducers and receptors that can be readily compiled into defined signaling networks. Here, we report a generic approach for the construction of protein-based mol. switches based on artficially autoinhibited proteases. Using structure-guided design and directed protein evolution, we created signal transducers based on artificially autoinhibited proteases that can be activated following site-specific proteolysis and also demonstrate the modular design of an allosterically regulated protease receptor following recombination with an affinity clamp peptide receptor. Notably, the receptor's mode of action can be varied from >5-fold switch-OFF to >30-fold switch-ON solely by changing the length of the connecting linkers, demonstrating a high functional plasticity not previously obsd. in naturally occurring receptor systems. We also create an integrated signaling circuit based on two orthogonal autoinhibited protease units that can propagate and amplify mol. queues generated by the protease receptor. Finally, we present a generic two-component receptor architecture based on proximity-based activation of two autoinhibited proteases. Overall, the approach allows the design of protease-based signaling networks that, in principle, can be connected to any biol. process.
- 14Zhou, J., Arugula, M. A., Halámek, J., Pita, M., and Katz, E. (2009) Enzyme-based NAND and NOR logic gates with modular design. J. Phys. Chem. B 113, 16065– 16070, DOI: 10.1021/jp907905214Enzyme-Based NAND and NOR Logic Gates with Modular DesignZhou, Jian; Arugula, Mary A.; Halamek, Jan; Pita, Marcos; Katz, EvgenyJournal of Physical Chemistry B (2009), 113 (49), 16065-16070CODEN: JPCBFK; ISSN:1520-6106. (American Chemical Society)The logic gates NAND/NOR were mimicked by enzyme biocatalyzed reactions activated by sucrose, maltose and phosphate. The subunits performing AND/OR Boolean logic operations were designed using maltose phosphorylase and cooperative work of invertase/amyloglucosidase, resp. Glucose produced as the output signal from the AND/OR subunits was applied as the input signal for the INVERTER gate composed of alc. dehydrogenase, glucose oxidase, microperoxidase-11, ethanol and NAD+, which generated the final output in the form of NADH inverting the logic signal from 0 to 1 or from 1 to 0. The final output signal was amplified by a self-promoting biocatalytic system. In order to fulfill the Boolean properties of associativity and commutativity in logic networks, the final NADH output signal was converted to the initial signals of maltose and phosphate, thus allowing assembling of the same std. units in concatenated sequences. The designed modular approach, signal amplification and conversion processes open the way toward complex logic networks composed of std. elements resembling electronic integrated circuitries.
- 15Gao, X. J., Chong, L. S., Kim, M. S., and Elowitz, M. B. (2018) Programmable protein circuits in living cells. Science 361, 1252– 1258, DOI: 10.1126/science.aat506215Programmable protein circuits in living cellsGao, Xiaojing J.; Chong, Lucy S.; Kim, Matthew S.; Elowitz, Michael B.Science (Washington, DC, United States) (2018), 361 (6408), 1252-1258CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Synthetic protein-level circuits could enable engineering of powerful new cellular behaviors. Rational protein circuit design would be facilitated by a composable protein-protein regulation system in which individual protein components can regulate one another to create a variety of different circuit architectures. In this study, we show that engineered viral proteases can function as composable protein components, which can together implement a broad variety of circuit-level functions in mammalian cells. In this system, termed CHOMP (circuits of hacked orthogonal modular proteases), input proteases dock with and cleave target proteases to inhibit their function. These components can be connected to generate regulatory cascades, binary logic gates, and dynamic analog signal-processing functions. To demonstrate the utility of this system, we rationally designed a circuit that induces cell death in response to upstream activators of the Ras oncogene. Because CHOMP circuits can perform complex functions yet be encoded as single transcripts and delivered without genomic integration, they offer a scalable platform to facilitate protein circuit engineering for biotechnol. applications.
- 16Hilburger, C. E., Jacobs, M. L., Lewis, K. R., Peruzzi, J. A., and Kamat, N. P. (2019) Controlling Secretion in Artificial Cells with a Membrane and Gate. ACS Synth. Biol. 8, 1224– 1230, DOI: 10.1021/acssynbio.8b0043516Controlling Secretion in Artificial Cells with a Membrane AND GateHilburger, Claire E.; Jacobs, Miranda L.; Lewis, Kamryn R.; Peruzzi, Justin A.; Kamat, Neha P.ACS Synthetic Biology (2019), 8 (6), 1224-1230CODEN: ASBCD6; ISSN:2161-5063. (American Chemical Society)The assembly of channel proteins into vesicle membranes is a useful strategy to control activities of vesicle-based systems. Here, the authors developed a membrane AND gate that responds to both a fatty acid and a pore-forming channel protein to induce the release of encapsulated cargo. The authors explored how membrane compn. affects the functional assembly of α-hemolysin into phospholipid vesicles as a function of oleic acid content and α-hemolysin concn. The authors then showed that the authors could induce α-hemolysin assembly when the authors added oleic acid micelles to a specific compn. of phospholipid vesicles. Finally, the authors demonstrated that the membrane AND gate could be coupled to a gene expression system. The study provides a new method to control the temporal dynamics of vesicle permeability by controlling when the functional assembly of a channel protein into synthetic vesicles occurs. Furthermore, a membrane AND gate that utilizes membrane-assocg. biomols. introduces a new way to implement Boolean logic that should complement genetic logic circuits and ultimately enhance the capabilities of artificial cellular systems.
- 17Adamala, K. P., Martin-Alarcon, D. A., Guthrie-Honea, K. R., and Boyden, E. S. (2017) Engineering genetic circuit interactions within and between synthetic minimal cells. Nat. Chem. 9, 431– 439, DOI: 10.1038/nchem.264417Engineering 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.
- 18Einfalt, T., Goers, R., Dinu, I. A., Najer, A., Spulber, M., Onaca-Fischer, O., and Palivan, C. G. (2015) Stimuli-Triggered Activity of Nanoreactors by Biomimetic Engineering Polymer Membranes. Nano Lett. 15, 7596– 7603, DOI: 10.1021/acs.nanolett.5b0338618Stimuli-Triggered Activity of Nanoreactors by Biomimetic Engineering Polymer MembranesEinfalt, Tomaz; Goers, Roland; Dinu, Ionel Adrian; Najer, Adrian; Spulber, Mariana; Onaca-Fischer, Ozana; Palivan, Cornelia G.Nano Letters (2015), 15 (11), 7596-7603CODEN: NALEFD; ISSN:1530-6984. (American Chemical Society)The development of advanced stimuli-responsive systems for medicine, catalysis, or technol. requires compartmentalized reaction spaces with triggered activity. Only very few stimuli-responsive systems preserve the compartment architecture, and none allows a triggered activity in situ. We present here a biomimetic strategy to mol. transmembrane transport by engineering synthetic membranes equipped with channel proteins so that they are stimuli-responsive. Nanoreactors with triggered activity were designed by simultaneously encapsulating an enzyme inside polymer compartments, and inserting protein "gates" in the membrane. The outer membrane protein F (OmpF) porin was chem. modified with a pH-responsive mol. cap to serve as "gate" producing pH-driven mol. flow through the membrane and control the in situ enzymic activity. This strategy provides complex reaction spaces necessary in "smart" medicine and for biomimetic engineering of artificial cells.
- 19Einfalt, T., Witzigmann, D., Edlinger, C., Sieber, S., Goers, R., Najer, A., Spulber, M., Onaca-Fischer, O., Huwyler, J., and Palivan, C. G. (2018) Biomimetic artificial organelles with in vitro and in vivo activity triggered by reduction in microenvironment. Nat. Commun. 9, 1127, DOI: 10.1038/s41467-018-03560-x19Biomimetic artificial organelles with in vitro and in vivo activity triggered by reduction in microenvironmentEinfalt T; Edlinger C; Goers R; Najer A; Spulber M; Onaca-Fischer O; Palivan C G; Einfalt T; Witzigmann D; Sieber S; Huwyler J; Goers RNature communications (2018), 9 (1), 1127 ISSN:.Despite tremendous efforts to develop stimuli-responsive enzyme delivery systems, their efficacy has been mostly limited to in vitro applications. Here we introduce, by using an approach of combining biomolecules with artificial compartments, a biomimetic strategy to create artificial organelles (AOs) as cellular implants, with endogenous stimuli-triggered enzymatic activity. AOs are produced by inserting protein gates in the membrane of polymersomes containing horseradish peroxidase enzymes selected as a model for natures own enzymes involved in the redox homoeostasis. The inserted protein gates are engineered by attaching molecular caps to genetically modified channel porins in order to induce redox-responsive control of the molecular flow through the membrane. AOs preserve their structure and are activated by intracellular glutathione levels in vitro. Importantly, our biomimetic AOs are functional in vivo in zebrafish embryos, which demonstrates the feasibility of using AOs as cellular implants in living organisms. This opens new perspectives for patient-oriented protein therapy.
- 20van Meer, G., Voelker, D. R., and Feigenson, G. W. (2008) Membrane lipids: where they are and how they behave. Nat. Rev. Mol. Cell Biol. 9, 112– 124, DOI: 10.1038/nrm233020Membrane lipids: where they are and how they behavevan Meer, Gerrit; Voelker, Dennis R.; Feigenson, Gerald W.Nature Reviews Molecular Cell Biology (2008), 9 (2), 112-124CODEN: NRMCBP; ISSN:1471-0072. (Nature Publishing Group)A review. Throughout the biol. world, a 30 Å hydrophobic film typically delimits the environments that serve as the margin between life and death for individual cells. Biochem. and biophys. findings have provided a detailed model of the compn. and structure of membranes, which includes levels of dynamic organization both across the lipid bilayer (lipid asymmetry) and in the lateral dimension (lipid domains) of membranes. How do cells apply anabolic and catabolic enzymes, translocases, and transporters, plus the intrinsic phys. phase behavior of lipids and their interactions with membrane proteins, to create the unique compns. and multiple functionalities of their individual membranes.
- 21Golynskiy, M. V., Koay, M. S., Vinkenborg, J. L., and Merkx, M. (2011) Engineering protein switches: sensors, regulators, and spare parts for biology and biotechnology. ChemBioChem 12, 353– 361, DOI: 10.1002/cbic.20100064221Engineering Protein Switches: Sensors, Regulators, and Spare Parts for Biology and BiotechnologyGolynskiy, Misha V.; Koay, Melissa S.; Vinkenborg, Jan L.; Merkx, MaartenChemBioChem (2011), 12 (3), 353-361CODEN: CBCHFX; ISSN:1439-4227. (Wiley-VCH Verlag GmbH & Co. KGaA)A review. The authors review the underlying engineering concepts for constructing protein switches, most of which are inspired by or reminiscent of natural examples. The authors focus on those strategies that have the potential to be generally applicable, and discuss them according to their mechanism of action in order of increasing modularity and decreasing domain integration. Finally, the authors identify the opportunities and challenges assocd. with these different engineering concepts and discuss future perspectives.
