pH-Responsive Peptide Nanoparticles Deliver Macromolecules to Cells via Endosomal Membrane NanoporationClick to copy article linkArticle link copied!
- Eric WuEric WuDepartment of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana 70112, United StatesMore by Eric Wu
- Ains EllisAins EllisDepartment of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana 70112, United StatesMore by Ains Ellis
- Keynon BellKeynon BellChemistry-Biology Interface Program, Johns Hopkins University, Baltimore, Maryland 21218, United StatesInstitute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, United StatesMore by Keynon Bell
- Daniel L. MossDaniel L. MossDepartment of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana 70112, United StatesMore by Daniel L. Moss
- Samuel J. LandrySamuel J. LandryDepartment of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana 70112, United StatesMore by Samuel J. Landry
- Kalina HristovaKalina HristovaInstitute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland 21218, United StatesDepartment of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland 21218, United StatesMore by Kalina Hristova
- William C. Wimley*William C. Wimley*Email: [email protected]Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, Louisiana 70112, United StatesMore by William C. Wimley
Abstract
The synthetically evolved pHD family of peptides is known to self-assemble into macromolecule-sized nanopores of 2–10 nm diameter in synthetic lipid bilayers, but only when the pH is below ∼6. Here, we show that a representative family member, pHD108, has the same pH-responsive nanopore-forming activity in the endosomal membranes of living human cells, which is triggered by endosomal acidification. This enables the cytosolic delivery of endocytosed proteins and other macromolecules. Acylation of either peptide terminus significantly decreases the concentration of peptide required for macromolecule delivery to the cell cytosol while not causing any measurable cytotoxicity. Longer acyl chains are more effective. The N-terminal palmitoylated C16-pHD108 is the most potent of all of the acyl-pHD108 variants and readily delivers a cytotoxic enzyme, fluorescent proteins, and a dye-labeled dextran to the cell cytosol. C16-pHD108 forms stable monodisperse micellar nanoparticles in a buffer at pH 7 with an average diameter of around 120 nm. These nanoparticles are not cytolytic or cytotoxic because the acylated pHD peptide does not partition from the nanoparticles into cell membranes at pH 7. At pH 5, the nanoparticles are unstable, driving acylated pHD108 to bind strongly to membranes. We hypothesize that passive endocytosis of macromolecular cargo and stable peptide nanoparticles, followed by endosomal acidification-dependent destabilization of the nanoparticles, triggers the nanopore-forming activity of acylated pHD peptides in the endosomal membrane, enabling internalized macromolecules to be delivered to the cytosol.
This publication is licensed under
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
Introduction
Results
pHD Peptides Self-Assemble into Nanopores
Figure 1
Figure 1. Synthetically evolved nanopore-forming peptide pHD108. (A) The amino acid sequence of pHD108 is shown. In this work, we use both l- and d-amino acid versions of pHD108. (B) Helical wheel diagram for pHD108, showing nonpolar residues in gray, basic residues in blue, acidic residues in red, and polar residues in orange. The P14 residue, which is essential for activity, is shown in green. (C) Nanoporation activity of pHD108 in synthetic lipid vesicles made from 1-palmitoyl-2-oleoly phosphatidylcholine (POPC), versus pH. (28,30) Nanoporation is measured by the release of a 40 kDa dextran from liposomes. (D) Concentration dependence of nanoporation activity (28,30) in synthetic liposomes.
Development of a High-Throughput Protein Delivery Assay
Figure 2
Figure 2. Development of an assay to measure the delivery of the PE-III protein to live cells. (A) The PE-III protein cargo is ∼25 kDa and has major and minor axes of 4.3 and 2.7 nm, respectively. (B) Fate of HeLa cells incubated with peptides, PE-III protein, or a combination. MelP5 was used at 25 μM, pHD108 was used at 200 μM, and PE-III was used at 40 nM. Cell viability was measured 48 h after incubation to allow for apoptosis to occur. Notation: l-P and d-P are l- and d-amino acid versions of pHD108. (C) Delivery of PE-III mutants is measured by Alamar Blue detection of cell viability. The R494A mutant, which is biochemically active but less stable, was tested with and without pHD108. The Δ485–492 mutant, which is biochemically inactive, was also tested with and without pHD108. (D) The PE-III delivery assay was tested on HeLa cells and RAW macrophages using 40 mM PE-III and a range of pHD108 concentrations.
Acylation Drives a Dramatic Increase in Macromolecule Delivery Activity
Figure 3
Figure 3. Variants of pHD108 were tested in this work. Two groups of acylated peptides were synthesized. (Left) pHD108 with a C-terminal cysteine had alkyl chains attached by a disulfide cross-link. This group includes a peptide with the sulfhydryl group alkylated with acetamide and a disulfide cross-linked peptide dimer. (Right) pHD108 with acyl chains attached to the amino-terminal amine by an amide bond. This group includes unmodified l- and d-pHD108, as well as l- and d-pHD108 with C16 chains. Notation: d-P, l-P: d- and l-amino acid pHD108. CN: Linear, saturated alkyl chain with N carbons. CN- at the start of the name indicates an N-terminal acylated peptide. −CN at the end of the name indicates a C-terminal modified peptide. These images are not drawn to scale─the acyl chain sizes are exaggerated for increased visibility.
Figure 4
Figure 4. PE-III protein delivery by acylated pHD108 variants. (A, B) Delivery of PE-III by C-terminal variants (A) and N-terminal variants (B). HeLa cells were incubated with 40 nM PE-III and serially diluted pHD108 variants. Cytosolic PE-III causes apoptosis. To assay for delivery, cell viability was measured 48 h after treatment using Alamar Blue. The signal from cells with no treatment was defined as 100% viability. Media without cells was defined as 0% viability. Peptide variant notation is defined in Figure 3. ONEG is an unrelated peptide (39) used as a control. (C, D) Concentration midpoint (EC50) values for the C-terminal variants (C) and N-terminal variants (D) determined from the data in panels (A, B). (E) Direct cytotoxicity of the pHD peptide variants in the absence of the PE-III cargo measured by Alamar Blue, 48 h after treatment with peptides as above.
C16-d-pHD108 Delivers Other Macromolecules to the Cytosol
Figure 5
Figure 5. Cytosolic delivery of dye-labeled dextran to cells by C16-d-pHD108. HeLa cells were incubated at 37 °C overnight with 25 μM AF488-dextran 10 kDa with and without 25 μM C16-d-pHD108. Just before imaging, external AF488-dextran (green) was washed off and replaced with AF647-dextran (lavender) to mark the external spaces and cell boundaries. Cells were also treated with Hoechst 33342 (blue) to stain the nuclei. (A) Cells were incubated overnight with AF488-dextran and C16-d-pHD108. (B) The same image as in Panel A, except only the dextran fluorescence, is shown. (C) Cells were incubated overnight with AF488-dextran in the absence of C16-d-pHD108. (D) The same image as in Panel C, except only the dextran fluorescence is shown. (E) Example of normalized intensity measurements across the extracellular spaces, cytosol, and nucleus of the cell indicated by the arrow in panel B. (F) Example of normalized intensity measurements across the extracellular spaces, cytosol, and nucleus of the cells indicated by the arrow in panel D. The intensity of a 10 μM standard solution of AF488-dextran, 40% of the external concentration, is shown as a dashed line in panels (E, F).
Figure 6
Figure 6. Measurement of cytosolic delivery of AF88-labeled dextran by variants of pHD108. (A) Confocal microscopy images of HeLa cells incubated at 37 °C overnight with 0.1 μM AF488-dextran 10 kDa, plus the pHD108 variants indicated. In all cases, we used confocal images to measure the diffuse fluorescence in the nuclei and cytosol and compare it to that of a standard curve consisting of different concentrations of a reference solution containing AF488-dextran. After incubation, all cells contain bright endosomal puncta that have unreleased dextran, including masses gathered in perinuclear regions. (B) Nuclear and cytosolic fluorescence, identified by diffuse uniform intensity exclusive of bright puncta, was measured in ∼50 individual cells, shown as individual points. In the absence of peptides, cytosolic and nuclear fluorescence is negligible. By one-factor ANOVA, all peptide treatments give statistically significant increases in cytosolic fluorescence, with delivery increasing with the peptide concentration. (C) Cytosolic delivery of dextran at 4 and 37 °C, with and without C16-d-pHD108. (D) Comparison of cytosolic delivery of AF488-dextran by C16-l-pHD108 and C16-d-pHD108, measured as described above for panel B.
Fluorescent Protein Delivery
Figure 7
Figure 7. Cytosolic delivery of fluorescent proteins to two different cell lines by C16-d-pHD108. (A–E) HeLa cells were incubated at 37 °C overnight with 10 μM green fluorescent protein (GFP) with and without 25 μM C16-d-pHD108. (F–J): Chinese Hamster Ovary (CHO) cells were incubated overnight with 3.5 μM yellow fluorescent protein (YFP) with or without C16-d-pHD108. (A, B) GFP delivery to HeLa cells was confirmed with C16-d-pHD108. Just before imaging, the external GFP was washed off and replaced with TAMRA-dextran (red) to mark the external spaces and cell boundaries. The nuclei were stained with Hoechst 33342 (blue). Panel A shows all colors and panel B shows only GFP. (C, D) GFP delivery to HeLa cells in experiments identical to panels (A and B), except that C16-d-pHD108 was absent. Panel C shows all colors, and panel D shows only GFP. (E) GFP intensities from the nuclei of the cells in panels (A–D). Dotted lines are the intensities of standard reference solutions of 0, 2, and 4 μM GFP measured under identical conditions. (F, G) YFP delivery to CHO cells was carried out with 25 μM C16-d-pHD108. Just before imaging, the external YFP was washed off and replaced with fluorescent protein mTurquoise (mTurq) (blue) to mark the external spaces and cell boundaries. Panel F shows both colors and panel G shows only YFP. (H, I) YFP delivery to CHO cells in experiments was identical to that of panels (G and F), except that C16-d-pHD108 was absent. Panel H shows both colors and panel I shows only YFP. (J) YFP intensities from the nuclei and cytosol of the cells in panels (F–I). The dotted lines are the intensities of the standard reference solution of media only and 3.5 μM YFP measured under identical conditions.
Acylation of pHD108 Drives Nanoparticle Formation
Figure 8
Figure 8. Solution properties and cell binding of pHD108 variants. (A) Circular dichroism spectra of C16-d-pHD108 at pH 7, and of unmodified l-pHD108 at pH 5 with and without liposomes. Spectra were collected at 25 μM peptide in phosphate-buffered saline. (B) Tryptophan fluorescence emission spectra of the same three solutions are characterized in panel A. The dotted line indicates the emission maximum observed for pHD108, which is monomeric and its tryptophan residue is exposed to bulk water. (C) Particle size distribution in individually prepared 25 μM solutions of C16-d-pHD108 measured by quasielastic light scattering on a Malvern Zetasizer nano DLS instrument. Buffers contained phosphate plus 138 mM, 69 mM, or 0 mM added NaCl. (D) Light scattering intensity, in counts per second, is shown as a function of peptide concentration. (E) Solubility of pHD108 and C16-d-pHD108. Peptides were suspended by gentle vortexing for 1 min, followed by bath sonication for 5 min. After at least 1 h of incubation, solutions were sampled by reverse phase HPLC with and without centrifugation at 1400g and subsequent centrifugation at 11,000g. The peptide remaining in the solution was normalized to an uncentrifuged sample. (F) Cell binding of the pHD108 variants. HeLa cells at 106 cells/ml were suspended in phosphate-buffered saline at pH 7 or pH 5, followed by the addition of peptide dissolved in water. After 1 h of incubation, the cells were pelleted by centrifugation and the peptide remaining in the solution was measured by HPLC. (G) Confocal microscopy images of HeLa cells incubated with 5 μM unlabeled C16-d-D-pHD108 mixed with 1 μM C16-d-pHD108 labeled with TAMRA. All images were collected with the same instrument settings and the same peptide concentrations. In the left panel, cells were incubated with peptide in media at pH 7 for 10 min. The other panels show one field of cells under the same conditions after the solution pH was reduced to pH 5. Cells are also stained with the DNA-binding dye Hoechst 33342.
Discussion
Study Limitations
Conclusions
Materials and Methods
Peptides
pHD108 Variants
TAMRA-Labeled C16-d-pHD108
PE-III Delivery Assay
Dextran Delivery
Protein Delivery
Cell Binding
Dynamic Light Scattering
Measurement of Peptide Solubility
Circular Dichroism Spectroscopy
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsnano.4c07525.
Confocal microscopy images of uptake and delivery; quantitation of protein delivery; quantitation of antibody fragment delivery; model calculation for uptake and delivery (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgments
The authors thank Alexander Mrozek for GFP purification. This work was funded by NIH R01GM151326, the National Science Foundation NSF DMR 1709892 (K.H.) and NSF DMR 1710053 (W.C.W.), and the Ladies Leukemia League of New Orleans. K.B. is supported by NIH T32 GM080189.
References
This article references 58 other publications.
- 1Hong, C. W.; Zeng, Q. Tapping the treasure of intracellular oncotargets with immunotherapy. FEBS Lett. 2014, 588 (2), 350– 355, DOI: 10.1016/j.febslet.2013.10.025Google ScholarThere is no corresponding record for this reference.
- 2Cheever, M. A.; Allison, J. P.; Ferris, A. S.; Finn, O. J.; Hastings, B. M.; Hecht, T. T.; Mellman, I.; Prindiville, S. A.; Viner, J. L.; Weiner, L. M.; Matrisian, L. M. The prioritization of cancer antigens: a national cancer institute pilot project for the acceleration of translational research. Clin. Cancer Res. 2009, 15 (17), 5323– 5337, DOI: 10.1158/1078-0432.CCR-09-0737Google ScholarThere is no corresponding record for this reference.
- 3Lazo, J. S.; Sharlow, E. R. Drugging Undruggable Molecular Cancer Targets. Annu. Rev. Pharmacol. Toxicol. 2016, 56, 23– 40, DOI: 10.1146/annurev-pharmtox-010715-103440Google Scholar3https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhslGlsr7O&md5=5ab0518d4155a00e2a898a6f68d551d8Drugging Undruggable Molecular Cancer TargetsLazo, John S.; Sharlow, Elizabeth R.Annual Review of Pharmacology and Toxicology (2016), 56 (), 23-40CODEN: ARPTDI; ISSN:0362-1642. (Annual Reviews)Cancer, more than any other human disease, now has a surfeit of potential mol. targets poised for therapeutic exploitation. Currently, a no. of attractive and validated cancer targets remain outside of the reach of pharmacol. regulation. Some have been described as undruggable, at least by traditional strategies. In this article, we outline the basis for the undruggable moniker, propose a reclassification of these targets as undrugged, and highlight three general classes of this imposing group as exemplars with some attendant strategies currently being explored to reclassify them. Expanding the spectrum of disease-relevant targets to pharmacol. manipulation is central to reducing cancer morbidity and mortality.
- 4Slastnikova, T. A.; Ulasov, A. V.; Rosenkranz, A. A.; Sobolev, A. S. Targeted Intracellular Delivery of Antibodies: The State of the Art. Front. Pharmacol. 2018, 9, 1208, DOI: 10.3389/fphar.2018.01208Google Scholar4https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtVGmtbvL&md5=fe66302eb7d6dd1e77f41c46dcffc635Targeted intracellular delivery of antibodies: the state of the artSlastnikova, Tatiana A.; Ulasov, A. V.; Rosenkranz, A. A.; Sobolev, A. S.Frontiers in Pharmacology (2018), 9 (), 1208CODEN: FPRHAU; ISSN:1663-9812. (Frontiers Media S.A.)A review. A dominant area of antibody research is the extension of the use of this mighty exptl. and therapeutic tool for the specific detection of mols. for diagnostics, visualization, and activity blocking. Despite the ability to raise antibodies against different proteins, numerous applications of antibodies in basic research fields, clin. practice, and biotechnol. are restricted to permeabilized cells or extracellular antigens, such as membrane or secreted proteins. With the exception of small groups of autoantibodies, natural antibodies to intracellular targets cannot be used within living cells. This excludes the scope of a major class of intracellular targets, including some infamous cancer-assocd. mols. Some of these targets are still not druggable via small mols. because of large flat contact areas and the absence of deep hydrophobic pockets in which small mols. can insert and perturb their activity. Thus, the development of technologies for the targeted intracellular delivery of antibodies, their fragments, or antibody-like mols. is extremely important. Various strategies for intracellular targeting of antibodies via protein-transduction domains or their mimics, liposomes, polymer vesicles, and viral envelopes, are reviewed in this article. The pitfalls, challenges, and perspectives of these technologies are discussed.
- 5Chonghaile, T. N.; Letai, A. Mimicking the BH3 domain to kill cancer cells. Oncogene 2008, 27 (Suppl 1), S149– S157, DOI: 10.1038/onc.2009.52Google ScholarThere is no corresponding record for this reference.
- 6Goldsmith, K. C.; Liu, X.; Dam, V.; Morgan, B. T.; Shabbout, M.; Cnaan, A.; Letai, A.; Korsmeyer, S. J.; Hogarty, M. D. BH3 peptidomimetics potently activate apoptosis and demonstrate single agent efficacy in neuroblastoma. Oncogene 2006, 25 (33), 4525– 4533, DOI: 10.1038/sj.onc.1209489Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xnsl2jsbs%253D&md5=2447d02a4be4f41fcf2939602b149948BH3 peptidomimetics potently activate apoptosis and demonstrate single agent efficacy in neuroblastomaGoldsmith, K. C.; Liu, X.; Dam, V.; Morgan, B. T.; Shabbout, M.; Cnaan, A.; Letai, A.; Korsmeyer, S. J.; Hogarty, M. D.Oncogene (2006), 25 (33), 4525-4533CODEN: ONCNES; ISSN:0950-9232. (Nature Publishing Group)The major impediment to cure for many malignancies is the development of therapy resistance with resultant tumor progression. Genetic alterations leading to subversion of inherent apoptosis pathways are common themes in therapy resistance. Bcl-2 family proteins play a crit. role in regulating mitochondrial apoptosis that governs chemotherapeutic effects, and defective engagement of these pathways contributes to treatment failure. The authors have studied the efficacy of BH3 peptidomimetics consisting of the minimal death, or BH3, domains of the proapoptotic BH3-only proteins Bid and Bad to induce apoptosis using neuroblastoma (NB) as a model system. The authors demonstrate that BH3 peptides, modified with an arginine homopolymer for membrane transduction (called r8-BidBH3 and r8-BadBH3, resp.), potently induce apoptosis in NB cells, including those with MYCN amplification. Cell death is caspase 9 dependent, consistent with a requirement for the intrinsic mitochondrial pathway. Substitutions at highly conserved residues within the r8-BidBH3 peptide abolish apoptotic efficacy supporting activity through specific BH domain interactions. Concomitant exposure to r8-BadBH3 and r8-BidBH3 at sublethal monotherapy doses revealed potent synergy consistent with a competitive displacement model, whereby BH3 peptides displace sequestered BH3 proteins to induce cell death. Further, BH3 peptides demonstrate antitumor efficacy in a xenograft model of NB in the absence of addnl. genotoxic or trophic stressors. These data provide proof of principle that targeted reengagement of apoptosis pathways may be of therapeutic utility, and BH3-like compds. are attractive lead agents to reestablish therapy-induced apoptosis in refractory malignancies.
- 7Walensky, L. D.; Kung, A. L.; Escher, I.; Malia, T. J.; Barbuto, S.; Wright, R. D.; Wagner, G.; Verdine, G. L.; Korsmeyer, S. J. Activation of apoptosis in vivo by a hydrocarbon-stapled BH3 helix. Science 2004, 305 (5689), 1466– 1470, DOI: 10.1126/science.1099191Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXntFart7c%253D&md5=53fca0524df7a584ccf9d0991bc1bb64Activation of Apoptosis in Vivo by a Hydrocarbon-Stapled BH3 HelixWalensky, Loren D.; Kung, Andrew L.; Escher, Iris; Malia, Thomas J.; Barbuto, Scott; Wright, Renee D.; Wagner, Gerhard; Verdine, Gregory L.; Korsmeyer, Stanley J.Science (Washington, DC, United States) (2004), 305 (5689), 1466-1470CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)BCL-2 family proteins constitute a crit. control point for the regulation of apoptosis. Protein interaction between BCL-2 members is a prominent mechanism of control and is mediated through the amphipathic α-helical BH3 segment, an essential death domain. We used a chem. strategy, termed hydrocarbon stapling, to generate BH3 peptides with improved pharmacol. properties. The stapled peptides, called "stabilized alpha-helix of BCL-2 domains" (SAHBs), proved to be helical, protease-resistant, and cell-permeable mols. that bound with increased affinity to multidomain BCL-2 member pockets. A SAHB of the BH3 domain from the BID protein specifically activated the apoptotic pathway to kill leukemia cells. In addn., SAHB effectively inhibited the growth of human leukemia xenografts in vivo. Hydrocarbon stapling of native peptides may provide a useful strategy for exptl. and therapeutic modulation of protein-protein interactions in many signaling pathways.
- 8Baar, M. P.; Brandt, R. M. C.; Putavet, D. A.; Klein, J. D. D.; Derks, K. W. J.; Bourgeois, B. R. M.; Stryeck, S.; Rijksen, Y.; van Willigenburg, H.; Feijtel, D. A. Targeted Apoptosis of Senescent Cells Restores Tissue Homeostasis in Response to Chemotoxicity and Aging. Cell 2017, 169 (1), 132– 147.e116, DOI: 10.1016/j.cell.2017.02.031Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXltVWnsrk%253D&md5=5b907eb349b0e3014186f56893af362eTargeted Apoptosis of Senescent Cells Restores Tissue Homeostasis in Response to Chemotoxicity and AgingBaar, Marjolein P.; Brandt, Renata M. C.; Putavet, Diana A.; Klein, Julian D. D.; Derks, Kasper W. J.; Bourgeois, Benjamin R. M.; Stryeck, Sarah; Rijksen, Yvonne; van Willigenburg, Hester; Feijtel, Danny A.; van der Pluijm, Ingrid; Essers, Jeroen; van Cappellen, Wiggert A.; van IJcken, Wilfred F.; Houtsmuller, Adriaan B.; Pothof, Joris; de Bruin, Ron W. F.; Madl, Tobias; Hoeijmakers, Jan H. J.; Campisi, Judith; de Keizer, Peter L. J.Cell (Cambridge, MA, United States) (2017), 169 (1), 132-147.e16CODEN: CELLB5; ISSN:0092-8674. (Cell Press)The accumulation of irreparable cellular damage restricts health span after acute stress or natural aging. Senescent cells are thought to impair tissue function, and their genetic clearance can delay features of aging. Identifying how senescent cells avoid apoptosis allows for the prospective design of anti-senescence compds. to address whether homeostasis can also be restored. Here, the authors identify FOXO4 as a pivot in senescent cell viability. The authors designed a FOXO4 peptide that perturbs the FOXO4 interaction with p53. In senescent cells, this selectively causes p53 nuclear exclusion and cell-intrinsic apoptosis. Under conditions where it was well tolerated in vivo, this FOXO4 peptide neutralized doxorubicin-induced chemotoxicity. Moreover, it restored fitness, fur d., and renal function in both fast aging XpdTTD/TTD and naturally aged mice. Thus, therapeutic targeting of senescent cells is feasible under conditions where loss of health has already occurred, and in doing so tissue homeostasis can effectively be restored.
- 9Bobone, S.; Pannone, L.; Biondi, B.; Solman, M.; Flex, E.; Canale, V. C.; Calligari, P.; De Faveri, C.; Gandini, T.; Quercioli, A. Targeting Oncogenic Src Homology 2 Domain-Containing Phosphatase 2 (SHP2) by Inhibiting Its Protein-Protein Interactions. J. Med. Chem. 2021, 64 (21), 15973– 15990, DOI: 10.1021/acs.jmedchem.1c01371Google ScholarThere is no corresponding record for this reference.
- 10Zottel, A.; Novak, M.; Samec, N.; Majc, B.; Colja, S.; Katrasnik, M.; Vittori, M.; Hrastar, B.; Rotter, A.; Porcnik, A. Anti-Vimentin Nanobody Decreases Glioblastoma Cell Invasion In Vitro and In Vivo. Cancers 2023, 15 (3), 573, DOI: 10.3390/cancers15030573Google ScholarThere is no corresponding record for this reference.