- 22Arosio, D., Ricci, F., Marchetti, L., Gualdani, R., Albertazzi, L., and Beltram, F. (2010) Simultaneous intracellular chloride and pH measurements using a GFP-based sensor. Nat. Methods 7, 516– 518, DOI: 10.1038/nmeth.147122Simultaneous intracellular chloride and pH measurements using a GFP-based sensorArosio, Daniele; Ricci, Fernanda; Marchetti, Laura; Gualdani, Roberta; Albertazzi, Lorenzo; Beltram, FabioNature Methods (2010), 7 (7), 516-518CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)Chloride and protons perform important closely related roles in many cellular responses. Here we developed a ratiometric biosensor, ClopHensor, based on a highly chloride-sensitive Aequorea victoria GFP variant that is suited for the combined real-time optical detection of pH changes and chloride fluxes in live cells. We detected high chloride concn. in large dense-core exocytosis granules by targeting ClopHensor to these intracellular compartments.
- 23Dal Peraro, M. and van der Goot, F. G. (2016) Pore-forming toxins: ancient, but never really out of fashion. Nat. Rev. Microbiol. 14, 77– 92, DOI: 10.1038/nrmicro.2015.323Pore-forming toxins: ancient, but never really out of fashionDal Peraro Matteo; van der Goot F GisouNature reviews. Microbiology (2016), 14 (2), 77-92 ISSN:.Pore-forming toxins (PFTs) are virulence factors produced by many pathogenic bacteria and have long fascinated structural biologists, microbiologists and immunologists. Interestingly, pore-forming proteins with remarkably similar structures to PFTs are found in vertebrates and constitute part of their immune system. Recently, structural studies of several PFTs have provided important mechanistic insights into the metamorphosis of PFTs from soluble inactive monomers to cytolytic transmembrane assemblies. In this Review, we discuss the diverse pore architectures and membrane insertion mechanisms that have been revealed by these studies, and we consider how these features contribute to binding specificity for different membrane targets. Finally, we explore the potential of these structural insights to enable the development of novel therapeutic strategies that would prevent both the establishment of bacterial resistance and an excessive immune response.
- 24Anderluh, G. and Lakey, J. H. (2008) Disparate proteins use similar architectures to damage membranes. Trends Biochem. Sci. 33, 482– 490, DOI: 10.1016/j.tibs.2008.07.00424Disparate proteins use similar architectures to damage membranesAnderluh, Gregor; Lakey, Jeremy H.Trends in Biochemical Sciences (2008), 33 (10), 482-490CODEN: TBSCDB; ISSN:0968-0004. (Elsevier B.V.)A review. Membrane disruption can efficiently alter cellular function; indeed, pore-forming toxins (PFTs) are well known as important bacterial virulence factors. However, recent data have revealed that structures similar to those found in PFTs are found in membrane active proteins across disparate phyla. Many similarities can be identified only at the 3D-structural level. Of note, domains found in membrane-attack complex proteins of complement and perforin (MACPF) resemble cholesterol-dependent cytolysins from Gram-pos. bacteria, and the Bcl family of apoptosis regulators share similar architectures with Escherichia coli pore-forming colicins. These and other correlations provide considerable help in understanding the structural requirements for membrane binding and pore formation.
- 25Laventie, B., Potrich, C., Atmanène, C., Saleh, M., Joubert, O., Viero, G., Bachmeyer, C., Antonini, V., Mancini, I., Cianferani-Sanglier, S., Keller, D., Colin, D. A., Bourcier, T., Anderluh, G., van Dorsselaer, A., Dalla Serra, M., and Prévost, G. (2013) p-Sulfonato-calix[n]arenes inhibit staphylococcal bicomponent leukotoxins by supramolecular interactions. Biochem. J. 450, 559– 571, DOI: 10.1042/BJ2012162825p-Sulfonato-calix[n]arenes inhibit staphylococcal bicomponent leukotoxins by supramolecular interactionsLaventie, Benoit-Joseph; Potrich, Cristina; Atmanene, Cedric; Saleh, Maher; Joubert, Olivier; Viero, Gabriella; Bachmeyer, Christoph; Antonini, Valeria; Mancini, Ines; Cianferani-Sanglier, Sarah; Keller, Daniel; Colin, Didier A.; Bourcier, Tristan; Anderluh, Gregor; van Dorsselaer, Alain; Dalla Serra, Mauro; Prevost, GillesBiochemical Journal (2013), 450 (3), 559-571CODEN: BIJOAK; ISSN:0264-6021. (Portland Press Ltd.)PVL (Panton-Valentine leukocidin) and other Staphylococcus aureus β-stranded pore-forming toxins are important virulence factors involved in various pathologies that are often necrotizing. The present study characterized leukotoxin inhibition by selected SCns (p-sulfonato-calix[n]arenes): SC4, SC6 and SC8. These chems. have no toxic effects on human erythrocytes or neutrophils, and some are able to inhibit both the activity of and the cell lysis by leukotoxins in a dose-dependent manner. Depending on the type of leukotoxins and SCns, flow cytometry revealed IC50 values of 6-22 μM for Ca2+ activation and of 2-50 μM for cell lysis. SCns were obsd. to affect membrane binding of class S proteins responsible for cell specificity. Electrospray MS and surface plasmon resonance established supramol. interactions (1:1 stoichiometry) between SCns and class S proteins in soln., but not class F proteins. The membrane-binding affinity of S proteins was Kd=0.07-6.2 nM. The binding ability was completely abolished by SCns at different concns. according to the no. of benzenes (30-300 μM; SC8>SC6»SC4). The inhibitory properties of SCns were also obsd. in vivo in a rabbit model of PVL-induced endophthalmitis. These calixarenes may represent new therapeutic avenues aimed at minimizing inflammatory reactions and necrosis due to certain virulence factors.
- 26Booth, M. J., Schild, V. R., Graham, A. D., Olof, S. N., and Bayley, H. (2016) Light-activated communication in synthetic tissues. Sci. Adv. 2, e1600056 DOI: 10.1126/sciadv.160005626Light-activated communication in synthetic tissuesBooth, Michael J.; Schild, Vanessa Restrepo; Graham, Alexander D.; Olof, Sam N.; Bayley, HaganScience Advances (2016), 2 (4), e1600056/1-e1600056/11CODEN: SACDAF; ISSN:2375-2548. (American Association for the Advancement of Science)We have previously used three-dimensional (3D) printing to prep. tissue-like materials in which picoliter aq. compartments are sepd. by lipid bilayers. These printed droplets are elaborated into synthetic cells by using a tightly regulated in vitro transcription/translation system. A light-activated DNA promoter has been developed that can be used to turn on the expression of any gene within the synthetic cells. We used light activation to express protein pores in 3D-printed patterns within synthetic tissues. The pores are incorporated into specific bilayer interfaces and thereby mediate rapid, directional elec. communication between subsets of cells. Accordingly, we have developed a functional mimic of neuronal transmission that can be controlled in a precise way.
- 27Provoda, C. J., Stier, E. M., and Lee, K. D. (2003) Tumor cell killing enabled by listeriolysin O-liposome-mediated delivery of the protein toxin gelonin. J. Biol. Chem. 278, 35102– 35108, DOI: 10.1074/jbc.M30541120027Tumor cell killing enabled by Listeriolysin O-liposome-mediated delivery of the protein toxin geloninProvoda, Chester J.; Stier, Ethan M.; Lee, Kyung-DallJournal of Biological Chemistry (2003), 278 (37), 35102-35108CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)Gelonin is a type I plant toxin that has potential as an effective anti-tumor agent by virtue of its enzymic capacity to inactivate ribosomes and arrest protein synthesis, thereby effectively limiting the growth of cancer cells. Being a hydrophilic macromol., however, gelonin has limited access to its target subcellular compartment, the cytosol; it is effectively plasma membrane-impermeant and subject to rapid degrdn. within endosomes and lysosomes upon cellular uptake as it lacks the membrane-translocating capability that is typically provided by a disulfide-linked B polypeptide found in the type II toxins (e.g. ricin). These inherent characteristics generate the need for the development of a specialized cytosolic delivery strategy for gelonin as an effective anti-tumor therapeutic agent. Here we describe an efficient means of delivering gelonin to the cytosol of B16 melanoma cells. Gelonin was co-encapsulated inside pH-sensitive liposomes with listeriolysin O, the pore-forming protein that mediates escape of the intracellular pathogen Listeria monocytogenes from the endosome into the cytosol. In in vitro expts., co-encapsulated listeriolysin O enabled liposomal gelonin-mediated B16 cell killing with a gelonin IC50 of ∼0.1 nM with an extreme efficiency requiring an incubation time of only 1 h. By contrast, cells treated with equiv. concns. of unencapsulated gelonin or gelonin encapsulated alone in pH-sensitive liposomes exhibited no detectable cytotoxicity. Moreover, treatment by direct intratumor injection into s.c. solid tumors of B16 melanoma in a mouse model showed that pH-sensitive liposomes contg. both listeriolysin O and gelonin were more effective than control formulations in curtailing tumor growth rates.
- 28Robertson, J. W. F., Rodrigues, C. G., Stanford, V. M., Rubinson, K. A., Krasilnikov, O. V., and Kasianowicz, J. J. (2007) Single-molecule mass spectrometry in solution using a solitary nanopore. Proc. Natl. Acad. Sci. U. S. A. 104, 8207– 8211, DOI: 10.1073/pnas.061108510428Single-molecule mass spectrometry in solution using a solitary nanoporeRobertson, Joseph W. F.; Rodrigues, Claudio G.; Stanford, Vincent M.; Rubinson, Kenneth A.; Krasilnikov, Oleg V.; Kasianowicz, John J.Proceedings of the National Academy of Sciences of the United States of America (2007), 104 (20), 8207-8211CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)The authors introduce a 2-dimensional method for mass spectrometry in soln. that is based on the interaction between a nanometer-scale pore and analytes. As an example, poly(ethylene glycol) mols. that enter a single α-hemolysin pore cause distinct mass-dependent conductance states with characteristic mean residence times. The conductance-based mass spectrum clearly resolves the repeat unit of ethylene glycol, and the mean residence time increases monotonically with the poly(ethylene glycol) mass. This technique could prove useful for the real-time characterization of mols. in soln.
- 29Clarke, J., Wu, H., Jayasinghe, L., Patel, A., Reid, S., and Bayley, H. (2009) Continuous base identification for single-molecule nanopore DNA sequencing. Nat. Nanotechnol. 4, 265– 270, DOI: 10.1038/nnano.2009.1229Continuous base identification for single-molecule nanopore DNA sequencingClarke, James; Wu, Hai-Chen; Jayasinghe, Lakmal; Patel, Alpesh; Reid, Stuart; Bayley, HaganNature Nanotechnology (2009), 4 (4), 265-270CODEN: NNAABX; ISSN:1748-3387. (Nature Publishing Group)A single-mol. method for sequencing DNA that does not require fluorescent labeling could reduce costs and increase sequencing speeds. An exonuclease enzyme might be used to cleave individual nucleotide mols. from the DNA, and when coupled to an appropriate detection system, these nucleotides could be identified in the correct order. Here, the authors show that a protein nanopore with a covalently attached adapter mol. can continuously identify unlabeled nucleoside 5'-monophosphate mols. with accuracies averaging 99.8%. Methylated cytosine can also be distinguished from the four std. DNA bases: guanine, adenine, thymine and cytosine. The operating conditions are compatible with the exonuclease, and the kinetic data show that the nucleotides have a high probability of translocation through the nanopore and, therefore, of not being registered twice. This highly accurate tool is suitable for integration into a system for sequencing nucleic acids and for analyzing epigenetic modifications.
- 30Wang, Y., Montana, V., Grubišić, V., Stout, R. F., Parpura, V., and Gu, L. Q. (2015) Nanopore sensing of botulinum toxin type B by discriminating an enzymatically cleaved peptide from a synaptic protein synaptobrevin 2 derivative. ACS Appl. Mater. Interfaces 7, 184– 192, DOI: 10.1021/am505659630Nanopore Sensing of Botulinum Toxin Type B by Discriminating an Enzymatically Cleaved Peptide from a Synaptic Protein Synaptobrevin 2 DerivativeWang, Yong; Montana, Vedrana; Grubisic, Vladimir; Stout, Randy F.; Parpura, Vladimir; Gu, Li-QunACS Applied Materials & Interfaces (2015), 7 (1), 184-192CODEN: AAMICK; ISSN:1944-8244. (American Chemical Society)Botulinum neurotoxins (BoNTs) are the most lethal toxin known to human. Biodefense requires early and rapid detection of BoNTs. Traditionally, BoNTs can be detected by looking for signs of botulism in mice that receive an injection of human material, serum or stool. While the living animal assay remains the most sensitive approach, it is costly, slow and assocd. with legal and ethical constrains. Various biochem., optical and mech. methods have been developed for BoNTs detection with improved speed, but with lesser sensitivity. Here, we report a novel nanopore-based BoNT type B (BoNT-B) sensor that monitors the toxin's enzymic activity on its substrate, a recombinant synaptic protein synaptobrevin 2 deriv. By analyzing the modulation of the pore current caused by the specific BoNT-B-digested peptide as a marker, the presence of BoNT-B at a subnanomolar concn. was identified within minutes. The nanopore detector would fill the niche for a much needed rapid and highly sensitive detection of neurotoxins, and provide an excellent system to explore biophys. mechanisms for biopolymer transportation.
- 31Wang, Y., Zheng, D., Tan, Q., Wang, M., and Gu, L. (2011) Nanopore-based detection of circulating microRNAs in lung cancer patients. Nat. Nanotechnol. 6, 668– 674, DOI: 10.1038/nnano.2011.14731Nanopore-based detection of circulating microRNAs in lung cancer patientsWang, Yong; Zheng, Da-Li; Tan, Qiu-Lin; Wang, Michael X.; Gu, Li-QunNature Nanotechnology (2011), 6 (10), 668-674CODEN: NNAABX; ISSN:1748-3387. (Nature Publishing Group)MicroRNAs are short RNA mols. that regulate gene expression, and have been investigated as potential biomarkers because their expression levels are correlated with various diseases. However, detecting microRNAs in the bloodstream remains difficult because current methods are not sufficiently selective or sensitive. Here, we show that a nanopore sensor based on the α-haemolysin protein can selectively detect microRNAs at the single mol. level in plasma samples from lung cancer patients without the need for labels or amplification of the microRNA. The sensor, which uses a programmable oligonucleotide probe to generate a target-specific signature signal, can quantify subpicomolar levels of cancer-assocd. microRNAs and can distinguish single-nucleotide differences between microRNA family members. This approach is potentially useful for quant. microRNA detection, the discovery of disease markers and non-invasive early diagnosis of cancer.
- 32Braha, O., Bayley, H., Gu, L. Q., Zhou, L., Lu, X., and Cheley, S. (2000) Simultaneous stochastic sensing of divalent metal ions. Nat. Biotechnol. 18, 1005– 1007, DOI: 10.1038/7927532Simultaneous stochastic sensing of divalent metal ionsBraha, Orit; Gu, Lin-Qun; Zhou, Li; Lu, Xiaofeng; Cheley, Stephen; Bayley, HaganNature Biotechnology (2000), 18 (9), 1005-1007CODEN: NABIF9; ISSN:1087-0156. (Nature America Inc.)Stochastic sensing is an emerging anal. technique that relies upon single-mol. detection. Transmembrane pores, into which binding sites for analytes have been placed by genetic engineering, have been developed as stochastic sensing elements. Reversible occupation of an engineered binding site modulates the ionic current passing through a pore in a transmembrane potential and thereby provides both the concn. of an analyte and, through a characteristic signature, its identity. Here, we show that the concns. of two or more divalent metal ions in soln. can be detd. simultaneously with a single sensor element. Further, the sensor element can be permanently calibrated without a detailed understanding of the kinetics of interaction of the metal ions with the engineered pore.
- 33Villar, G., Graham, A. D., and Bayley, H. (2013) A tissue-like printed material. Science 340, 48– 52, DOI: 10.1126/science.122949533A Tissue-Like Printed MaterialVillar, Gabriel; Graham, Alexander D.; Bayley, HaganScience (Washington, DC, United States) (2013), 340 (6128), 48-52CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)Living cells communicate and cooperate to produce the emergent properties of tissues. Synthetic mimics of cells, such as liposomes, are typically incapable of cooperation and therefore cannot readily display sophisticated collective behavior. We printed tens of thousands of picoliter aq. droplets that become joined by single lipid bilayers to form a cohesive material with cooperating compartments. Three-dimensional structures can be built with heterologous droplets in software-defined arrangements. The droplet networks can be functionalized with membrane proteins; for example, to allow rapid elec. communication along a specific path. The networks can also be programmed by osmolarity gradients to fold into otherwise unattainable designed structures. Printed droplet networks might be interfaced with tissues, used as tissue engineering substrates, or developed as mimics of living tissue.
- 34Elani, Y., Law, R. V., and Ces, O. (2014) Vesicle-based artificial cells as chemical microreactors with spatially segregated reaction pathways. Nat. Commun. 5, 5305, DOI: 10.1038/ncomms630534Vesicle-based artificial cells as chemical microreactors with spatially segregated reaction pathwaysElani, Yuval; Law, Robert V.; Ces, OscarNature Communications (2014), 5 (), 5305CODEN: NCAOBW; ISSN:2041-1723. (Nature Publishing Group)In the discipline of bottom-up synthetic biol., vesicles define the boundaries of artificial cells and are increasingly being used as biochem. microreactors operating in physiol. environments. As the field matures, there is a need to compartmentalize processes in different spatial localities within vesicles, and for these processes to interact with one another. Here we address this by designing and constructing multi-compartment vesicles within which an engineered multi-step enzymic pathway is carried out. The individual steps are isolated in distinct compartments, and their products traverse into adjacent compartments with the aid of transmembrane protein pores, initiating subsequent steps. Thus, an engineered signalling cascade is recreated in an artificial cellular system. Importantly, by allowing different steps of a chem. pathway to be sepd. in space, this platform bridges the gap between table-top chem. and chem. that is performed within vesicles.
- 35Hamon, M. A., Ribet, D., Stavru, F., and Cossart, P. (2012) Listeriolysin O: the Swiss army knife of Listeria. Trends Microbiol. 20, 360– 368, DOI: 10.1016/j.tim.2012.04.00635Listeriolysin O: the Swiss army knife of ListeriaHamon, Melanie Anne; Ribet, David; Stavru, Fabrizia; Cossart, PascaleTrends in Microbiology (2012), 20 (8), 360-368CODEN: TRMIEA; ISSN:0966-842X. (Elsevier Ltd.)A review. Listeriolysin O (LLO) is a toxin produced by Listeria monocytogenes, an opportunistic bacterial pathogen responsible for the disease listeriosis. This disease starts with the ingestion of contaminated foods and mainly affects immunocompromised individuals, newborns, and pregnant women. In the lab., L. monocytogenes is used as a model organism to study processes such as cell invasion, intracellular survival, and cell-to-cell spreading, as this Gram-pos. bacterium has evolved elaborate mol. strategies to subvert host cell functions. LLO is a major virulence factor originally shown to be crucial for bacterial escape from the internalization vacuole after entry into cells. However, recent studies are revisiting the role of LLO during infection and are revealing new insights into the action of LLO, in particular before bacterial entry. These latest findings along with their impact on the infectious process will be discussed.
- 36Köster, S., van Pee, K., Hudel, M., Leustik, M., Rhinow, D., Kühlbrandt, W., Chakraborty, T., and Yildiz, O. (2014) Crystal structure of listeriolysin O reveals molecular details of oligomerization and pore formation. Nat. Commun. 5, 3690, DOI: 10.1038/ncomms469036Crystal structure of listeriolysin O reveals molecular details of oligomerization and pore formationKoster Stefan; van Pee Katharina; Rhinow Daniel; Kuhlbrandt Werner; Yildiz Ozkan; Hudel Martina; Leustik Martin; Chakraborty TrinadNature communications (2014), 5 (), 3690 ISSN:.Listeriolysin O (LLO) is an essential virulence factor of Listeria monocytogenes that causes listeriosis. Listeria monocytogenes owes its ability to live within cells to the pH- and temperature-dependent pore-forming activity of LLO, which is unique among cholesterol-dependent cytolysins. LLO enables the bacteria to cross the phagosomal membrane and is also involved in activation of cellular processes, including the modulation of gene expression or intracellular Ca(2+) oscillations. Neither the pore-forming mechanism nor the mechanisms triggering the signalling processes in the host cell are known in detail. Here, we report the crystal structure of LLO, in which we identified regions important for oligomerization and pore formation. Mutants were characterized by determining their haemolytic and Ca(2+) uptake activity. We analysed the pore formation of LLO and its variants on erythrocyte ghosts by electron microscopy and show that pore formation requires precise interface interactions during toxin oligomerization on the membrane.