- 11Deprey, K.; Becker, L.; Kritzer, J.; Pluckthun, A. Trapped! A Critical Evaluation of Methods for Measuring Total Cellular Uptake versus Cytosolic Localization. Bioconjugate Chem. 2019, 30 (4), 1006– 1027, DOI: 10.1021/acs.bioconjchem.9b00112Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXkvFWjtrw%253D&md5=3d134d4d06429a27f0561329228ac2ecTrapped! A Critical Evaluation of Methods for Measuring Total Cellular Uptake versus Cytosolic LocalizationDeprey, Kirsten; Becker, Lukas; Kritzer, Joshua; Pluckthun, AndreasBioconjugate Chemistry (2019), 30 (4), 1006-1027CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)Biomols. have many properties that make them promising for intracellular therapeutic applications, but delivery remains a key challenge because large biomols. cannot easily enter the cytosol. Furthermore, quantification of total intracellular vs. cytosolic concns. remains demanding, and the detn. of delivery efficiency is thus not straightforward. In this review, we discuss strategies for delivering biomols. into the cytosol and briefly summarize the mechanisms of uptake for these systems. We then describe commonly used methods to measure total cellular uptake and, more selectively, cytosolic localization, and discuss the major advantages and drawbacks of each method. We critically evaluate methods of measuring "cell penetration" that do not adequately distinguish total cellular uptake and cytosolic localization, which often lead to inaccurate interpretations of a mol.'s cytosolic localization. Finally, we summarize the properties and components of each method, including the main caveats of each, to allow for informed decisions about method selection for specific applications. When applied correctly and interpreted carefully, methods for quantifying cytosolic localization offer valuable insight into the bioactivity of biomols. and potentially the prospects for their eventual development into therapeutics.
- 12Schneider, A. F. L.; Kithil, M.; Cardoso, M. C.; Lehmann, M.; Hackenberger, C. P. R. Cellular uptake of large biomolecules enabled by cell-surface-reactive cell-penetrating peptide additives. Nat. Chem. 2021, 13 (6), 530– 539, DOI: 10.1038/s41557-021-00661-xGoogle Scholar12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXptVWntL4%253D&md5=573ed27e9ca9ba324141e1d4761b9e8dCellular uptake of large biomolecules enabled by cell-surface-reactive cell-penetrating peptide additivesSchneider, Anselm F. L.; Kithil, Marina; Cardoso, M. Cristina; Lehmann, Martin; Hackenberger, Christian P. R.Nature Chemistry (2021), 13 (6), 530-539CODEN: NCAHBB; ISSN:1755-4330. (Nature Portfolio)Abstr.: Enabling the cellular delivery and cytosolic bioavailability of functional proteins constitutes a major challenge for the life sciences. Here we demonstrate that thiol-reactive arginine-rich peptide additives can enhance the cellular uptake of protein-CPP conjugates in a non-endocytic mode, even at low micromolar concn. We show that such thiol- or HaloTag-reactive additives can result in covalently anchored CPPs on the cell surface, which are highly effective at co-delivering protein cargoes. Taking advantage of the thiol reactivity of our most effective CPP additive, we show that Cys-contg. proteins can be readily delivered into the cytosol by simple co-addn. of a slight excess of this CPP. Furthermore, we demonstrate the application of our 'CPP-additive technique' in the delivery of functional enzymes, nanobodies and full-length Ig-G antibodies. This new cellular uptake protocol greatly simplifies both the accessibility and efficiency of protein and antibody delivery, with minimal chem. or genetic engineering.
- 13Kauffman, W. B.; Guha, S.; Wimley, W. C. Synthetic molecular evolution of hybrid cell penetrating peptides. Nat. Commun. 2018, 9 (1), 2568 DOI: 10.1038/s41467-018-04874-6Google Scholar13https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3c%252Fkt1Kktg%253D%253D&md5=50a72964b3069b3d25abfb5967f7b8daSynthetic molecular evolution of hybrid cell penetrating peptidesKauffman W Berkeley; Guha Shantanu; Wimley William CNature communications (2018), 9 (1), 2568 ISSN:.Peptides and analogs such as peptide nucleic acids (PNA) are promising tools and therapeutics, but the cell membrane remains a barrier to intracellular targets. Conjugation to classical cell penetrating peptides (CPPs) such as pTat48-60 (tat) and pAntp43-68 (penetratin) facilitates delivery; however, efficiencies are low. Lack of explicit design principles hinders rational improvement. Here, we use synthetic molecular evolution (SME) to identify gain-of-function CPPs with dramatically improved ability to deliver cargoes to cells at low concentration. A CPP library containing 8192 tat/penetratin hybrid peptides coupled to an 18-residue PNA is screened using the HeLa pTRE-LucIVS2 splice correction reporter system. The daughter CPPs identified are one to two orders of magnitude more efficient than the parent sequences at delivery of PNA, and also deliver a dye cargo and an anionic peptide cargo. The significant increase in performance following a single iteration of SME demonstrates the power of this approach to peptide sequence optimization.
- 14Kauffman, W. B.; Fuselier, T.; He, J.; WC, W. Mechanism matters: A taxonomy of cell penetrating peptides. Trends Biochem. Sci. 2015, 40, 739– 764, DOI: 10.1016/j.tibs.2015.10.004Google ScholarThere is no corresponding record for this reference.
- 15Bartoš, L.; Vacha, R. Peptide translocation across asymmetric phospholipid membranes. Biophys. J. 2024, 123, 693– 702, DOI: 10.1016/j.bpj.2024.02.006Google ScholarThere is no corresponding record for this reference.
- 16Bus, T.; Traeger, A.; Schubert, U. S. The great escape: how cationic polyplexes overcome the endosomal barrier. J. Mater. Chem. B 2018, 6 (43), 6904– 6918, DOI: 10.1039/C8TB00967HGoogle Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhslSrsLfJ&md5=62d63fe76a4b75545afeaa170bffce43The great escape: how cationic polyplexes overcome the endosomal barrierBus, Tanja; Traeger, Anja; Schubert, Ulrich S.Journal of Materials Chemistry B: Materials for Biology and Medicine (2018), 6 (43), 6904-6918CODEN: JMCBDV; ISSN:2050-7518. (Royal Society of Chemistry)A review. The targeted and efficiency-oriented delivery of (therapeutic) nucleic acids raises hope for successful gene therapy, i.e., for the local and individual treatment of acquired and inherited genetic disorders. Despite promising achievements in the field of polymer-mediated gene delivery, the efficiency of the non-viral vectors remains orders of magnitude lower than viral-mediated ones. Several obstacles on the mol. and cellular level along the gene delivery process were identified, starting from the design and formulation of the nano-sized carriers up to the targeted release to their site of action. In particular, the efficient escape from endo-lysosomal compartments was demonstrated to be a major barrier and its exact mechanism still remains unclear. Different hypotheses and theories of the endosomal escape were postulated. The most popular one is the so-called "proton sponge" hypothesis, claiming an escape by rupture of the endosome through osmotic swelling. It was the first effort to explain the excellent transfection efficiency of poly(ethylene imine). Moreover, it was thought that a unique mechanism based on the ability to capture protons and to buffer the endosomal pH is the basis of endosomal escape. Recent theories deal with the direct interaction of the cationic polyplex or free polymer with the exoplasmic lipid leaflet causing membrane destabilization, permeability or polymer-supported nanoscale hole formation. Both escape strategies are more related to viral-mediated escape compared to the "proton sponge" effect. This review addresses the different endosomal release theories and highlights their key mechanism.
- 17Moulay, G.; Leborgne, C.; Mason, A. J.; Aisenbrey, C.; Kichler, A.; Bechinger, B. Histidine-rich designer peptides of the LAH4 family promote cell delivery of a multitude of cargo. J. Peptide Sci. 2017, 23 (4), 320– 328, DOI: 10.1002/psc.2955Google ScholarThere is no corresponding record for this reference.
- 18Langel, U. CPP, Cell Penetrating Peptides; Springer Nature, 2019.Google ScholarThere is no corresponding record for this reference.
- 19Wimley, W. C. Describing the mechanism of antimicrobial peptide action with the interfacial activity model. ACS Chem. Biol. 2010, 5 (10), 905– 917, DOI: 10.1021/cb1001558Google Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhtVGgsLbF&md5=975166f203aa8a570841091eb8501beeDescribing the Mechanism of Antimicrobial Peptide Action with the Interfacial Activity ModelWimley, William C.ACS Chemical Biology (2010), 5 (10), 905-917CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)A review. Antimicrobial peptides (AMPs) have been studied for three decades, and yet a mol. understanding of their mechanism of action is still lacking. Here we summarize current knowledge for both synthetic vesicle expts. and microbe expts., with a focus on comparisons between the two. Microbial expts. are done at peptide to lipid ratios that are at least 4 orders of magnitude higher than vesicle-based expts. To close the gap between the two concn. regimes, we propose an "interfacial activity model", which is based on an exptl. testable mol. image of AMP-membrane interactions. The interfacial activity model may be useful in driving engineering and design of novel AMPs.
- 20Marschall, A. L.; Zhang, C.; Frenzel, A.; Schirrmann, T.; Hust, M.; Perez, F.; Dubel, S. Delivery of antibodies to the cytosol: debunking the myths. MAbs 2014, 6 (4), 943– 956, DOI: 10.4161/mabs.29268Google ScholarThere is no corresponding record for this reference.
- 21Wimley, W. C. Synthetic Molecular Evolution of Cell Penetrating Peptides. Methods Mol. Biol. 2022, 2383, 73– 89, DOI: 10.1007/978-1-0716-1752-6_5Google ScholarThere is no corresponding record for this reference.
- 22Starr, C. G.; Ghimire, J.; Guha, S.; Hoffmann, J. P.; Wang, Y.; Sun, L.; Landreneau, B. N.; Kolansky, Z. D.; Kilanowski-Doroh, I. M.; Sammarco, M. C. Synthetic molecular evolution of host cell-compatible, antimicrobial peptides effective against drug-resistant, biofilm-forming bacteria. Proc. Natl. Acad. Sci. U.S.A. 2020, 117 (15), 8437– 8448, DOI: 10.1073/pnas.1918427117Google ScholarThere is no corresponding record for this reference.
- 23Li, S.; Kim, S. Y.; Pittman, A. E.; King, G. M.; Wimley, W. C.; Hristova, K. Potent Macromolecule-Sized Poration of Lipid Bilayers by the Macrolittins, A Synthetically Evolved Family of Pore-Forming Peptides. J. Am. Chem. Soc. 2018, 140 (20), 6441– 6447, DOI: 10.1021/jacs.8b03026Google Scholar23https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXosVWitrs%253D&md5=f2f6ba0fc30294d7e94b957db4fc856fPotent Macromolecule-Sized Poration of Lipid Bilayers by the Macrolittins, A Synthetically Evolved Family of Pore-Forming PeptidesLi, Sijia; Kim, Sarah Y.; Pittman, Anna E.; King, Gavin M.; Wimley, William C.; Hristova, KalinaJournal of the American Chemical Society (2018), 140 (20), 6441-6447CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Pore-forming peptides with novel functions have potential utility in many biotechnol. applications. However, the sequence-structure-function relationships of pore forming peptides are not understood well enough to empower rational design. Therefore, in this work we used synthetic mol. evolution to identify a novel family of peptides that are highly potent and cause macromol. poration in synthetic lipid vesicles at low peptide concn. and at neutral pH. These unique 26-residue peptides, which we call macrolittins, release macromols. from lipid bilayer vesicles made from zwitterionic PC lipids at peptide to lipid ratios as low as 1:1000, a property that is almost unprecedented among known membrane permeabilizing peptides. The macrolittins exist as membrane-spanning α-helixes. They cause dramatic bilayer thinning and form large pores in planar supported bilayers. The high potency of these peptides is likely due to their ability to stabilize bilayer edges by a process that requires specific electrostatic interactions between peptides.
- 24Krauson, A. J.; He, J.; Hoffmann, A. R.; Wimley, A. W.; Wimley, W. C. Synthetic molecular evolution of pore-forming peptides by Iterative combinatorial library screening. ACS Chem. Biol. 2013, 8, 823– 831, DOI: 10.1021/cb300598kGoogle Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXit1Squrw%253D&md5=f0999c8059edf784b706174f62aab14aSynthetic Molecular Evolution of Pore-Forming Peptides by Iterative Combinatorial Library ScreeningKrauson, Aram J.; He, Jing; Wimley, Andrew W.; Hoffmann, Andrew R.; Wimley, William C.ACS Chemical Biology (2013), 8 (4), 823-831CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)We previously reported the de novo design of a combinatorial peptide library that was subjected to high-throughput screening to identify membrane-permeabilizing antimicrobial peptides that have β-sheet-like secondary structure. Those peptides do not form discrete pores in membranes but instead partition into membrane interfaces and cause transient permeabilization by membrane disruption, but only when present at high concn. In this work, we used a consensus sequence from that initial screen as a template to design an iterative, second generation library. In the 24-26-residue, 16,200-member second generation library we varied six residues. Two diad repeat motifs of alternating polar and nonpolar amino acids were preserved to maintain a propensity for non-helical secondary structure. We used a new high-throughput assay to identify members that self- assemble into equil. pores in synthetic lipid bilayers. This screen was done at a very stringent peptide to lipid ratio of 1:1000 where most known membrane-permeabilizing peptides, including the template peptide, are not active. In a screen of 10,000 library members we identified 16 (∼0.2%) that are equil. pore-formers at this high stringency. These rare and highly active peptides, which share a common sequence motif, are as potent as the most active pore-forming peptides known. Furthermore, they are not α-helical, which makes them unusual, as most of the highly potent pore-forming peptides are amphipathic α-helixes. Here we demonstrate that this synthetic mol. evolution-based approach, taken together with the new high-throughput tools we have developed, enables the identification, refinement, and optimization of unique membrane active peptides.