- 37Ruan, Y., Rezelj, S., Bedina Zavec, A., Anderluh, G., and Scheuring, S. (2016) Listeriolysin O membrane damaging activity involves arc formation and lineaction - implication for Listeria monocytogenes escape from phagocytic vacuole. PLoS Pathog. 12, e1005597 DOI: 10.1371/journal.ppat.100559737Listeriolysin O membrane damaging activity involves arc formation and lineaction - implication for Listeria monocytogenes escape from phagocytic vacuoleRuan, Yi; Rezelj, Sasa; Zavec, Apolonija Bedina; Anderluh, Gregor; Scheuring, SimonPLoS Pathogens (2016), 12 (4), e1005597/1-e1005597/18CODEN: PPLACN; ISSN:1553-7374. (Public Library of Science)Listeriolysin-O (LLO) plays a crucial role during infection by Listeria monocytogenes. It enables escape of bacteria from phagocytic vacuole, which is the basis for its spread to other cells and tissues. It is not clear how LLO acts at phagosomal membranes to allow bacterial escape. The mechanism of action of LLO remains poorly understood, probably due to unavailability of suitable exptl. tools that could monitor LLO membrane disruptive activity in real time. Here, we used high-speed at. force microscopy (HS-AFM) featuring high spatio-temporal resoln. on model membranes and optical microscopy on giant unilamellar vesicles (GUVs) to investigate LLO activity. We analyze the assembly kinetics of toxin oligomers, the prepore-to-pore transition dynamics and the membrane disruption in real time. We reveal that LLO toxin efficiency and mode of action as a membrane-disrupting agent varies strongly depending on the membrane cholesterol concn. and the environmental pH. We discovered that LLO is able to form arc pores as well as damage lipid membranes as a lineactant, and this leads to large-scale membrane defects. These results altogether provide a mechanistic basis of how large-scale membrane disruption leads to release of Listeria from the phagocytic vacuole in the cellular context.
- 38Podobnik, M., Marchioretto, M., Zanetti, M., Bavdek, A., Kisovec, M., Cajnko, M. M., Lunelli, L., Serra, M. D., and Anderluh, G. (2015) Plasticity of lysteriolysin O pores and its regulation by pH and unique histidine. Sci. Rep. 5, 9623, DOI: 10.1038/srep0962338Plasticity of Listeriolysin O Pores and its Regulation by pH and Unique HistidinePodobnik, Marjetka; Marchioretto, Marta; Zanetti, Manuela; Bavdek, Andrej; Kisovec, Matic; Cajnko, Misa Mojca; Lunelli, Lorenzo; Dalla Serra, Mauro; Anderluh, GregorScientific Reports (2015), 5 (), 9623/1-9623/10CODEN: SRCEC3; ISSN:2045-2322. (Nature Publishing Group)Pore formation of cellular membranes is an ancient mechanism of bacterial pathogenesis that allows efficient damaging of target cells. Several mechanisms have been described, however, relatively little is known about the assembly and properties of pores. Listeriolysin O (LLO) is a pH-regulated cholesterol-dependent cytolysin from the intracellular pathogen Listeria monocytogenes, which forms transmembrane β-barrel pores. Here we report that the assembly of LLO pores is rapid and efficient irresp. of pH. While pore diams. at the membrane surface are comparable at either pH 5.5 or 7.4, the distribution of pore conductances is significantly pH-dependent. This is directed by the unique residue H311, which is also important for the conformational stability of the LLO monomer and the rate of pore formation. The functional pores exhibit variations in height profiles and can reconfigure significantly by merging to other full pores or arcs. Our results indicate significant plasticity of large β-barrel pores, controlled by environmental cues like pH.
- 39Mulvihill, E., Van Pee, K., Mari, S. A., Müller, D. J., and Yildiz, Ö. (2015) Directly observing the lipid-dependent self-assembly and pore-forming mechanism of the cytolytic toxin listeriolysin O. Nano Lett. 15, 6965– 6973, DOI: 10.1021/acs.nanolett.5b0296339Directly Observing the Lipid-Dependent Self-Assembly and Pore-Forming Mechanism of the Cytolytic Toxin Listeriolysin OMulvihill Estefania; Mari Stefania A; Muller Daniel J; van Pee Katharina; Yildiz OzkanNano letters (2015), 15 (10), 6965-73 ISSN:.Listeriolysin O (LLO) is the major virulence factor of Listeria monocytogenes and a member of the cholesterol-dependent cytolysin (CDC) family. Gram-positive pathogenic bacteria produce water-soluble CDC monomers that bind cholesterol-dependent to the lipid membrane of the attacked cell or of the phagosome, oligomerize into prepores, and insert into the membrane to form transmembrane pores. However, the mechanisms guiding LLO toward pore formation are poorly understood. Using electron microscopy and time-lapse atomic force microscopy, we show that wild-type LLO binds to membranes, depending on the presence of cholesterol and other lipids. LLO oligomerizes into arc- or slit-shaped assemblies, which merge into complete rings. All three oligomeric assemblies can form transmembrane pores, and their efficiency to form pores depends on the cholesterol and the phospholipid composition of the membrane. Furthermore, the dynamic fusion of arcs, slits, and rings into larger rings and their formation of transmembrane pores does not involve a height difference between prepore and pore. Our results reveal new insights into the pore-forming mechanism and introduce a dynamic model of pore formation by LLO and other CDC pore-forming toxins.
- 40Kisovec, M., Rezelj, S., Knap, P., Cajnko, M., Caserman, S., Flašker, A., Žnidaršič, N., Repič, M., Mavri, J., Ruan, Y., Scheuring, S., Podobnik, M., and Anderluh, G. (2017) Engineering a pH responsive pore forming protein. Sci. Rep. 7, 42231, DOI: 10.1038/srep4223140Engineering a pH responsive pore forming proteinKisovec, Matic; Rezelj, Sasa; Knap, Primoz; Cajnko, Misa Mojca; Caserman, Simon; Flasker, Ajda; Znidarsic, Nada; Repic, Matej; Mavri, Janez; Ruan, Yi; Scheuring, Simon; Podobnik, Marjetka; Anderluh, GregorScientific Reports (2017), 7 (), 42231CODEN: SRCEC3; ISSN:2045-2322. (Nature Publishing Group)Listeriolysin O (LLO) is a cytolysin capable of forming pores in cholesterol-rich lipid membranes of host cells. It is conveniently suited for engineering a pH-governed responsiveness, due to a pH sensor identified in its structure that was shown before to affect its stability. Here we introduced a new level of control of its hemolytic activity by making a variant with hemolytic activity that was pH-dependent. Based on detailed structural anal. coupled with mol. dynamics and mutational anal., we found that the bulky side chain of Tyr406 allosterically affects the pH sensor. Mol. dynamics simulation further suggested which other amino acid residues may also allosterically influence the pH-sensor. LLO was engineered to the point where it can, in a pH-regulated manner, perforate artificial and cellular membranes. The single mutant Tyr406Ala bound to membranes and oligomerized similarly to the wild-type LLO, however, the final membrane insertion step was pH-affected by the introduced mutation. We show that the mutant toxin can be activated at the surface of artificial membranes or living cells by a single wash with slightly acidic pH buffer. Y406A mutant has a high potential in development of novel nanobiotechnol. applications such as controlled release of substances or as a sensor of environmental pH.
- 41Dreier, B., and Plückthun, A. (2011) Ribosome Display: A Technology for Selecting and Evolving Proteins from Large Libraries, in PCR Protocols. Methods in Molecular Biology (Methods and Protocols), (Park, D. J., Ed.), pp 283– 306, Humana Press, Totowa, NJ.There is no corresponding record for this reference.
- 42Binz, H. K., Amstutz, P., Kohl, A., Stumpp, M. T., Briand, C., Forrer, P., Grütter, M. G., and Plückthun, A. (2004) High-affinity binders selected from designed ankyrin repeat protein libraries. Nat. Biotechnol. 22, 575– 82, DOI: 10.1038/nbt96242High-affinity binders selected from designed ankyrin repeat protein librariesBinz, H. Kaspar; Amstutz, Patrick; Kohl, Andreas; Stumpp, Michael T.; Briand, Christophe; Forrer, Patrik; Gruetter, Markus G.; Plueckthun, AndreasNature Biotechnology (2004), 22 (5), 575-582CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)We report here the evolution of ankyrin repeat (AR) proteins in vitro for specific, high-affinity target binding. Using a consensus design strategy, we generated combinatorial libraries of AR proteins of varying repeat nos. with diversified binding surfaces. Libraries of two and three repeats, flanked by 'capping repeats,' were used in ribosome-display selections against maltose binding protein (MBP) and two eukaryotic kinases. We rapidly enriched target-specific binders with affinities in the low nanomolar range and detd. the crystal structure of one of the selected AR proteins in complex with MBP at 2.3 Å resoln. The interaction relies on the randomized positions of the designed AR protein and is comparable to natural, heterodimeric protein-protein interactions. Thus, our AR protein libraries are valuable sources for binding mols. and, because of the very favorable biophys. properties of the designed AR proteins, an attractive alternative to antibody libraries.
- 43Bavdek, A., Gekara, N. O., Priselac, D., Gutiérrez Aguirre, I., Darji, A., Chakraborty, T., Macek, P., Lakey, J. H., Weiss, S., and Anderluh, G. (2007) Sterol and pH interdependence in the binding, oligomerization, and pore formation of Listeriolysin O. Biochemistry 46, 4425– 4437, DOI: 10.1021/bi602497g43Sterol and pH Interdependence in the Binding, Oligomerization, and Pore Formation of Listeriolysin OBavdek, Andrej; Gekara, Nelson O.; Priselac, Dragan; Gutierrez Aguirre, Ion; Darji, Ayub; Chakraborty, Trinad; Macek, Peter; Lakey, Jeremy H.; Weiss, Siegfried; Anderluh, GregorBiochemistry (2007), 46 (14), 4425-4437CODEN: BICHAW; ISSN:0006-2960. (American Chemical Society)Listeriolysin O (LLO) is the most important virulence factor of the intracellular pathogen Listeria monocytogenes. Its main task is to enable escape of bacteria from the phagosomal vacuole into the cytoplasm. LLO belongs to the cholesterol-dependent cytolysin (CDC) family but differs from other members, as it exhibits optimal activity at low pH. Its pore forming ability at higher pH values has been largely disregarded in Listeria pathogenesis. Here we show that high cholesterol concns. in the membrane restore the low activity of LLO at high pH values. LLO binds to lipid membranes, at physiol. or even slightly basic pH values, in a cholesterol-dependent fashion. Binding, insertion into lipid monolayers, and permeabilization of calcein-loaded liposomes are maximal above approx. 35 mol % cholesterol, a concn. range typically found in lipid rafts. The narrow transition region of cholesterol concn. sepg. low and high activity indicates that cholesterol not only allows the binding of LLO to membranes but also affects other steps in pore formation. We were able to detect some of these by surface plasmon resonance-based assays. In particular, we show that LLO recognition of cholesterol is detd. by the most exposed 3β-hydroxy group of cholesterol. In addn., LLO binds and permeabilizes J774 cells and human erythrocytes in a cholesterol-dependent fashion at physiol. or slightly basic pH values. The results clearly show that LLO activity at physiol. pH cannot be neglected and that its action at sites distal to cell entry may have important physiol. consequences for Listeria pathogenesis.
- 44Sallee, N. A., Yeh, B. J., and Lim, W. A. (2007) Engineering modular protein interaction switches by sequence overlap. J. Am. Chem. Soc. 129, 4606– 4611, DOI: 10.1021/ja067272844Engineering Modular Protein Interaction Switches by Sequence OverlapSallee, Nathan A.; Yeh, Brian J.; Lim, Wendell A.Journal of the American Chemical Society (2007), 129 (15), 4606-4611CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Many cellular signaling pathways contain proteins whose interactions change in response to upstream inputs, allowing for conditional activation or repression of the interaction based on the presence of the input mol. The ability to engineer similar regulation into protein interaction elements would provide us with powerful tools for controlling cell signaling. Here we describe an approach for engineering diverse synthetic protein interaction switches. Specifically, by overlapping the sequences of pairs of protein interaction domains and peptides, we have been able to generate mutually exclusive regulation over their interactions. Thus, the hybrid protein (which is composed of the two overlapped interaction modules) can bind to either of the two resp. ligands for those modules, but not to both simultaneously. We show that these synthetic switch proteins can be used to regulate specific protein-protein interactions in vivo. These switches allow us to disrupt an interaction with the addn. or activation of a protein input that has no natural connection to the interaction in question. Therefore, they give us the ability to make novel connections between normally unrelated signaling pathways and to rewire the input/output relationships of cellular behaviors. Our expts. also suggest a possible mechanism by which complex regulatory proteins might have evolved from simpler components.
- 45Karginov, A. V., Ding, F., Kota, P., Dokholyan, N. V., and Hahn, K. M. (2010) Engineered allosteric activation of kinases in living cells. Nat. Biotechnol. 28, 743– 747, DOI: 10.1038/nbt.163945Engineered allosteric activation of kinases in living cellsKarginov, Andrei V.; Ding, Feng; Kota, Pradeep; Dokholyan, Nikolay V.; Hahn, Klaus M.Nature Biotechnology (2010), 28 (7), 743-747CODEN: NABIF9; ISSN:1087-0156. (Nature Publishing Group)Studies of cellular and tissue dynamics benefit greatly from tools that can control protein activity with specificity and precise timing in living systems. Here we describe an approach to confer allosteric regulation specifically on the catalytic activity of protein kinases. A highly conserved portion of the kinase catalytic domain is modified with a small protein insert that inactivates catalytic activity but does not affect other protein functions. Catalytic activity is restored by addn. of rapamycin or non-immunosuppresive rapamycin analogs. Mol. modeling and mutagenesis indicate that the protein insert reduces activity by increasing the flexibility of the catalytic domain. Drug binding restores activity by increasing rigidity. We demonstrate the approach by specifically activating focal adhesion kinase (FAK) within minutes in living cells and show that FAK is involved in the regulation of membrane dynamics. Successful regulation of Src and p38 by insertion of the rapamycin-responsive element at the same conserved site used in FAK suggests that our strategy will be applicable to other kinases.
- 46Edwards, W. R., Busse, K., Allemann, R. K., and Jones, D. D. (2008) Linking the functions of unrelated proteins using a novel directed evolution domain insertion method. Nucleic Acids Res. 36, e78 DOI: 10.1093/nar/gkn36346Linking the functions of unrelated proteins using a novel directed evolution domain insertion methodEdwards, Wayne R.; Busse, Kathy; Allemann, Rudolf K.; Jones, D. DafyddNucleic Acids Research (2008), 36 (13), e78/1-e78/9CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)We have successfully developed a new directed evolution method for generating integral protein fusions comprising of one domain inserted within another. Creating two connections between the insert and accepting parent domain can result in the inter-dependence of the sep. protein activities, thus providing a general strategy for constructing mol. switches. Using an engineered transposon termed MuDel, contiguous trinucleotide sequences were removed at random positions from the bla gene encoding TEM-1 β-lactamase. The deleted trinucleotide sequence was then replaced by a DNA cassette encoding cytochrome b562 with differing linking sequences at each terminus and sampling all three reading frames. The result was a variety of chimeric genes encoding novel integral fusion proteins that retained TEM-1 activity. While most of the tolerated insertions were obsd. in loops, several also occurred close to the termini of α-helixes and β-strands. Several variants conferred a switching phenotype on Escherichia coli, with bacterial tolerance to ampicillin being dependent on the presence of haem in the growth medium. The magnitude of the switching phenotype ranged from 4- to 128-fold depending on the insertion position within TEM-1 and the linker sequences that join the two domains.
- 47Ha, J. H., Butler, J. S., Mitrea, D. M., and Loh, S. N. (2006) Modular enzyme design: Regulation by mutually exclusive protein folding. J. Mol. Biol. 357, 1058– 1062, DOI: 10.1016/j.jmb.2006.01.07347Modular Enzyme Design: Regulation by Mutually Exclusive Protein FoldingHa, Jeung-Hoi; Butler, James S.; Mitrea, Diana M.; Loh, Stewart N.Journal of Molecular Biology (2006), 357 (4), 1058-1062CODEN: JMOBAK; ISSN:0022-2836. (Elsevier B.V.)A regulatory mechanism is introduced whereupon the catalytic activity of a given enzyme is controlled by ligand binding to a receptor domain of choice. A small enzyme (barnase) and a ligand-binding polypeptide (GCN4) are fused so that a simple topol. constraint prevents them from existing simultaneously in their folded states. The two domains consequently engage in a thermodn. tug-of-war in which the more stable domain forces the less stable domain to unfold. In the absence of ligand, the barnase domain is more stable and is therefore folded and active; the GCN4 domain is substantially unstructured. DNA binding induces folding of GCN4, forcibly unfolding and inactivating the barnase domain. Barnase-GCN4 is thus a "natively unfolded" protein that uses ligand binding to switch between partially folded forms. The key characteristics of each parent protein (catalytic efficiency of barnase, DNA binding affinity and sequence specificity of GCN4) are retained in the chimera. Barnase-GCN4 thus defines a modular approach for assembling enzymes with novel sensor capabilities from a variety of catalytic and ligand binding domains.
- 48Tweten, R. K., Hotze, E. M., and Wade, K. R. (2015) The unique molecular choreography of giant pore formation by the cholesterol-dependent cytolysins of Gram-positive bacteria. Annu. Rev. Microbiol. 69, 323– 340, DOI: 10.1146/annurev-micro-091014-10423348The Unique Molecular Choreography of Giant Pore Formation by the Cholesterol-Dependent Cytolysins of Gram-Positive BacteriaTweten, Rodney K.; Hotze, Eileen M.; Wade, Kristin R.Annual Review of Microbiology (2015), 69 (), 323-340CODEN: ARMIAZ; ISSN:0066-4227. (Annual Reviews)A review. The mechanism by which the cholesterol-dependent cytolysins (CDCs) assemble their giant β-barrel pore in cholesterol-rich membranes has been the subject of intense study in the past two decades. A combination of structural, biophys., and biochem. analyses revealed deep insights into the series of complex and highly choreographed secondary and tertiary structural transitions that the CDCs undergo to assemble their β-barrel pore in eukaryotic membranes. The authors' knowledge of the mol. details of these dramatic structural changes in CDCs has transformed the understanding of how giant pore complexes are assembled and has been crit. to the understanding of the mechanisms of other important classes of pore-forming toxins and proteins across the kingdoms of life. Finally, there are tantalizing hints that the CDC pore-forming mechanism is more sophisticated than previously imagined and that some CDCs were employed in pore-independent processes.
- 49Song, L., Hobaugh, M. R., Shustak, C., Cheley, S., Bayley, H., and Gouaux, J. E. (1996) Structure of staphylococcal alpha-hemolysin, a heptameric transmembrane pore. Science 274, 1859– 1866, DOI: 10.1126/science.274.5294.185949Structure of staphylococcal α-hemolysin, a heptameric transmembrane poreSong, Langzhou; Hobaugh, Michael R.; Shustak, Christopher; Cheley, Stephen; Bayley, Hagan; Gouaux, J. EricScience (Washington, D. C.) (1996), 274 (5294), 1859-1866CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)The structure of the Staphylococcus aureus α-hemolysin pore has been detd. to 1.9 Å resoln. Contained within the mushroom-shaped homo-oligomeric heptamer is a solvent-filled channel, 100 Å in length, that runs along the sevenfold axis and ranges from 14 Å to 46 Å in diam. The lytic, transmembrane domain comprises the lower half of a 14-strand antiparallel β barrel, to which each promoter contributes two β strands, each 65 Å long. The interior of the β barrel is primarily hydrophilic, and the exterior has a hydrophobic belt 28 Å wide. The structure proves the heptameric subunit stoichiometry of the α-hemolysin oligomer, shows that a glycine-rich and solvent-exposed region of a water-sol. protein can self-assemble to form a transmembrane pore of defined structure, and provides insight into the principles of membrane interaction and transport activity of β barrel pore-forming toxins.
- 50Podobnik, M., Savory, P., Rojko, N., Kisovec, M., Wood, N., Hambley, R., Pugh, J., Wallace, E. J., McNeill, L., Bruce, M., Liko, I., Allison, T. M., Mehmood, S., Yilmaz, N., Kobayashi, T., Gilbert, R. J. C., Robinson, C. V., Jayasinghe, L., and Anderluh, G. (2016) Crystal structure of an invertebrate cytolysin pore reveals unique properties and mechanism of assembly. Nat. Commun. 7, 11598, DOI: 10.1038/ncomms1159850Crystal structure of an invertebrate cytolysin pore reveals unique properties and mechanism of assemblyPodobnik, Marjetka; Savory, Peter; Rojko, Nejc; Kisovec, Matic; Wood, Neil; Hambley, Richard; Pugh, Jonathan; Wallace, E. Jayne; McNeill, Luke; Bruce, Mark; Liko, Idlir; Allison, Timothy M.; Mehmood, Shahid; Yilmaz, Neval; Kobayashi, Toshihide; Gilbert, Robert J. C.; Robinson, Carol V.; Jayasinghe, Lakmal; Anderluh, GregorNature Communications (2016), 7 (), 11598CODEN: NCAOBW; ISSN:2041-1723. (Nature Publishing Group)The invertebrate cytolysin lysenin is a member of the aerolysin family of pore-forming toxins that includes many representatives from pathogenic bacteria. Here we report the crystal structure of the lysenin pore and provide insights into its assembly mechanism. The lysenin pore is assembled from nine monomers via dramatic reorganization of almost half of the monomeric subunit structure leading to a β-barrel pore ∼10 nm long and 1.6-2.5 nm wide. The lysenin pore is devoid of addnl. luminal compartments as commonly found in other toxin pores. Mutagenic anal. and at. force microscopy imaging, together with these structural insights, suggest a mechanism for pore assembly for lysenin. These insights are relevant to the understanding of pore formation by other aerolysin-like pore-forming toxins, which often represent crucial virulence factors in bacteria.