- 25Tosteson, M. T.; Tosteson, D. C. The sting. Melittin forms channels in lipid bilayers. Biophys. J. 1981, 36 (1), 109– 116, DOI: 10.1016/S0006-3495(81)84719-4Google ScholarThere is no corresponding record for this reference.
- 26Krauson, A. J.; He, J.; Wimley, W. C. Gain-of-Function Analogues of the Pore-Forming Peptide Melittin Selected by Orthogonal High-Throughput Screening. J. Am. Chem. Soc. 2012, 134 (30), 12732– 12741, DOI: 10.1021/ja3042004Google Scholar26https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XptF2hsrY%253D&md5=e15fc7f6b1869548ec0154cbcc24c686Gain-of-function analogues of the pore-forming peptide melittin selected by orthogonal high-throughput screeningKrauson, Aram J.; He, Jing; Wimley, William C.Journal of the American Chemical Society (2012), 134 (30), 12732-12741CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)The authors recently developed an orthogonal, high-throughput assay to identify peptides that self-assemble into potent, equil. pores in synthetic lipid bilayers. Here, the authors use this assay as a high-throughput screen to select highly potent pore-forming peptides from a 7776-member rational combinatorial peptide library based on the sequence of the natural pore-forming peptide toxin melittin. In the library the authors varied ten crit. residues in the melittin sequence, chosen to test specific structural hypotheses about the mechanism of pore formation. Using the new high-throughput assay, the authors screened the library for gain-of-function sequences at a peptide to lipid ratio of 1:1000 where native melittin is not active. More than 99% of the library sequences were also inactive under these conditions. A small no. of library members (0.1%) were highly active. From these the authors identified 14 potent, gain-of-function, pore-forming sequences. These sequences differed from melittin in only 2-6 amino acids out of 26. Some native residues were highly conserved and others were consistently changed. The two factors that were essential for gain-of-function were the preservation of melittin's proline-dependent break in the middle of the helix and the improvement and extension the amphipathic nature of the α-helix. In particular the highly cationic carboxyl-terminal sequence of melittin is consistently changed in the gain-of-function variants to a sequence that it is capable of participating in an extended amphipathic α-helix. The most potent variants reside in a membrane-spanning orientation, in contrast to the parent melittin, which is predominantly surface bound. This structural information, taken together with the high-throughput tools developed for this work, enable the identification, refinement and optimization of pore-forming peptides for many potential applications.
- 27Wiedman, G.; Fuselier, T.; He, J.; Searson, P. C.; Hristova, K.; Wimley, W. C. Highly efficient macromolecule-sized poration of lipid bilayers by a synthetically evolved peptide. J. Am. Chem. Soc. 2014, 136 (12), 4724– 4731, DOI: 10.1021/ja500462sGoogle Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXjtlOlsbg%253D&md5=5accca4a4bc0e23542e62683ed1e8056Highly Efficient Macromolecule-Sized Poration of Lipid Bilayers by a Synthetically Evolved PeptideWiedman, Gregory; Fuselier, Taylor; He, Jing; Searson, Peter C.; Hristova, Kalina; Wimley, William C.Journal of the American Chemical Society (2014), 136 (12), 4724-4731CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Peptides that self-assemble, at low concn., into bilayer-spanning pores which allow the passage of macromols. would be beneficial in multiple areas of biotechnol. However, there are few, if any, natural or designed peptides that have this property. The 26-residue peptide "MelP5", a synthetically evolved gain-of-function variant of the bee venom lytic peptide melittin identified in a high-throughput screen for small mol. leakage, enables the passage of macromols. across bilayers under conditions where melittin and other pore-forming peptides do not. In surface-supported bilayers, MelP5 forms unusually high conductance, equil. pores at peptide:lipid ratios as low as 1:25000. The increase in bilayer conductance due to MelP5 is dramatically higher, per peptide, than the increase due to the parent sequence of melittin or other peptide pore formers. Here the authors also develop two novel assays for macromol. leakage from vesicles, and they use them to characterize MelP5 pores in bilayers. MelP5 allows the passage of macromols. across vesicle membranes at peptide:lipid ratios as low as 1:500, and under conditions where neither osmotic lysis nor gross vesicle destabilization occur. The macromol.-sized, equil. pores formed by MelP5 are unique as neither melittin nor other pore-forming peptides release macromols. significantly under the same conditions. MelP5 thus appears to belong to a novel functional class of peptide that could form the foundation of multiple potential biotechnol. applications.
- 28Wiedman, G.; Kim, S. Y.; Zapata-Mercado, E.; Wimley, W. C.; Hristova, K. PH-Triggered, Macromolecule-Sized Poration of Lipid Bilayers by Synthetically Evolved Peptides. J. Am. Chem. Soc. 2017, 139, 937– 945, DOI: 10.1021/jacs.6b11447Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XitFaiurjN&md5=22f0314dfb54a347cf980012204fa247pH-Triggered, Macromolecule-Sized Poration of Lipid Bilayers by Synthetically Evolved PeptidesWiedman, Gregory; Kim, Sarah Y.; Zapata-Mercado, Elmer; Wimley, William C.; Hristova, KalinaJournal of the American Chemical Society (2017), 139 (2), 937-945CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)PH-triggered membrane-permeabilizing peptides could be exploited in a variety of applications, such as to enable cargo release from endosomes for cellular delivery, or as cancer therapeutics that selectively permeabilize the plasma membranes of malignant cells. Such peptides would be esp. useful if they could enable the movement of macromols. across membranes, a rare property in membrane-permeabilizing peptides. Here the authors approach this goal by using an orthogonal high-throughput screen of an iterative peptide library to identify peptide sequences that have the following two properties: (1) little synthetic lipid membrane permeabilization at physiol. pH 7 at high peptide concn. and (2) efficient formation of macromol.-sized defects in synthetic lipid membranes at acidic pH 5 and low peptide concn. The peptides the authors selected are remarkably potent macromol. sized pore-formers at pH 5, while having little or no activity at pH 7, as intended. The action of these peptides likely relies on tight coupling between membrane partitioning, α-helix formation, and electrostatic repulsions between acidic side chains, which collectively drive a sharp pH-triggered transition between inactive and active configurations with apparent pKa values of 5.5-5.8. This work opens new doors to developing applications that use peptides with membrane-permeabilizing activities that are triggered by physiol. relevant decreases in pH.
- 29Kim, S. Y.; Bondar, A. N.; Wimley, W. C.; Hristova, K. pH-triggered pore-forming peptides with strong composition-dependent membrane selectivity. Biophys. J. 2021, 120 (4), 618– 630, DOI: 10.1016/j.bpj.2021.01.010Google Scholar29https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXisVyktbc%253D&md5=80fdd94720a95e5d9c15ffd1185137fcpH-triggered pore-forming peptides with strong composition-dependent membrane selectivityKim, Sarah Y.; Bondar, Ana-Nicoleta; Wimley, William C.; Hristova, KalinaBiophysical Journal (2021), 120 (4), 618-630CODEN: BIOJAU; ISSN:0006-3495. (Cell Press)Peptides that self-assemble into nanometer-sized pores in lipid bilayers could have utility in a variety of biotechnol. and clin. applications if we can understand their phys. chem. properties and learn to control their membrane selectivity. To empower such control, we have used synthetic mol. evolution to identify the pH-dependent delivery peptides, a family of peptides that assemble into macromol.-sized pores in membranes at low peptide concn. but only at pH < ∼6. Further advancements will also require better selectivity for specific membranes. Here, we det. the effect of anionic headgroups and bilayer thickness on the mechanism of action of the pH-dependent delivery peptides by measuring binding, secondary structure, and macromol. poration. The peptide pHD15 partitions and folds equally well into zwitterionic and anionic membranes but is less potent at pore formation in phosphatidylserine-contg. membranes. The peptide also binds and folds similarly in membranes of various thicknesses, but its ability to release macromols. changes dramatically. It causes potent macromol. poration in vesicles made from phosphatidylcholine with 14 carbon acyl chains, but macromol. poration decreases sharply with increasing bilayer thickness and does not occur at any peptide concn. in fluid bilayers made from phosphatidylcholine lipids with 20-carbon acyl chains. The effects of headgroup and bilayer thickness on macromol. poration cannot be accounted for by the amt. of peptide bound but instead reflect an inherent selectivity of the peptide for inserting into the membrane-spanning pore state. Mol. dynamics simulations suggest that the effect of thickness is due to hydrophobic match/mismatch between the membrane-spanning peptide and the bilayer hydrocarbon. This remarkable degree of selectivity based on headgroup and esp. bilayer thickness is unusual and suggests ways that pore-forming peptides with exquisite selectivity for specific membranes can be designed or evolved.
- 30Kim, S. Y.; Pittman, A. E.; Zapata-Mercado, E.; King, G. M.; Wimley, W. C.; Hristova, K. Mechanism of Action of Peptides That Cause the pH-Triggered Macromolecular Poration of Lipid Bilayers. J. Am. Chem. Soc. 2019, 141 (16), 6706– 6718, DOI: 10.1021/jacs.9b01970Google Scholar30https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXmtFGntbs%253D&md5=2e292bc579d5cd82b71f412ba386acd1Mechanism of action of peptides that cause the pH-triggered macromolecular poration of lipid bilayersKim, Sarah Y.; Pittman, Anna E.; Zapata-Mercado, Elmer; King, Gavin M.; Wimley, William C.; Hristova, KalinaJournal of the American Chemical Society (2019), 141 (16), 6706-6718CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Using synthetic mol. evolution, we previously discovered a family of peptides that cause macromol. poration in synthetic membranes at low peptide concn. in a way that is triggered by acidic pH. To understand the mechanism of action of these "pHD peptides", here we systematically explored structure-function relationships through measurements of the effect of pH and peptide concn. on membrane binding, peptide structure, and the formation of macromol.-sized pores in membranes. Both AFM and functional assays demonstrate the peptide-induced appearance of large pores in bilayers. Pore formation has a very steep pH dependence and is also dependent on peptide concn. In vesicles, 50% leakage of 40 kDa dextrans occurs at 1 bound peptide per 1300 lipids or only 75 peptides per vesicle, an observation that holds true across a wide range of acidic pH values. The major role of pH is to regulate the amt. of peptide bound per vesicle. The phys. chem. and sequence of the pHD peptides affect their potency and pH dependence; therefore, the sequence-structure-function relationships described here can be used for the future design and optimization of membrane permeabilizing peptides for specific applications.
- 31Wiedman, G.; Wimley, W. C.; Hristova, K. Testing the limits of rational design by engineering pH sensitivity into membrane-active peptides. Biochim. Biophys. Acta 2015, 1848 (4), 951– 957, DOI: 10.1016/j.bbamem.2014.12.023Google ScholarThere is no corresponding record for this reference.
- 32Guha, S.; Ferrie, R. P.; Ghimire, J.; Ventura, C. R.; Wu, E.; Sun, L.; Kim, S. Y.; Wiedman, G. R.; Hristova, K.; Wimley, W. C. Applications and evolution of melittin, the quintessential membrane active peptide. Biochem. Pharmacol. 2021, 193, 114769 DOI: 10.1016/j.bcp.2021.114769Google Scholar32https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXitFGhtrvI&md5=e16c4e779ab73131267b2942d2aa5d4eApplications and evolution of melittin, the quintessential membrane active peptideGuha, Shantanu; Ferrie, Ryan P.; Ghimire, Jenisha; Ventura, Cristina R.; Wu, Eric; Sun, Leisheng; Kim, Sarah Y.; Wiedman, Gregory R.; Hristova, Kalina; Wimley, Wimley C.Biochemical Pharmacology (Amsterdam, Netherlands) (2021), 193 (), 114769CODEN: BCPCA6; ISSN:0006-2952. (Elsevier B.V.)A review. Melittin, the main venom component of the European Honeybee, is a cationic linear peptide-amide of 26 amino acid residues with the sequence: GIGAVLKVLTTGLPALISWIKRKRQQ-NH2. Melittin binds to lipid bilayer membranes, folds into amphipathic α-helical secondary structure and disrupts the permeability barrier. Since melittin was first described, a remarkable array of activities and potential applications in biol. and medicine have been described. Melittin is also a favorite model system for biophysicists to study the structure, folding and function of peptides and proteins in membranes. Melittin has also been used as a template for the evolution of new activities in membranes. Here we overview the rich history of scientific research into the many activities of melittin and outline exciting future applications.