- 51Tsuchikama, K. and An, Z. (2018) Antibody-drug conjugates: recent advances in conjugation and linker chemistries. Protein Cell 9, 33– 46, DOI: 10.1007/s13238-016-0323-051Antibody-drug conjugates: recent advances in conjugation and linker chemistriesTsuchikama, Kyoji; An, ZhiqiangProtein & Cell (2018), 9 (1), 33-46CODEN: PCREFB; ISSN:1674-800X. (Higher Education Press)The antibody-drug conjugate (ADC), a humanized or human monoclonal antibody conjugated with highly cytotoxic small mols. (payloads) through chem. linkers, is a novel therapeutic format and has great potential to make a paradigm shift in cancer chemotherapy. This new antibody-based mol. platform enables selective delivery of a potent cytotoxic payload to target cancer cells, resulting in improved efficacy, reduced systemic toxicity, and preferable pharmacokinetics (PK)/pharmacodynamics (PD) and biodistribution compared to traditional chemotherapy. Boosted by the successes of FDA-approved Adcetris and Kadcyla, this drug class has been rapidly growing along with about 60 ADCs currently in clin. trials. In this article, we briefly review mol. aspects of each component (the antibody, payload, and linker) of ADCs, and then mainly discuss traditional and new technologies of the conjugation and linker chemistries for successful construction of clin. effective ADCs. Current efforts in the conjugation and linker chemistries will provide greater insights into mol. design and strategies for clin. effective ADCs from medicinal chem. and pharmacol. standpoints. The development of site-specific conjugation methodologies for constructing homogeneous ADCs is an esp. promising path to improving ADC design, which will open the way for novel cancer therapeutics.
- 52Leriche, G., Chisholm, L., and Wagner, A. (2012) Cleavable linkers in chemical biology. Bioorg. Med. Chem. 20, 571– 582, DOI: 10.1016/j.bmc.2011.07.04852Cleavable linkers in chemical biologyLeriche, Geoffray; Chisholm, Louise; Wagner, AlainBioorganic & Medicinal Chemistry (2012), 20 (2), 571-582CODEN: BMECEP; ISSN:0968-0896. (Elsevier B.V.)A review. Interest in cleavable linkers is growing due to the rapid development and expansion of chem. biol. The chem. constrains imposed by the biol. conditions cause significant challenges for org. chemists. In this review we will present an overview of the cleavable linkers used in chem. biol. classified according to their cleavage conditions by enzymes, nucleophilic/basic reagents, reducing agents, photo-irradn., electrophilic/acidic reagents, organometallic and metal reagents, oxidizing reagents.
- 53Cajnko, M. M., Marušić, M., Kisovec, M., Rojko, N., Benčina, M., Caserman, S., and Anderluh, G. (2015) Listeriolysin O affects the permeability of Caco-2 monolayer in a pore-dependent and Ca2+-independent manner. PLoS One 10, e0130471 DOI: 10.1371/journal.pone.013047153Listeriolysin O affects the permeability of Caco-2 monolayer in a pore-dependent and Ca2+-independent mannerCajnko Misa, Mojca; Marusic, Maja; Kisovec, Matic; Rojko, Nejc; Bencina, Mojca; Caserman, Simon; Anderluh, GregorPLoS One (2015), 10 (6), e0130471/1-e0130471/21CODEN: POLNCL; ISSN:1932-6203. (Public Library of Science)Listeria monocytogenes is a food and soil-borne pathogen that secretes a pore-forming toxin listeriolysin O (LLO) as its major virulence factor. We tested the effects of LLO on an intestinal epithelial cell line Caco-2 and compared them to an unrelated pore-forming toxin equinatoxin II (EqtII). Apical application of both toxins causes a significant drop in transepithelial elec. resistance (TEER), with higher LLO concns. or prolonged exposure time needed to achieve the same magnitude of response than with EqtII. The drop in TEER was due to pore formation and coincided with rearrangement of claudin-1 within tight junctions and assocd. actin cytoskeleton; however, no significant increase in permeability to fluorescein or 3 kDa FITC-dextran was obsd. Influx of calcium after pore formation affected the magnitude of the drop in TEER. Both toxins exhibit similar effects on epithelium morphol. and physiol. Importantly, LLO action upon the membrane is much slower and results in compromised epithelium on a longer time scale at lower concns. than EqtII. This could favor listerial invasion in hosts resistant to E-cadherin related infection.
- 54Gerweck, L. E. and Seetharaman, K. (1996) Cellular pH gradient in tumor versus normal tissue: Potential exploitation for the treatment of cancer. Cancer Res. 56, 1194– 119854Cellular pH gradient in tumor versus normal tissue: potential exploitation for the treatment of cancerGerweck, Leo E.; Seetharaman, KalaCancer Research (1996), 56 (6), 1194-8CODEN: CNREA8; ISSN:0008-5472. (American Association for Cancer Research)Although limited data exist, electrode-measured pH values of human tumors and adjacent normal tissues, which are concurrently obtained by the same investigator in the same patient, consistently show that the electrode pH (believed to primarily represent tissue extracellular pH) is substantially and consistently lower in tumor than in normal tissue. In contrast, the 31P-magnetic resonance spectroscopy estd. that intracellular pH is essentially identical or slightly more basic in tumor compared to normal tissue. As a consequence, the cellular pH gradient is substantially reduced or reversed in tumor compared to normal tissue: in normal tissue the extracellular pH is relatively basic, and in tumor tissue the magnitude of the pH gradient is reduced or reversed. This difference provides an exploitable avenue for the treatment of cancer. The extent to which drugs exhibiting weakly acid or basic properties are ionized is strongly dependent on the pH of their milieu. Weakly acidic drugs which are relatively lipid sol. in their nonionized state may diffuse freely across the cell membrane and, upon entering a relatively basic intracellular compartment, become trapped and accumulate within a cell, leading to substantial differences in the intracellular/extracellular drug distribution between tumor and normal tissue for drugs exhibiting appropriate pKas.
- 55Cerutti, P. A. (1985) Prooxidant states and tumor promotion. Science 227, 375– 381, DOI: 10.1126/science.298143355Prooxidant states and tumor promotionCerutti, Peter A.Science (Washington, DC, United States) (1985), 227 (4685), 375-81CODEN: SCIEAS; ISSN:0036-8075.There is convincing evidence that cellular prooxidant states-i.e., increased concns. of active O and org. peroxides and radicals-can promote initiated cells to neoplastic growth. Prooxidant states can be caused by different classes of agents, including hyperbaric O, radiation, xenobiotic metabolites and Fenton-type reagents, modulators of the cytochrome P 450 electron-transport chain, peroxisome proliferators, inhibitors of the antioxidant defense, and membrane-active agents. Many of these agents are promoters or complete carcinogens. They cause chromosomal damage by indirect action, but the role of this damage in carcinogenesis remains unclear. Prooxidant states can be prevented or suppressed by the enzymes of the cellular antioxidant defense and low mol. wt. scavenger mols., and many antioxidants are antipromoters and anticarcinogens. Finally, prooxidant states may modulate the expression of a family of prooxidant genes, which are related to cell growth and differentiation, by inducing alterations in DNA structure or by epigenetic mechanisms, for example, by polyadenosine diphosphate-ribosylation of chromosomal proteins.
- 56Bergers, G., Brekken, R., McMahon, G., Vu, T. H., Itoh, T., Tamaki, K., Tanzawa, K., Thorpe, P., Itohara, S., Werb, Z., and Hanahan, D. (2000) Matrix metalloproteinase-9 triggers the angiogenic switch during carcinogenesis. Nat. Cell Biol. 2, 737– 744, DOI: 10.1038/3503637456Matrix metalloproteinase-9 triggers the angiogenic switch during carcinogenesisBergers, Gabriele; Brekken, Rolf; McMahon, Gerald; Vu, Thiennu H.; Itoh, Takeshi; Tamaki, Kazuhiko; Tanzawa, Kazuhiko; Thorpe, Philip; Itohara, Shigeyoshi; Werb, Zena; Hanahan, DouglasNature Cell Biology (2000), 2 (10), 737-744CODEN: NCBIFN; ISSN:1465-7392. (Nature Publishing Group)During carcinogenesis of pancreatic islets in transgenic mice, an angiogenic switch activates the quiescent vasculature. Paradoxically, vascular endothelial growth factor (VEGF) and its receptors are expressed constitutively. Nevertheless, a synthetic inhibitor (SU5416) of VEGF signalling impairs angiogenic switching and tumor growth. Two metalloproteinases, MMP-2/gelatinase-A and MMP-9/gelatinase-B, are upregulated in angiogenic lesions. MMP-9 can render normal islets angiogenic, releasing VEGF. MMP inhibitors reduce angiogenic switching, and tumor no. and growth, as does genetic ablation of MMP-9. Absence of MMP-2 does not impair induction of angiogenesis, but retards tumor growth, whereas lack of urokinase has no effect. Our results show that MMP-9 is a component of the angiogenic switch.
- 57Karimi, M., Ghasemi, A., Sahandi Zangabad, P., Rahighi, R., Moosavi Basri, S. M., Mirshekari, H., Amiri, M., Shafaei Pishabad, Z., Aslani, A., Bozorgomid, M., Ghosh, D., Beyzavi, A., Vaseghi, A., Aref, A. R., Haghani, L., Bahrami, S., and Hamblin, M. R. (2016) Smart micro/nanoparticles in stimulus-responsive drug/gene delivery systems. Chem. Soc. Rev. 45, 1457– 1501, DOI: 10.1039/C5CS00798D57Smart micro/nanoparticles in stimulus-responsive drug/gene delivery systemsKarimi, Mahdi; Ghasemi, Amir; Sahandi Zangabad, Parham; Rahighi, Reza; Moosavi Basri, S. Masoud; Mirshekari, H.; Amiri, M.; Shafaei Pishabad, Z.; Aslani, A.; Bozorgomid, M.; Ghosh, D.; Beyzavi, A.; Vaseghi, A.; Aref, A. R.; Haghani, L.; Bahrami, S.; Hamblin, Michael R.Chemical Society Reviews (2016), 45 (5), 1457-1501CODEN: CSRVBR; ISSN:0306-0012. (Royal Society of Chemistry)New achievements in the realm of nanoscience and innovative techniques of nanomedicine have moved micro/nanoparticles (MNPs) to the point of becoming actually useful for practical applications in the near future. Various differences between the extracellular and intracellular environments of cancerous and normal cells and the particular characteristics of tumors such as physicochem. properties, neovasculature, elasticity, surface elec. charge, and pH have motivated the design and fabrication of inventive "smart" MNPs for stimulus-responsive controlled drug release. These novel MNPs can be tailored to be responsive to pH variations, redox potential, enzymic activation, thermal gradients, magnetic fields, light, and ultrasound (US), or can even be responsive to dual or multi-combinations of different stimuli. This unparalleled capability has increased their importance as site-specific controlled drug delivery systems (DDSs) and has encouraged their rapid development in recent years. An in-depth understanding of the underlying mechanisms of these DDS approaches is expected to further contribute to this groundbreaking field of nanomedicine. Smart nanocarriers in the form of MNPs that can be triggered by internal or external stimulus are summarized and discussed in the present review, including pH-sensitive peptides and polymers, redox-responsive micelles and nanogels, thermo- or magnetic-responsive nanoparticles (NPs), mech.- or elec.-responsive MNPs, light or ultrasound-sensitive particles, and multi-responsive MNPs including dual stimuli-sensitive nanosheets of graphene. This review highlights the recent advances of smart MNPs categorized according to their activation stimulus (phys., chem., or biol.) and looks forward to future pharmaceutical applications.
- 58Zahnd, C., Amstutz, P., and Plückthun, A. (2007) Ribosome display: selecting and evolving proteins in vitro that specifically bind to a target. Nat. Methods 4, 269– 279, DOI: 10.1038/nmeth100358Ribosome display: selecting and evolving proteins in vitro that specifically bind to a targetZahnd, Christian; Amstutz, Patrick; Plueckthun, AndreasNature Methods (2007), 4 (3), 269-279CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)Ribosome display is an in vitro selection and evolution technol. for proteins and peptides from large libraries. As it is performed entirely in vitro, there are two main advantages over other selection technologies. First, the diversity of the library is not limited by the transformation efficiency of bacterial cells, but only by the no. of ribosomes and different mRNA mols. present in the test tube. Second, random mutations can be introduced easily after each selection round, as no library must be transformed after any diversification step. This allows facile directed evolution of binding proteins over several generations (Box 1). A prerequisite for the selection of proteins from libraries is the coupling of genotype (RNA, DNA) and phenotype (protein). In ribosome display, this link is accomplished during in vitro translation by stabilizing the complex consisting of the ribosome, the mRNA and the nascent, correctly folded polypeptide (Fig. 1). The DNA library coding for a particular library of binding proteins is genetically fused to a spacer sequence lacking a stop codon. This spacer sequence, when translated, is still attached to the peptidyl tRNA and occupies the ribosomal tunnel, and thus allows the protein of interest to protrude out of the ribosome and fold. The ribosomal complexes are allowed to bind to surface-immobilized target. Whereas non-bound complexes are washed away, mRNA of the complexes displaying a binding polypeptide can be recovered, and thus, the genetic information of the binding polypeptides is available for anal. Here we describe a step-by-step procedure to perform ribosome display selection using an Escherichia coli S30 ext. for in vitro translation, based on the work originally described and further refined in our lab. A protocol that makes use of eukaryotic in vitro translation systems for ribosome display is also included in this issue.
- 59Zahnd, C., Wyler, E., Schwenk, J. M., Steiner, D., Lawrence, M. C., McKern, N. M., Pecorari, F., Ward, C. W., Joos, T. O., and Plückthun, A. (2007) A designed ankyrin repeat protein evolved to picomolar affinity to Her2. J. Mol. Biol. 369, 1015– 28, DOI: 10.1016/j.jmb.2007.03.02859A Designed Ankyrin Repeat Protein Evolved to Picomolar Affinity to Her2Zahnd, Christian; Wyler, Emanuel; Schwenk, Jochen M.; Steiner, Daniel; Lawrence, Michael C.; McKern, Neil M.; Pecorari, Frederic; Ward, Colin W.; Joos, Thomas O.; Plueckthun, AndreasJournal of Molecular Biology (2007), 369 (4), 1015-1028CODEN: JMOBAK; ISSN:0022-2836. (Elsevier Ltd.)Designed ankyrin repeat proteins (DARPins) are a novel class of binding mols., which can be selected to recognize specifically a wide variety of target proteins. DARPins were previously selected against human epidermal growth factor receptor 2 (Her2) with low nanomolar affinities. We describe here their affinity maturation by error-prone PCR and ribosome display yielding clones with zero to seven (av. 2.5) amino acid substitutions in framework positions. The DARPin with highest affinity (90 pM) carried four mutations at framework positions, leading to a 3000-fold affinity increase compared to the consensus framework variant, mainly coming from a 500-fold increase of the on-rate. This DARPin was found to be highly sensitive in detecting Her2 in human carcinoma exts. We have detd. the crystal structure of this DARPin at 1.7 Å, and found that a His to Tyr mutation at the framework position 52 alters the inter-repeat H-bonding pattern and causes a significant conformational change in the relative disposition of the repeat subdomains. These changes are thought to be the reason for the enhanced on-rate of the mutated DARPin. The DARPin not bearing the residue 52 mutation has an unusually slow on-rate, suggesting that binding occurred via conformational selection of a relatively rare state, which was stabilized by this His52Tyr mutation, increasing the on-rate again to typical values. An anal. of the structural location of the framework mutations suggests that randomization of some framework residues either by error-prone PCR or by design in a future library could increase affinities and the target binding spectrum.
- 60Liu, S., Netzel-Arnett, S., Birkedal-Hansen, H., and Leppla, S. H. (2000) Tumor cell-selective cytotoxicity of matrix metalloproteinase-activated anthrax toxin. Cancer Res. 60, 6061– 606760Tumor cell-selective cytotoxicity of matrix metalloproteinase-activated anthrax toxinLiu, Shihui; Netzel-Arnett, Sarah; Birkedal-Hansen, Henning; Leppla, Stephen H.Cancer Research (2000), 60 (21), 6061-6067CODEN: CNREA8; ISSN:0008-5472. (American Association for Cancer Research)Matrix metalloproteinases (MMPs) are overexpressed in a variety of tumor tissues and cell lines, and their expression is highly correlated to tumor invasion and metastasis. To exploit these characteristics in the design of tumor cell-selective cytotoxins, we constructed two mutated anthrax toxin protective antigen (PA) proteins in which the furin protease cleavage site is replaced by sequences selectively cleaved by MMPs. These MMP-targeted PA proteins were activated rapidly and selectively on the surface of MMP-overexpressing tumor cells. The activated PA proteins caused internalization of a recombinant cytotoxin, FP59, consisting of anthrax toxin lethal factor residues 1-254 fused to the ADP-ribosylation domain of Pseudomonas exotoxin A. The toxicity of the mutated PA proteins for MMP-overexpressing cells was blocked by hydroxamate inhibitors of MMPs, including BB94, and by a tissue inhibitor of matrix metalloproteinases (TIMP-2). The mutated PA proteins killed MMP-overexpressing tumor cells while sparing nontumorigenic normal cells when these were grown together in a coculture model, indicating that PA activation occurred on the tumor cell surface and not in the supernatant. This method of achieving cell-type specificity is conceptually distinct from, and potentially synergistic with, the more common strategy of retargeting a protein toxin by fusion to a growth factor, cytokine, or antibody.
- 61Franke, D., Petoukhov, M. V., Konarev, P. V., Panjkovich, A., Tuukkanen, A., Mertens, H. D. T., Kikhney, A. G., Hajizadeh, N. R., Franklin, J. M., Jeffries, C. M., and Svergun, D. I. (2017) ATSAS 2.8: a comprehensive data analysis suite for small-angle scattering from macromolecular solutions. J. Appl. Crystallogr. 50, 1212– 1225, DOI: 10.1107/S160057671700778661ATSAS 2.8: a comprehensive data analysis suite for small-angle scattering from macromolecular solutionsFranke, D.; Petoukhov, M. V.; Konarev, P. V.; Panjkovich, A.; Tuukkanen, A.; Mertens, H. D. T.; Kikhney, A. G.; Hajizadeh, N. R.; Franklin, J. M.; Jeffries, C. M.; Svergun, D. I.Journal of Applied Crystallography (2017), 50 (4), 1212-1225CODEN: JACGAR; ISSN:1600-5767. (International Union of Crystallography)A review. ATSAS is a comprehensive software suite for the anal. of small-angle scattering data from dil. solns. of biol. macromols. or nanoparticles. It contains applications for primary data processing and assessment, ab initio bead modeling, and model validation, as well as methods for the anal. of flexibility and mixts. In addn., approaches are supported that utilize information from X-ray crystallog., NMR spectroscopy or atomistic homol. modeling to construct hybrid models based on the scattering data. This article summarizes the progress made during the 2.5-2.8 ATSAS release series and highlights the latest developments. These include AMBIMETER, an assessment of the reconstruction ambiguity of exptl. data; DATCLASS, a multiclass shape classification based on exptl. data; SASRES, for estg. the resoln. of ab initio model reconstructions; CHROMIXS, a convenient interface to analyze in-line size exclusion chromatog. data; SHANUM, to evaluate the useful angular range in measured data; SREFLEX, to refine available high-resoln. models using normal mode anal.; SUPALM for a rapid superposition of low- and high-resoln. models; and SASPy, the ATSAS plugin for interactive modeling in PyMOL. All these features and other improvements are included in the ATSAS release 2.8, freely available for academic users from https://www.embl-hamburg.de/biosaxs/software.html.</a></a></a></a></a></a></a></a></a>.
- 62Franke, D. and Svergun, D. I. (2009) DAMMIF, a program for rapid ab-initio shape determination in small-angle scattering. J. Appl. Crystallogr. 42, 342– 346, DOI: 10.1107/S002188980900033862DAMMIF, a program for rapid ab-initio shape determination in small-angle scatteringFranke, Daniel; Svergun, Dmitri I.Journal of Applied Crystallography (2009), 42 (2), 342-346CODEN: JACGAR; ISSN:0021-8898. (International Union of Crystallography)DAMMIF, a revised implementation of the ab-initio shape-detn. program DAMMIN for small-angle scattering data, is presented. The program was fully rewritten, and its algorithm was optimized for speed of execution and modified to avoid limitations due to the finite search vol. Symmetry and anisometry constraints can be imposed on the particle shape, similar to DAMMIN. In equivalent conditions, DAMMIF is 25-40 times faster than DAMMIN on a single CPU. The possibility to utilize multiple CPUs is added to DAMMIF. The application is available in binary form for major platforms.