- 33Guha, S.; Ghimire, J.; Wu, E.; Wimley, W. C. Mechanistic Landscape of Membrane-Permeabilizing Peptides. Chem. Rev. 2019, 119, 6040– 6085, DOI: 10.1021/acs.chemrev.8b00520Google Scholar33https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXltV2jug%253D%253D&md5=a591e43ebc009cbe483ba932a2b93feeMechanistic landscape of membrane-permeabilizing peptidesGuha, Shantanu; Ghimire, Jenisha; Wu, Eric; Wimley, William C.Chemical Reviews (Washington, DC, United States) (2019), 119 (9), 6040-6085CODEN: CHREAY; ISSN:0009-2665. (American Chemical Society)A review. Membrane-permeabilizing peptides (MPPs) are as ubiquitous as the lipid bilayer membranes they act upon. Produced by all forms of life, most MPPs are used offensively or defensively against the membranes of other organisms. Just as Nature has found many uses for them, translational scientists have worked for decades to design or optimize MPPs for applications in the lab. and in the clinic ranging from antibacterial and antiviral therapy and prophylaxis to anticancer therapeutics and drug delivery. Here, we review the field of MPPs. We discuss the diversity of their sources and structures, the systems and methods used to measure their activities, and the behaviors that are obsd. We discuss the fact that "mechanism" is not a discrete or a static entity for a MPP, but rather the result of a heterogeneous and dynamic ensemble of structural states that vary in response to many different exptl. conditions. This has led to an almost complete lack of discrete 3-dimensional active structures among the thousands of known MPPs and a lack of useful or predictive sequence-structure-function relation rules. Ultimately, we discuss how it may be more useful to think of MPP mechanisms as broad regions of a mechanistic landscape rather than discrete mol. processes.
- 34Sun, L.; Hristova, K.; Bondar, A.-N.; Wimley, W. C. Structural Determinants of Peptide Nanopore Formation. ACS Nano 2024, 18, 15831– 15854, DOI: 10.1021/acsnano.4c02824Google ScholarThere is no corresponding record for this reference.
- 35Michalska, M.; Wolf, P. Pseudomonas Exotoxin A: optimized by evolution for effective killing. Front. Microbiol. 2015, 6, 963, DOI: 10.3389/fmicb.2015.00963Google Scholar35https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC283ps1akug%253D%253D&md5=20c632c00c9c4e16292642a971b6ad47Pseudomonas Exotoxin A: optimized by evolution for effective killingMichalska Marta; Wolf PhilippFrontiers in microbiology (2015), 6 (), 963 ISSN:1664-302X.Pseudomonas Exotoxin A (PE) is the most toxic virulence factor of the pathogenic bacterium Pseudomonas aeruginosa. This review describes current knowledge about the intoxication pathways of PE. Moreover, PE represents a remarkable example for pathoadaptive evolution, how bacterial molecules have been structurally and functionally optimized under evolutionary pressure to effectively impair and kill their host cells.
- 36Deng, Q.; Barbieri, J. T. Molecular mechanisms of the cytotoxicity of ADP-ribosylating toxins. Annu. Rev. Microbiol. 2008, 62, 271– 288, DOI: 10.1146/annurev.micro.62.081307.162848Google Scholar36https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXht1Gls73N&md5=248e3f5084b0338ecfb1af1df7b31ad8Molecular mechanisms of the cytotoxicity of ADP-ribosylating toxinsDeng, Qing; Barbieri, Joseph T.Annual Review of Microbiology (2008), 62 (), 271-288CODEN: ARMIAZ; ISSN:0066-4227. (Annual Reviews Inc.)A review. Bacterial pathogens utilize toxins to modify or kill host cells. The bacterial ADP-ribosyltransferases are a family of protein toxins that covalently transfer the ADP-ribose portion of NAD to host proteins. Each bacterial ADP-ribosyltransferase toxin modifies a specific host protein(s) that yields a unique pathol. These toxins possess the capacity to enter a host cell or to use a bacterial type III app. for delivery into the host cell. Advances in the understanding of bacterial toxin action parallel the development of biophys. and structural biol. as well as an understanding of the mammalian cell. Bacterial toxins have been utilized as vaccines, as tools to dissect host cell physiol., and more recently for the development of novel therapies to treat human disease.
- 37Moss, D. L.; Park, H. W.; Mettu, R. R.; Landry, S. J. Deimmunizing substitutions in Pseudomonas exotoxin domain III perturb antigen processing without eliminating T-cell epitopes. J. Biol. Chem. 2019, 294 (12), 4667– 4681, DOI: 10.1074/jbc.RA118.006704Google ScholarThere is no corresponding record for this reference.
- 38Wedekind, J. E.; Trame, C. B.; Dorywalska, M.; Koehl, P.; Raschke, T. M.; McKee, M.; FitzGerald, D.; Collier, R. J.; McKay, D. B. Refined crystallographic structure of Pseudomonas aeruginosa exotoxin A and its implications for the molecular mechanism of toxicity. J. Mol. Biol. 2001, 314 (4), 823– 837, DOI: 10.1006/jmbi.2001.5195Google ScholarThere is no corresponding record for this reference.
- 39Marks, J. R.; Placone, J.; Hristova, K.; Wimley, W. C. Spontaneous membrane-translocating peptides by orthogonal high-throughput screening. J. Am. Chem. Soc. 2011, 133 (23), 8995– 9004, DOI: 10.1021/ja2017416Google Scholar39https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXmtlWrtrk%253D&md5=c47082603ff99de9b48c0f6777249ae9Spontaneous Membrane-Translocating Peptides by Orthogonal High-Throughput ScreeningMarks, Jessica R.; Placone, Jesse; Hristova, Kalina; Wimley, William C.Journal of the American Chemical Society (2011), 133 (23), 8995-9004CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Combinatorial peptide chem. and orthogonal high-throughput screening were used to select peptides that spontaneously translocate across synthetic lipid bilayer membranes without permeabilization. A conserved sequence motif was identified that contains several cationic residues in conserved positions in an otherwise hydrophobic sequence. This 9-residue motif rapidly translocates across synthetic multibilayer vesicles and into cells while carrying a large polar dye as a "cargo" moiety. The extraordinary ability of this family of peptides to spontaneously translocate across bilayers without an energy source of any kind is distinctly different from the behavior of the well-known, highly cationic cell-penetrating peptides, such as the HIV tat peptide, which do not translocate across synthetic bilayers, and enter cells mostly by active endocytosis. Peptides that translocate spontaneously across membranes have the potential to transform the field of drug design by enabling the delivery of otherwise membrane-impermeant polar drugs into cells and tissues. Here we describe the chem. tools needed to rapidly identify spontaneous membrane translocating peptides.
- 40He, J.; Kauffman, W. B.; Fuselier, T.; Naveen, S. K.; Voss, T. G.; Hristova, K.; Wimley, W. C. Direct Cytosolic Delivery of Polar Cargo to Cells by Spontaneous Membrane-translocating Peptides. J. Biol. Chem. 2013, 288 (41), 29974– 29986, DOI: 10.1074/jbc.M113.488312Google ScholarThere is no corresponding record for this reference.
- 41Armstrong, J. K.; Wenby, R. B.; Meiselman, H. J.; Fisher, T. C. The hydrodynamic radii of macromolecules and their effect on red blood cell aggregation. Biophys. J. 2004, 87 (6), 4259– 4270, DOI: 10.1529/biophysj.104.047746Google Scholar41https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXhtVOmur7L&md5=1426af6c85786a5eead545b528518fc3The hydrodynamic radii of macromolecules and their effect on red blood cell aggregationArmstrong, J. K.; Wenby, R. B.; Meiselman, H. J.; Fisher, T. C.Biophysical Journal (2004), 87 (6), 4259-4270CODEN: BIOJAU; ISSN:0006-3495. (Biophysical Society)The effects of nonionic polymers on human red blood cell (RBC) aggregation were investigated. The hydrodynamic radius (Rh) of individual samples of dextran, polyvinylpyrrolidone, and polyoxyethylene over a range of mol. wts. (1500-2,000,000) were calcd. from their intrinsic viscosities using the Einstein viscosity relation and directly measured by quasi-elastic light scattering, and the effect of each polymer sample on RBC aggregation was studied by nephelometry and low-shear viscometry. For all three polymers, despite their different structures, samples with Rh <4 nm were found to inhibit aggregation, whereas those with Rh >4 nm enhanced aggregation. Inhibition increased with Rh and was maximal at ∼3 nm; above 4 nm, the pro-aggregant effect increased with Rh. For comparison, the Rh of 12 plasma proteins were calcd. from literature values of intrinsic viscosity or diffusion coeff. Each protein known to promote RBC aggregation had Rh >4 nm, whereas those with Rh <4 nm either inhibited or had no effect on aggregation. These results suggest that the influence of a nonionic polymer or plasma protein on RBC aggregation is simply a consequence of its size in an aq. environment and that the specific type of macromol. is of minor importance.
- 42Bohrer, M. P.; Deen, W. M.; Robertson, C. R.; Troy, J. L.; Brenner, B. M. Influence of molecular configuration on the passage of macromolecules across glomerular capillary wall. J. Gen. Physiol 1979, 74, 583– 593, DOI: 10.1085/jgp.74.5.583Google ScholarThere is no corresponding record for this reference.
- 43Loret, C.; Chaufer, B.; Sebille, B.; Hamelin, M.; Blain, Y.; Le Hir, A. Characterization and hydrodynamic behaviour of modified gelatin: ii. Characterization by high performance size exclusion chromatography comparison with dextrans and proteins. Int. J. Biol. Macromol. 1988, 10, 366– 372, DOI: 10.1016/0141-8130(88)90031-1Google ScholarThere is no corresponding record for this reference.
- 44Wu, E.; Jenschke, R. M.; Hristova, K.; Wimley, W. C. Rational Modulation of pH-Triggered Macromolecular Poration by Peptide Acylation and Dimerization. J. Phys. Chem. B 2020, 124, 8835– 8843, DOI: 10.1021/acs.jpcb.0c05363Google ScholarThere is no corresponding record for this reference.
- 45Johnson, W. C. Protein secondary structure and circular dichroism: A practical guide. Proteins 1990, 7, 205– 214, DOI: 10.1002/prot.340070302Google Scholar45https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3cXksFGitbY%253D&md5=f1316896de77d441523c349575dbaaabProtein secondary structure and circular dichroism: a practical guideJohnson, W. Curtis, Jr.Proteins: Structure, Function, and Genetics (1990), 7 (3), 205-14CODEN: PSFGEY; ISSN:0887-3585.A review, with 24 refs., of CD spectroscopy in the study of protein secondary structure.
- 46Lakowicz, J. R. Principles of Fluorescence Spectroscopy; Plenum Press, 1983.Google ScholarThere is no corresponding record for this reference.
- 47Ladokhin, A. S.; Jayasinghe, S.; White, S. H. How to measure and analyze tryptophan fluorescence in membranes properly, and why bother?. Anal. Biochem. 2000, 285 (2), 235– 245, DOI: 10.1006/abio.2000.4773Google Scholar47https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXmvFOksL0%253D&md5=99c2776187f13c33656bd1f4e59e2142How to Measure and Analyze Tryptophan Fluorescence in Membranes Properly, and Why Bother?Ladokhin, Alexey S.; Jayasinghe, Sajith; White, Stephen H.Analytical Biochemistry (2000), 285 (2), 235-245CODEN: ANBCA2; ISSN:0003-2697. (Academic Press)Tryptophan fluorescence is a powerful tool for studying protein structure and function, esp. membrane-active proteins and peptides. It is arguably the most frequently used tool for examg. the interactions of proteins and peptides with vesicular unilamellar model membranes. However, high light scattering assocd. with vesicular membrane systems presents special challenges. Because of their reduced light scattering compared to large unilamellar vesicles (LUV), small unilamellar vesicles (SUV) produced by sonication are widely used membrane models. Unfortunately, SUV, unlike LUV, are metastable and consequently unsuitable for equil. thermodn. measurements. We present simple and easily implemented exptl. procedures for the accurate detn. of tryptophan (Trp) fluorescence in either LUV or SUV. Specifically, we show that Trp spectra can be obtained in the presence of up to 6 mM LUV that are virtually identical to spectra obtained in buffer alone, which obviates the use of SUV. We show how the widths and peak positions of such spectra can be used to evaluate the heterogeneity of the membrane conformation and penetration of peptides. Finally, we show how to use a ref. fluorophore for the correction of intensity measurements so that the energetics of peptide partitioning into membranes can be accurately detd. (c) 2000 Academic Press.