- 63Volkov, V. V. and Svergun, D. I. (2003) Uniqueness of ab initio shape determination in small-angle scattering. J. Appl. Crystallogr. 36, 860– 864, DOI: 10.1107/S002188980300026863Uniqueness of ab initio shape determination in small-angle scatteringVolkov, Vladimir V.; Svergun, Dmitri I.Journal of Applied Crystallography (2003), 36 (3, Pt. 1), 860-864CODEN: JACGAR; ISSN:0021-8898. (Blackwell Munksgaard)Scattering patterns from geometrical bodies with different shapes and anisometry (solid and hollow spheres, cylinders, prisms) are computed and the shapes are reconstructed ab initio using envelope function and bead modeling methods. A procedure is described to analyze multiple solns. provided by bead modeling methods and to est. stability and reliability of the shape reconstruction. It is demonstrated that flat shapes are more difficult to restore than elongated ones and types of shapes are indicated, which require addnl. information for reliable shape reconstruction from the scattering data.
- 64The PyMOL Molecular Graphics System, Version 2.0, Schrödinger, LLC.There is no corresponding record for this reference.
- 65Webb, B., and Sali, A. (2017) Protein Structure Modeling with MODELLER, in Functional Genomics: Methods and Protocols, Methods in Molecular Biology, (Kaufmann, M., Klinger, C., and Savelsbergh, A., Eds.), pp 39– 54, Humana Press, New York, NY.There is no corresponding record for this reference.
- 66Waterhouse, A., Bertoni, M., Bienert, S., Studer, G., Tauriello, G., Gumienny, R., Heer, F. T., De Beer, T. A. P., Rempfer, C., Bordoli, L., Lepore, R., and Schwede, T. (2018) SWISS-MODEL: Homology modelling of protein structures and complexes. Nucleic Acids Res. 46, W296– W303, DOI: 10.1093/nar/gky42766SWISS-MODEL: homology modelling of protein structures and complexesWaterhouse, Andrew; Bertoni, Martino; Bienert, Stefan; Studer, Gabriel; Tauriello, Gerardo; Gumienny, Rafal; Heer, Florian T.; de Beer, Tjaart A. P.; Rempfer, Christine; Bordoli, Lorenza; Lepore, Rosalba; Schwede, TorstenNucleic Acids Research (2018), 46 (W1), W296-W303CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)Homol. modeling has matured into an important technique in structural biol., significantly contributing to narrowing the gap between known protein sequences and exptl. detd. structures. Fully automated workflows and servers simplify and streamline the homol. modeling process, also allowing users without a specific computational expertise to generate reliable protein models and have easy access to modeling results, their visualization and interpretation. Here, we present an update to the SWISS-MODEL server, which pioneered the field of automated modeling 25 years ago and been continuously further developed. Recently, its functionality has been extended to the modeling of homo- and heteromeric complexes. Starting from the amino acid sequences of the interacting proteins, both the stoichiometry and the overall structure of the complex are inferred by homol. modeling. Other major improvements include the implementation of a new modeling engine, ProMod3 and the introduction a new local model quality estn. method, QMEANDisCo.
- 67Schweizer, A., Roschitzki-Voser, H., Amstutz, P., Briand, C., Gulotti-Georgieva, M., Prenosil, E., Binz, H. K., Capitani, G., Baici, A., Plückthun, A., and Grütter, M. G. (2007) Inhibition of caspase-2 by a designed ankyrin repeat protein: specificity, structure, and inhibition mechanism. Structure 15, 625– 36, DOI: 10.1016/j.str.2007.03.01467Inhibition of Caspase-2 by a Designed Ankyrin Repeat Protein: Specificity, Structure, and Inhibition MechanismSchweizer, Andreas; Roschitzki-Voser, Heidi; Amstutz, Patrick; Briand, Christophe; Gulotti-Georgieva, Maya; Prenosil, Eva; Binz, H. Kaspar; Capitani, Guido; Baici, Antonio; Plueckthun, Andreas; Gruetter, Markus G.Structure (Cambridge, MA, United States) (2007), 15 (5), 625-636CODEN: STRUE6; ISSN:0969-2126. (Cell Press)Summary: Specific and potent caspase inhibitors are indispensable for the dissection of the intricate pathways leading to apoptosis. We selected a designed ankyrin repeat protein (DARPin) from a combinatorial library that inhibits caspase-2 in vitro with a subnanomolar inhibition const. and, in contrast to the peptidic caspase inhibitors, with very high specificity for this particular caspase. The crystal structure of this inhibitor (AR_F8) in complex with caspase-2 reveals the mol. basis for the specificity and, together with kinetic analyses, the allosteric mechanism of inhibition. The structure also shows a conformation of the active site that can be exploited for the design of inhibitory compds. AR_F8 is a specific inhibitor of an initiator caspase and has the potential to help identify the function of caspase-2 in the complex biol. apoptotic signaling network.
- 68Petoukhov, M. V. and Svergun, D. I. (2005) Global rigid body modeling of macromolecular complexes against small-angle scattering data. Biophys. J. 89, 1237– 1250, DOI: 10.1529/biophysj.105.06415468Global rigid body modeling of macromolecular complexes against small-angle scattering dataPetoukhov, Maxim V.; Svergun, Dmitri I.Biophysical Journal (2005), 89 (2), 1237-1250CODEN: BIOJAU; ISSN:0006-3495. (Biophysical Society)New methods to automatically build models of macromol. complexes from high-resoln. structures or homol. models of their subunits or domains against x-ray or neutron small-angle scattering data are presented. Depending on the complexity of the object, different approaches are employed for the global search of the optimum configuration of subunits fitting the exptl. data. An exhaustive grid search is used for hetero- and homodimeric particles and for sym. oligomers formed by identical subunits. For the assemblies or multidomain proteins contg. more then one subunit/domain per asym. unit, heuristic algorithms based on simulated annealing are used. Fast computational algorithms based on spherical harmonics representation of scattering amplitudes are employed. The methods allow one to construct interconnected models without steric clashes, to account for the particle symmetry and to incorporate information from other methods, on distances between specific residues or nucleotides. For multidomain proteins, addn. of missing linkers between the domains is possible. Simultaneous fitting of multiple scattering patterns from subcomplexes or deletion mutants is incorporated. The efficiency of the methods is illustrated by their application to complexes of different types in several simulated and practical examples. Limitations and possible ambiguity of rigid body modeling are discussed and simplified docking criteria are provided to rank multiple models. The methods described are implemented in publicly available computer programs running on major hardware platforms.
- 69Panjkovich, A. and Svergun, D. I. (2016) SASpy: A PyMOL plugin for manipulation and refinement of hybrid models against small angle X-ray scattering data. Bioinformatics 32, 2062– 2064, DOI: 10.1093/bioinformatics/btw07169SASpy: a PyMOL plugin for manipulation and refinement of hybrid models against small angle X-ray scattering dataPanjkovich, Alejandro; Svergun, Dmitri I.Bioinformatics (2016), 32 (13), 2062-2064CODEN: BOINFP; ISSN:1367-4803. (Oxford University Press)Complex formation and conformational transitions of biol. macromols. in soln. can be effectively studied using the information about overall shape and size provided by small angle X-ray scattering (SAXS). Hybrid modeling is often applied to integrate high-resoln. models into SAXS data anal. To facilitate this task, we present SASpy, a PyMOL plugin that provides an easy-to-use graphical interface for SAXS-based hybrid modeling. Through a few mouse clicks in SASpy, low-resoln. models can be superimposed to high-resoln. structures, theor. scattering profiles and fits can be calcd. and displayed on-the-fly. Mouse-based manual rearrangements of complexes are conveniently applied to rapidly check and interactively refine tentative models. Interfaces to automated rigid-body and flexible refinement of macromol. models against the exptl. SAXS data are provided.
- 70Hodnik, V., and Anderluh, G. (2013) Surface Plasmon Resonance for Measuring Interactions of Proteins with Lipid Membranes, in Lipid-Protein Interactions: Methods and Protocols, Methods in Molecular Biology, (Kleinschmidt, J. H., Ed.), pp 23– 36, Humana Press, Totowa, NJ.There is no corresponding record for this reference.
- 71Anderluh, G., Beseničar, M., Kladnik, A., Lakey, J. H., and Maček, P. (2005) Properties of nonfused liposomes immobilized on an L1 Biacore chip and their permeabilization by a eukaryotic pore-forming toxin. Anal. Biochem. 344, 43– 52, DOI: 10.1016/j.ab.2005.06.01371Properties of nonfused liposomes immobilized on an L1 Biacore chip and their permeabilization by a eukaryotic pore-forming toxinAnderluh, Gregor; Besenicar, Mojca; Kladnik, Ales; Lakey, Jeremy H.; Macek, PeterAnalytical Biochemistry (2005), 344 (1), 43-52CODEN: ANBCA2; ISSN:0003-2697. (Elsevier)The L1 chip is used intensively for protein-membrane interaction studies in Biacore surface plasmon resonance systems. The exact form of captured lipid membranes on the chip is, however, not precisely known. Evidence exists that the vesicles both remain intact after the binding to the chip and fuse to form a large single-bilayer membrane. In this study, the authors were able to bind up to approx. 11,500 resonance units of zwitterionic liposomes (100 nm in diam.) at a low flow rate. The authors show by fluorescence microscopy that the entire surface of the flow cell is covered homogeneously by liposomes. Neg. charged vesicles (i.e., those composed of phosphatidylcholine/phosphatidylglycerol [1:1]) always deposited less densely, but the authors were able to increase the d. slightly with the use of calcium chloride that promotes fusion of the vesicles. Finally, the authors used zwitterionic liposomes loaded with fluorescent probe calcein to show that they remain intact after the capture on the L1 chip. The fluorescence was lost only after the authors used equinatoxin, a well-studied pore-forming toxin, to perform on-chip permeabilization of vesicles. The characteristics of permeabilization process for chip-immobilized liposomes are similar to those of liposomes free in soln. All results collectively suggest that liposomes do not fuse to form a single bilayer on the surface of the chip.
- 72Schneider, C. A., Rasband, W. S., and Eliceiri, K. W. (2012) NIH Image to ImageJ: 25 years of image analysis. Nat. Methods 9, 671– 675, DOI: 10.1038/nmeth.208972NIH Image to ImageJ: 25 years of image analysisSchneider, Caroline A.; Rasband, Wayne S.; Eliceiri, Kevin W.Nature Methods (2012), 9 (7_part1), 671-675CODEN: NMAEA3; ISSN:1548-7091. (Nature Publishing Group)For the past 25 years NIH Image and ImageJ software have been pioneers as open tools for the anal. of scientific images. We discuss the origins, challenges and solns. of these two programs, and how their history can serve to advise and inform other software projects.