- 48Pittman, A. E.; Marsh, B. P.; King, G. M. Conformations and Dynamic Transitions of a Melittin Derivative That Forms Macromolecule-Sized Pores in Lipid Bilayers. Langmuir 2018, 34 (28), 8393– 8399, DOI: 10.1021/acs.langmuir.8b00804Google ScholarThere is no corresponding record for this reference.
- 49Sun, L.; Hristova, K.; Wimley, W. C. Membrane-selective nanoscale pores in liposomes by a synthetically evolved peptide: implications for triggered release. Nanoscale 2021, 13 (28), 12185– 12197, DOI: 10.1039/D1NR03084AGoogle ScholarThere is no corresponding record for this reference.
- 50Di, L. Strategic approaches to optimizing peptide ADME properties. AAPS J. 2015, 17 (1), 134– 143, DOI: 10.1208/s12248-014-9687-3Google Scholar50https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhvFSnurvJ&md5=530597d3719d65509105096f433fee0aStrategic Approaches to Optimizing Peptide ADME PropertiesDi, LiAAPS Journal (2015), 17 (1), 134-143CODEN: AJAOB6; ISSN:1550-7416. (Springer)Development of peptide drugs is challenging but also quite rewarding. Five blockbuster peptide drugs are currently on the market, and six new peptides received first marketing approval as new mol. entities in 2012. Although peptides only represent 2% of the drug market, the market is growing twice as quickly and might soon occupy a larger niche. Natural peptides typically have poor absorption, distribution, metab., and excretion (ADME) properties with rapid clearance, short half-life, low permeability, and sometimes low soly. Strategies have been developed to improve peptide drugability through enhancing permeability, reducing proteolysis and renal clearance, and prolonging half-life. In vivo, in vitro, and in silico tools are available to evaluate ADME properties of peptides, and structural modification strategies are in place to improve peptide developability.
- 51Brunsveld, L.; Waldmann, H.; Huster, D. Membrane binding of lipidated Ras peptides and proteins--the structural point of view. Biochim. Biophys. Acta 2009, 1788 (1), 273– 288, DOI: 10.1016/j.bbamem.2008.08.006Google ScholarThere is no corresponding record for this reference.
- 52Zhang, L.; Bulaj, G. Converting peptides into drug leads by lipidation. Curr. Med. Chem. 2012, 19 (11), 1602– 1618, DOI: 10.2174/092986712799945003Google Scholar52https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XmsF2jt7s%253D&md5=a9f289aea1cea75a566370a1b2c06583Converting peptides into drug leads by lipidationZhang, L.; Bulaj, G.Current Medicinal Chemistry (2012), 19 (11), 1602-1618CODEN: CMCHE7; ISSN:0929-8673. (Bentham Science Publishers Ltd.)A review. Lipidation is a posttranslational modification of proteins that has also found its use in designing peptide drugs. The presence of a lipid group in peptides modulates their hydrophobicity, secondary structures and self-assembling propensities while retaining their abilities to bind to target receptors. Lipidation improves peptides' metabolic stability, membrane permeability, bioavailability, and changes peptides' pharmacokinetic and pharmacodynamic properties. Herein, we review the applications of various lipidation strategies in peptide drug design, the effects of the chain length and anchor position of fatty acids in peptide lipidation, the physicochem. and biol. properties of selected lipidated peptides and the synthesis strategies for peptide lipidation.
- 53Ezzat, K.; Andaloussi, S. E.; Zaghloul, E. M.; Lehto, T.; Lindberg, S.; Moreno, P. M.; Viola, J. R.; Magdy, T.; Abdo, R.; Guterstam, P. PepFect 14, a novel cell-penetrating peptide for oligonucleotide delivery in solution and as solid formulation. Nucleic Acids Res. 2011, 39 (12), 5284– 5298, DOI: 10.1093/nar/gkr072Google Scholar53https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXoslOgtbw%253D&md5=64e66305efb772c25386977d53802340PepFect 14, a novel cell-penetrating peptide for oligonucleotide delivery in solution and as solid formulationEzzat, Kariem; El Andaloussi, Samir; Zaghloul, Eman M.; Lehto, Taavi; Lindberg, Staffan; Moreno, Pedro M. D.; Viola, Joana R.; Magdy, Tarek; Abdo, Rania; Guterstam, Peter; Sillard, Rannar; Hammond, Suzan M.; Wood, Matthew J. A.; Arzumanov, Andrey A.; Gait, Michael J.; Smith, C. I. Edvard; Haellbrink, Mattias; Langel, UeloNucleic Acids Research (2011), 39 (12), 5284-5298CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)Numerous human genetic diseases are caused by mutations that give rise to aberrant alternative splicing. Recently, several of these debilitating disorders have been shown to be amenable for splice-correcting oligonucleotides (SCOs) that modify splicing patterns and restore the phenotype in exptl. models. However, translational approaches are required to transform SCOs into usable drug products. In this study, we present a new cell-penetrating peptide, PepFect14 (PF14), which efficiently delivers SCOs to different cell models including HeLa pLuc705 and mdx mouse myotubes; a cell culture model of Duchenne's muscular dystrophy (DMD). Non-covalent PF14-SCO nanocomplexes induce splice-correction at rates higher than the com. available lipid-based vector Lipofectamine 2000 (LF2000) and remain active in the presence of serum. Furthermore, we demonstrate the feasibility of incorporating this delivery system into solid formulations that could be suitable for several therapeutic applications. Solid dispersion technique is utilized and the formed solid formulations are as active as the freshly prepd. nanocomplexes in soln. even when stored at an elevated temps. for several weeks. In contrast, LF2000 drastically loses activity after being subjected to same procedure. This shows that using PF14 is a very promising translational approach for the delivery of SCOs in different pharmaceutical forms.
- 54Gatto, E.; Mazzuca, C.; Stella, L.; Venanzi, M.; Toniolo, C.; Pispisa, B. Effect of peptide lipidation on membrane perturbing activity: a comparative study on two trichogin analogues. J. Phys. Chem. B 2006, 110 (45), 22813– 22818, DOI: 10.1021/jp064580jGoogle Scholar54https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XhtVyntbnN&md5=76fb933226981c03757eb547119aee16Effect of Peptide Lipidation on Membrane Perturbing Activity: A Comparative Study on Two Trichogin AnaloguesGatto, Emanuela; Mazzuca, Claudia; Stella, Lorenzo; Venanzi, Mariano; Toniolo, Claudio; Pispisa, BasilioJournal of Physical Chemistry B (2006), 110 (45), 22813-22818CODEN: JPCBFK; ISSN:1520-6106. (American Chemical Society)The effect of lipidation on the membrane perturbing activity of peptaibol antibiotics was investigated by performing a comparative study on two synthetic analogs of the natural peptide trichogin GA IV. Both analogs were labeled with a hydrophobic fluorescent probe, but one of them lacked the N-terminal n-octanoyl chain, present in the natural peptide. Spectroscopic studies show that the fatty acyl chain produces two opposite effects: It increases the affinity of the monomeric peptide for the membrane phase, but, at the same time, it favors peptide aggregation in water, thus inhibiting membrane binding by reducing the effective monomer concn. In the membrane phase the two analogs exhibit the same aggregation and orientation behavior, indicating that the n-octanoyl chain plays no specific role in detg. their orientation or membrane perturbing activity. Indeed, the dependence of peptide-induced membrane leakage on total peptide concn. is basically the same for the two analogs, because the aforementioned opposite effects, caused by peptide lipidation, tend to balance. These findings make questionable the use of lipidation as a general method for increasing the peptide membrane-perturbing activity, as its validity seems to be restricted to parent compds. of limited overall hydrophobicity.
- 55Müller, A.; Wenzel, M.; Strahl, H.; Grein, F.; Saaki, T. N. V.; Kohl, B.; Siersma, T.; Bandow, J. E.; Sahl, H. G.; Schneider, T.; Hamoen, L. W. Daptomycin inhibits cell envelope synthesis by interfering with fluid membrane microdomains. Proc. Natl. Acad. Sci. U.S.A. 2016, 113 (45), E7077– E7086, DOI: 10.1073/pnas.1611173113Google Scholar55https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhslalsLrL&md5=900e3e7b23f443b37e0a0a1ba6ec590fDaptomycin inhibits cell envelope synthesis by interfering with fluid membrane microdomainsMueller, Anna; Wenzel, Michaela; Strahl, Henrik; Grein, Fabian; Saaki, Terrens N. V.; Kohl, Bastian; Siersma, Tjalling; Bandow, Julia E.; Sahl, Hans-Georg; Schneider, Tanja; Hamoen, Leendert W.Proceedings of the National Academy of Sciences of the United States of America (2016), 113 (45), E7077-E7086CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Daptomycin is a highly efficient last-resort antibiotic that targets the bacterial cell membrane. Despite its clin. importance, the exact mechanism by which daptomycin kills bacteria is not fully understood. Different expts. have led to different models, including (i) blockage of cell wall synthesis, (ii) membrane pore formation, and (iii) the generation of altered membrane curvature leading to aberrant recruitment of proteins. To det. which model is correct, the authors carried out a comprehensive mode-of-action study using the model organism Bacillus subtilis and different assays, including proteomics, ionomics, and fluorescence light microscopy. The authors found that daptomycin causes a gradual decrease in membrane potential but does not form discrete membrane pores. Although the authors found no evidence for altered membrane curvature, the authors confirmed that daptomycin inhibits cell wall synthesis. Interestingly, using different fluorescent lipid probes, the authors showed that binding of daptomycin led to a drastic rearrangement of fluid lipid domains, affecting overall membrane fluidity. Importantly, these changes resulted in the rapid detachment of the membrane-assocd. lipid II synthase MurG and the phospholipid synthase PlsX. Both proteins preferentially colocalize with fluid membrane microdomains. Delocalization of these proteins presumably is a key reason why daptomycin blocks cell wall synthesis. Finally, clustering of fluid lipids by daptomycin likely causes hydrophobic mismatches between fluid and more rigid membrane areas. This mismatch can facilitate proton leakage and may explain the gradual membrane depolarization obsd. with daptomycin. Targeting of fluid lipid domains has not been described before for antibiotics and adds another dimension to the authors' understanding of membrane-active antibiotics.
- 56Lin, B. F.; Missirlis, D.; Krogstad, D. V.; Tirrell, M. Structural effects and lipid membrane interactions of the pH-responsive GALA peptide with fatty acid acylation. Biochemistry 2012, 51 (23), 4658– 4668, DOI: 10.1021/bi300314hGoogle Scholar56https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XntFSgsLs%253D&md5=0d8f3bc422caab96b4f5b8f373ef21f2Structural Effects and Lipid Membrane Interactions of the pH-Responsive GALA Peptide with Fatty Acid AcylationLin, Brian F.; Missirlis, Dimitris; Krogstad, Daniel V.; Tirrell, MatthewBiochemistry (2012), 51 (23), 4658-4668CODEN: BICHAW; ISSN:0006-2960. (American Chemical Society)GALA is a pH-responsive, membrane-perturbing peptide designed to fold from a random coil at physiol. pH to an amphipathic α-helix under mildly acidic conditions. Because of its pH-activated function, GALA has been sought-after as a component of intracellular drug delivery systems that could actively propel endosomal escape. In this study, we conjugated GALA with lauryl and palmitoyl fatty acid tails as model hydrophobic moieties and examd. the physicochem. characteristics and activities of the resulting peptide amphiphiles (PAs). The fatty acid variants of GALA exhibited distinctly different membrane perturbing mechanisms at pH 7.5 and 5.5. At physiol. pH, the PAs ruptured liposomes through a surfactant-like mechanism. At pH 5.5, lauryl-GALA was shown to form transmembrane pores with a higher potency as compared to its unmodified peptide counterpart; however, after prolonged exposure it also caused liposome lysis. The lytic activity of fatty acid-conjugated GALA did not impair cell viability. Lauryl-GALA was tolerated well by SJSA-1 osteocarcinoma cells and enhanced cell internalization of the PA was obsd. Our findings are discussed with the overarching goal of developing efficient therapeutic delivery systems.
- 57Chu-Kung, A. F.; Nguyen, R.; Bozzelli, K. N.; Tirrell, M. Chain length dependence of antimicrobial peptide-fatty acid conjugate activity. J. Colloid Interface Sci. 2010, 345 (2), 160– 167, DOI: 10.1016/j.jcis.2009.11.057Google Scholar57https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXkt1OktbY%253D&md5=1b283ed963ec56a1255b0706d3767253Chain length dependence of antimicrobial peptide-fatty acid conjugate activityChu-Kung, Alexander F.; Nguyen, Rose; Bozzelli, Kristen N.; Tirrell, MatthewJournal of Colloid and Interface Science (2010), 345 (2), 160-167CODEN: JCISA5; ISSN:0021-9797. (Elsevier B.V.)The rise of resistant bacteria has prompted the search for new antimicrobial agents. Antimicrobial membrane lytic peptides have potential as future microbial agents due to their novel mode of action. Recently conjugation of a fatty acid to antimicrobial peptides has been explored as a method to modulate the activity and selectivity of the peptide. Our work further explores these phenomena by testing two peptides, YGAAKKAAKAAKKAAKAA (AKK) and LKKLLKLLKLLKL (LKK), conjugated to fatty acids of varying length for their activity, structure, soln. assembly properties and the ability to bind model membranes. We found that increasing the length of fatty acids conjugated to peptide AKK, up to a 16 carbons in length, increases the antimicrobial activity. Peptide AKK appears to lose activity when the minimal active concn. is higher than the crit. micelle concn. (CMC) of the mol. Thus, if the CMC of the peptide conjugate is too low the activity is lost. Peptide LKK has no activity when conjugated to lauric acid and appears to aggregate at very low concns. Conjugation of AKK with a fatty acid increases its affinity to model supported lipid membranes. It appears that the increased hydrophobic interaction imparted by the fatty acid increases the affinity of the peptide to the surface thus increasing its activity. At concns. above the CMC, soln. self-assembly inhibits binding of the peptide to cell membranes.
- 58Sweatt, A. J.; Griffiths, C. D.; Groves, S. M.; Paudel, B. B.; Wang, L.; Kashatus, D. F.; Janes, K. A. Proteome-wide copy-number estimation from transcriptomics. Mol. Syst. Biol. 2024, 20 (11), 1230– 1256, DOI: 10.1038/s44320-024-00064-3Google ScholarThere is no corresponding record for this reference.
Cited By
This article has not yet been cited by other publications.
Article Views
Altmetric
Citations
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. Synthetically evolved nanopore-forming peptide pHD108. (A) The amino acid sequence of pHD108 is shown. In this work, we use both l- and d-amino acid versions of pHD108. (B) Helical wheel diagram for pHD108, showing nonpolar residues in gray, basic residues in blue, acidic residues in red, and polar residues in orange. The P14 residue, which is essential for activity, is shown in green. (C) Nanoporation activity of pHD108 in synthetic lipid vesicles made from 1-palmitoyl-2-oleoly phosphatidylcholine (POPC), versus pH. (28,30) Nanoporation is measured by the release of a 40 kDa dextran from liposomes. (D) Concentration dependence of nanoporation activity (28,30) in synthetic liposomes.
Figure 2
Figure 2. Development of an assay to measure the delivery of the PE-III protein to live cells. (A) The PE-III protein cargo is ∼25 kDa and has major and minor axes of 4.3 and 2.7 nm, respectively. (B) Fate of HeLa cells incubated with peptides, PE-III protein, or a combination. MelP5 was used at 25 μM, pHD108 was used at 200 μM, and PE-III was used at 40 nM. Cell viability was measured 48 h after incubation to allow for apoptosis to occur. Notation: l-P and d-P are l- and d-amino acid versions of pHD108. (C) Delivery of PE-III mutants is measured by Alamar Blue detection of cell viability. The R494A mutant, which is biochemically active but less stable, was tested with and without pHD108. The Δ485–492 mutant, which is biochemically inactive, was also tested with and without pHD108. (D) The PE-III delivery assay was tested on HeLa cells and RAW macrophages using 40 mM PE-III and a range of pHD108 concentrations.
Figure 3
Figure 3. Variants of pHD108 were tested in this work. Two groups of acylated peptides were synthesized. (Left) pHD108 with a C-terminal cysteine had alkyl chains attached by a disulfide cross-link. This group includes a peptide with the sulfhydryl group alkylated with acetamide and a disulfide cross-linked peptide dimer. (Right) pHD108 with acyl chains attached to the amino-terminal amine by an amide bond. This group includes unmodified l- and d-pHD108, as well as l- and d-pHD108 with C16 chains. Notation: d-P, l-P: d- and l-amino acid pHD108. CN: Linear, saturated alkyl chain with N carbons. CN- at the start of the name indicates an N-terminal acylated peptide. −CN at the end of the name indicates a C-terminal modified peptide. These images are not drawn to scale─the acyl chain sizes are exaggerated for increased visibility.
Figure 4
Figure 4. PE-III protein delivery by acylated pHD108 variants. (A, B) Delivery of PE-III by C-terminal variants (A) and N-terminal variants (B). HeLa cells were incubated with 40 nM PE-III and serially diluted pHD108 variants. Cytosolic PE-III causes apoptosis. To assay for delivery, cell viability was measured 48 h after treatment using Alamar Blue. The signal from cells with no treatment was defined as 100% viability. Media without cells was defined as 0% viability. Peptide variant notation is defined in Figure 3. ONEG is an unrelated peptide (39) used as a control. (C, D) Concentration midpoint (EC50) values for the C-terminal variants (C) and N-terminal variants (D) determined from the data in panels (A, B). (E) Direct cytotoxicity of the pHD peptide variants in the absence of the PE-III cargo measured by Alamar Blue, 48 h after treatment with peptides as above.
Figure 5
Figure 5. Cytosolic delivery of dye-labeled dextran to cells by C16-d-pHD108. HeLa cells were incubated at 37 °C overnight with 25 μM AF488-dextran 10 kDa with and without 25 μM C16-d-pHD108. Just before imaging, external AF488-dextran (green) was washed off and replaced with AF647-dextran (lavender) to mark the external spaces and cell boundaries. Cells were also treated with Hoechst 33342 (blue) to stain the nuclei. (A) Cells were incubated overnight with AF488-dextran and C16-d-pHD108. (B) The same image as in Panel A, except only the dextran fluorescence, is shown. (C) Cells were incubated overnight with AF488-dextran in the absence of C16-d-pHD108. (D) The same image as in Panel C, except only the dextran fluorescence is shown. (E) Example of normalized intensity measurements across the extracellular spaces, cytosol, and nucleus of the cell indicated by the arrow in panel B. (F) Example of normalized intensity measurements across the extracellular spaces, cytosol, and nucleus of the cells indicated by the arrow in panel D. The intensity of a 10 μM standard solution of AF488-dextran, 40% of the external concentration, is shown as a dashed line in panels (E, F).
Figure 6
Figure 6. Measurement of cytosolic delivery of AF88-labeled dextran by variants of pHD108. (A) Confocal microscopy images of HeLa cells incubated at 37 °C overnight with 0.1 μM AF488-dextran 10 kDa, plus the pHD108 variants indicated. In all cases, we used confocal images to measure the diffuse fluorescence in the nuclei and cytosol and compare it to that of a standard curve consisting of different concentrations of a reference solution containing AF488-dextran. After incubation, all cells contain bright endosomal puncta that have unreleased dextran, including masses gathered in perinuclear regions. (B) Nuclear and cytosolic fluorescence, identified by diffuse uniform intensity exclusive of bright puncta, was measured in ∼50 individual cells, shown as individual points. In the absence of peptides, cytosolic and nuclear fluorescence is negligible. By one-factor ANOVA, all peptide treatments give statistically significant increases in cytosolic fluorescence, with delivery increasing with the peptide concentration. (C) Cytosolic delivery of dextran at 4 and 37 °C, with and without C16-d-pHD108. (D) Comparison of cytosolic delivery of AF488-dextran by C16-l-pHD108 and C16-d-pHD108, measured as described above for panel B.
Figure 7
Figure 7. Cytosolic delivery of fluorescent proteins to two different cell lines by C16-d-pHD108. (A–E) HeLa cells were incubated at 37 °C overnight with 10 μM green fluorescent protein (GFP) with and without 25 μM C16-d-pHD108. (F–J): Chinese Hamster Ovary (CHO) cells were incubated overnight with 3.5 μM yellow fluorescent protein (YFP) with or without C16-d-pHD108. (A, B) GFP delivery to HeLa cells was confirmed with C16-d-pHD108. Just before imaging, the external GFP was washed off and replaced with TAMRA-dextran (red) to mark the external spaces and cell boundaries. The nuclei were stained with Hoechst 33342 (blue). Panel A shows all colors and panel B shows only GFP. (C, D) GFP delivery to HeLa cells in experiments identical to panels (A and B), except that C16-d-pHD108 was absent. Panel C shows all colors, and panel D shows only GFP. (E) GFP intensities from the nuclei of the cells in panels (A–D). Dotted lines are the intensities of standard reference solutions of 0, 2, and 4 μM GFP measured under identical conditions. (F, G) YFP delivery to CHO cells was carried out with 25 μM C16-d-pHD108. Just before imaging, the external YFP was washed off and replaced with fluorescent protein mTurquoise (mTurq) (blue) to mark the external spaces and cell boundaries. Panel F shows both colors and panel G shows only YFP. (H, I) YFP delivery to CHO cells in experiments was identical to that of panels (G and F), except that C16-d-pHD108 was absent. Panel H shows both colors and panel I shows only YFP. (J) YFP intensities from the nuclei and cytosol of the cells in panels (F–I). The dotted lines are the intensities of the standard reference solution of media only and 3.5 μM YFP measured under identical conditions.
Figure 8
Figure 8. Solution properties and cell binding of pHD108 variants. (A) Circular dichroism spectra of C16-d-pHD108 at pH 7, and of unmodified l-pHD108 at pH 5 with and without liposomes. Spectra were collected at 25 μM peptide in phosphate-buffered saline. (B) Tryptophan fluorescence emission spectra of the same three solutions are characterized in panel A. The dotted line indicates the emission maximum observed for pHD108, which is monomeric and its tryptophan residue is exposed to bulk water. (C) Particle size distribution in individually prepared 25 μM solutions of C16-d-pHD108 measured by quasielastic light scattering on a Malvern Zetasizer nano DLS instrument. Buffers contained phosphate plus 138 mM, 69 mM, or 0 mM added NaCl. (D) Light scattering intensity, in counts per second, is shown as a function of peptide concentration. (E) Solubility of pHD108 and C16-d-pHD108. Peptides were suspended by gentle vortexing for 1 min, followed by bath sonication for 5 min. After at least 1 h of incubation, solutions were sampled by reverse phase HPLC with and without centrifugation at 1400g and subsequent centrifugation at 11,000g. The peptide remaining in the solution was normalized to an uncentrifuged sample. (F) Cell binding of the pHD108 variants. HeLa cells at 106 cells/ml were suspended in phosphate-buffered saline at pH 7 or pH 5, followed by the addition of peptide dissolved in water. After 1 h of incubation, the cells were pelleted by centrifugation and the peptide remaining in the solution was measured by HPLC. (G) Confocal microscopy images of HeLa cells incubated with 5 μM unlabeled C16-d-D-pHD108 mixed with 1 μM C16-d-pHD108 labeled with TAMRA. All images were collected with the same instrument settings and the same peptide concentrations. In the left panel, cells were incubated with peptide in media at pH 7 for 10 min. The other panels show one field of cells under the same conditions after the solution pH was reduced to pH 5. Cells are also stained with the DNA-binding dye Hoechst 33342.
References
This article references 58 other publications.
- 1Hong, C. W.; Zeng, Q. Tapping the treasure of intracellular oncotargets with immunotherapy. FEBS Lett. 2014, 588 (2), 350– 355, DOI: 10.1016/j.febslet.2013.10.025There is no corresponding record for this reference.
- 2Cheever, M. A.; Allison, J. P.; Ferris, A. S.; Finn, O. J.; Hastings, B. M.; Hecht, T. T.; Mellman, I.; Prindiville, S. A.; Viner, J. L.; Weiner, L. M.; Matrisian, L. M. The prioritization of cancer antigens: a national cancer institute pilot project for the acceleration of translational research. Clin. Cancer Res. 2009, 15 (17), 5323– 5337, DOI: 10.1158/1078-0432.CCR-09-0737There is no corresponding record for this reference.
- 3Lazo, J. S.; Sharlow, E. R. Drugging Undruggable Molecular Cancer Targets. Annu. Rev. Pharmacol. Toxicol. 2016, 56, 23– 40, DOI: 10.1146/annurev-pharmtox-010715-1034403https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXhslGlsr7O&md5=5ab0518d4155a00e2a898a6f68d551d8Drugging Undruggable Molecular Cancer TargetsLazo, John S.; Sharlow, Elizabeth R.Annual Review of Pharmacology and Toxicology (2016), 56 (), 23-40CODEN: ARPTDI; ISSN:0362-1642. (Annual Reviews)Cancer, more than any other human disease, now has a surfeit of potential mol. targets poised for therapeutic exploitation. Currently, a no. of attractive and validated cancer targets remain outside of the reach of pharmacol. regulation. Some have been described as undruggable, at least by traditional strategies. In this article, we outline the basis for the undruggable moniker, propose a reclassification of these targets as undrugged, and highlight three general classes of this imposing group as exemplars with some attendant strategies currently being explored to reclassify them. Expanding the spectrum of disease-relevant targets to pharmacol. manipulation is central to reducing cancer morbidity and mortality.
- 4Slastnikova, T. A.; Ulasov, A. V.; Rosenkranz, A. A.; Sobolev, A. S. Targeted Intracellular Delivery of Antibodies: The State of the Art. Front. Pharmacol. 2018, 9, 1208, DOI: 10.3389/fphar.2018.012084https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXhtVGmtbvL&md5=fe66302eb7d6dd1e77f41c46dcffc635Targeted intracellular delivery of antibodies: the state of the artSlastnikova, Tatiana A.; Ulasov, A. V.; Rosenkranz, A. A.; Sobolev, A. S.Frontiers in Pharmacology (2018), 9 (), 1208CODEN: FPRHAU; ISSN:1663-9812. (Frontiers Media S.A.)A review. A dominant area of antibody research is the extension of the use of this mighty exptl. and therapeutic tool for the specific detection of mols. for diagnostics, visualization, and activity blocking. Despite the ability to raise antibodies against different proteins, numerous applications of antibodies in basic research fields, clin. practice, and biotechnol. are restricted to permeabilized cells or extracellular antigens, such as membrane or secreted proteins. With the exception of small groups of autoantibodies, natural antibodies to intracellular targets cannot be used within living cells. This excludes the scope of a major class of intracellular targets, including some infamous cancer-assocd. mols. Some of these targets are still not druggable via small mols. because of large flat contact areas and the absence of deep hydrophobic pockets in which small mols. can insert and perturb their activity. Thus, the development of technologies for the targeted intracellular delivery of antibodies, their fragments, or antibody-like mols. is extremely important. Various strategies for intracellular targeting of antibodies via protein-transduction domains or their mimics, liposomes, polymer vesicles, and viral envelopes, are reviewed in this article. The pitfalls, challenges, and perspectives of these technologies are discussed.
- 5Chonghaile, T. N.; Letai, A. Mimicking the BH3 domain to kill cancer cells. Oncogene 2008, 27 (Suppl 1), S149– S157, DOI: 10.1038/onc.2009.52There is no corresponding record for this reference.
- 6Goldsmith, K. C.; Liu, X.; Dam, V.; Morgan, B. T.; Shabbout, M.; Cnaan, A.; Letai, A.; Korsmeyer, S. J.; Hogarty, M. D. BH3 peptidomimetics potently activate apoptosis and demonstrate single agent efficacy in neuroblastoma. Oncogene 2006, 25 (33), 4525– 4533, DOI: 10.1038/sj.onc.12094896https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28Xnsl2jsbs%253D&md5=2447d02a4be4f41fcf2939602b149948BH3 peptidomimetics potently activate apoptosis and demonstrate single agent efficacy in neuroblastomaGoldsmith, K. C.; Liu, X.; Dam, V.; Morgan, B. T.; Shabbout, M.; Cnaan, A.; Letai, A.; Korsmeyer, S. J.; Hogarty, M. D.Oncogene (2006), 25 (33), 4525-4533CODEN: ONCNES; ISSN:0950-9232. (Nature Publishing Group)The major impediment to cure for many malignancies is the development of therapy resistance with resultant tumor progression. Genetic alterations leading to subversion of inherent apoptosis pathways are common themes in therapy resistance. Bcl-2 family proteins play a crit. role in regulating mitochondrial apoptosis that governs chemotherapeutic effects, and defective engagement of these pathways contributes to treatment failure. The authors have studied the efficacy of BH3 peptidomimetics consisting of the minimal death, or BH3, domains of the proapoptotic BH3-only proteins Bid and Bad to induce apoptosis using neuroblastoma (NB) as a model system. The authors demonstrate that BH3 peptides, modified with an arginine homopolymer for membrane transduction (called r8-BidBH3 and r8-BadBH3, resp.), potently induce apoptosis in NB cells, including those with MYCN amplification. Cell death is caspase 9 dependent, consistent with a requirement for the intrinsic mitochondrial pathway. Substitutions at highly conserved residues within the r8-BidBH3 peptide abolish apoptotic efficacy supporting activity through specific BH domain interactions. Concomitant exposure to r8-BadBH3 and r8-BidBH3 at sublethal monotherapy doses revealed potent synergy consistent with a competitive displacement model, whereby BH3 peptides displace sequestered BH3 proteins to induce cell death. Further, BH3 peptides demonstrate antitumor efficacy in a xenograft model of NB in the absence of addnl. genotoxic or trophic stressors. These data provide proof of principle that targeted reengagement of apoptosis pathways may be of therapeutic utility, and BH3-like compds. are attractive lead agents to reestablish therapy-induced apoptosis in refractory malignancies.
- 7Walensky, L. D.; Kung, A. L.; Escher, I.; Malia, T. J.; Barbuto, S.; Wright, R. D.; Wagner, G.; Verdine, G. L.; Korsmeyer, S. J. Activation of apoptosis in vivo by a hydrocarbon-stapled BH3 helix. Science 2004, 305 (5689), 1466– 1470, DOI: 10.1126/science.10991917https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXntFart7c%253D&md5=53fca0524df7a584ccf9d0991bc1bb64Activation of Apoptosis in Vivo by a Hydrocarbon-Stapled BH3 HelixWalensky, Loren D.; Kung, Andrew L.; Escher, Iris; Malia, Thomas J.; Barbuto, Scott; Wright, Renee D.; Wagner, Gerhard; Verdine, Gregory L.; Korsmeyer, Stanley J.Science (Washington, DC, United States) (2004), 305 (5689), 1466-1470CODEN: SCIEAS; ISSN:0036-8075. (American Association for the Advancement of Science)BCL-2 family proteins constitute a crit. control point for the regulation of apoptosis. Protein interaction between BCL-2 members is a prominent mechanism of control and is mediated through the amphipathic α-helical BH3 segment, an essential death domain. We used a chem. strategy, termed hydrocarbon stapling, to generate BH3 peptides with improved pharmacol. properties. The stapled peptides, called "stabilized alpha-helix of BCL-2 domains" (SAHBs), proved to be helical, protease-resistant, and cell-permeable mols. that bound with increased affinity to multidomain BCL-2 member pockets. A SAHB of the BH3 domain from the BID protein specifically activated the apoptotic pathway to kill leukemia cells. In addn., SAHB effectively inhibited the growth of human leukemia xenografts in vivo. Hydrocarbon stapling of native peptides may provide a useful strategy for exptl. and therapeutic modulation of protein-protein interactions in many signaling pathways.
- 8Baar, M. P.; Brandt, R. M. C.; Putavet, D. A.; Klein, J. D. D.; Derks, K. W. J.; Bourgeois, B. R. M.; Stryeck, S.; Rijksen, Y.; van Willigenburg, H.; Feijtel, D. A. Targeted Apoptosis of Senescent Cells Restores Tissue Homeostasis in Response to Chemotoxicity and Aging. Cell 2017, 169 (1), 132– 147.e116, DOI: 10.1016/j.cell.2017.02.0318https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2sXltVWnsrk%253D&md5=5b907eb349b0e3014186f56893af362eTargeted Apoptosis of Senescent Cells Restores Tissue Homeostasis in Response to Chemotoxicity and AgingBaar, Marjolein P.; Brandt, Renata M. C.; Putavet, Diana A.; Klein, Julian D. D.; Derks, Kasper W. J.; Bourgeois, Benjamin R. M.; Stryeck, Sarah; Rijksen, Yvonne; van Willigenburg, Hester; Feijtel, Danny A.; van der Pluijm, Ingrid; Essers, Jeroen; van Cappellen, Wiggert A.; van IJcken, Wilfred F.; Houtsmuller, Adriaan B.; Pothof, Joris; de Bruin, Ron W. F.; Madl, Tobias; Hoeijmakers, Jan H. J.; Campisi, Judith; de Keizer, Peter L. J.Cell (Cambridge, MA, United States) (2017), 169 (1), 132-147.e16CODEN: CELLB5; ISSN:0092-8674. (Cell Press)The accumulation of irreparable cellular damage restricts health span after acute stress or natural aging. Senescent cells are thought to impair tissue function, and their genetic clearance can delay features of aging. Identifying how senescent cells avoid apoptosis allows for the prospective design of anti-senescence compds. to address whether homeostasis can also be restored. Here, the authors identify FOXO4 as a pivot in senescent cell viability. The authors designed a FOXO4 peptide that perturbs the FOXO4 interaction with p53. In senescent cells, this selectively causes p53 nuclear exclusion and cell-intrinsic apoptosis. Under conditions where it was well tolerated in vivo, this FOXO4 peptide neutralized doxorubicin-induced chemotoxicity. Moreover, it restored fitness, fur d., and renal function in both fast aging XpdTTD/TTD and naturally aged mice. Thus, therapeutic targeting of senescent cells is feasible under conditions where loss of health has already occurred, and in doing so tissue homeostasis can effectively be restored.
- 9Bobone, S.; Pannone, L.; Biondi, B.; Solman, M.; Flex, E.; Canale, V. C.; Calligari, P.; De Faveri, C.; Gandini, T.; Quercioli, A. Targeting Oncogenic Src Homology 2 Domain-Containing Phosphatase 2 (SHP2) by Inhibiting Its Protein-Protein Interactions. J. Med. Chem. 2021, 64 (21), 15973– 15990, DOI: 10.1021/acs.jmedchem.1c01371There is no corresponding record for this reference.
- 10Zottel, A.; Novak, M.; Samec, N.; Majc, B.; Colja, S.; Katrasnik, M.; Vittori, M.; Hrastar, B.; Rotter, A.; Porcnik, A. Anti-Vimentin Nanobody Decreases Glioblastoma Cell Invasion In Vitro and In Vivo. Cancers 2023, 15 (3), 573, DOI: 10.3390/cancers15030573There is no corresponding record for this reference.
- 11Deprey, K.; Becker, L.; Kritzer, J.; Pluckthun, A. Trapped! A Critical Evaluation of Methods for Measuring Total Cellular Uptake versus Cytosolic Localization. Bioconjugate Chem. 2019, 30 (4), 1006– 1027, DOI: 10.1021/acs.bioconjchem.9b0011211https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXkvFWjtrw%253D&md5=3d134d4d06429a27f0561329228ac2ecTrapped! A Critical Evaluation of Methods for Measuring Total Cellular Uptake versus Cytosolic LocalizationDeprey, Kirsten; Becker, Lukas; Kritzer, Joshua; Pluckthun, AndreasBioconjugate Chemistry (2019), 30 (4), 1006-1027CODEN: BCCHES; ISSN:1043-1802. (American Chemical Society)Biomols. have many properties that make them promising for intracellular therapeutic applications, but delivery remains a key challenge because large biomols. cannot easily enter the cytosol. Furthermore, quantification of total intracellular vs. cytosolic concns. remains demanding, and the detn. of delivery efficiency is thus not straightforward. In this review, we discuss strategies for delivering biomols. into the cytosol and briefly summarize the mechanisms of uptake for these systems. We then describe commonly used methods to measure total cellular uptake and, more selectively, cytosolic localization, and discuss the major advantages and drawbacks of each method. We critically evaluate methods of measuring "cell penetration" that do not adequately distinguish total cellular uptake and cytosolic localization, which often lead to inaccurate interpretations of a mol.'s cytosolic localization. Finally, we summarize the properties and components of each method, including the main caveats of each, to allow for informed decisions about method selection for specific applications. When applied correctly and interpreted carefully, methods for quantifying cytosolic localization offer valuable insight into the bioactivity of biomols. and potentially the prospects for their eventual development into therapeutics.
- 12Schneider, A. F. L.; Kithil, M.; Cardoso, M. C.; Lehmann, M.; Hackenberger, C. P. R. Cellular uptake of large biomolecules enabled by cell-surface-reactive cell-penetrating peptide additives. Nat. Chem. 2021, 13 (6), 530– 539, DOI: 10.1038/s41557-021-00661-x12https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXptVWntL4%253D&md5=573ed27e9ca9ba324141e1d4761b9e8dCellular uptake of large biomolecules enabled by cell-surface-reactive cell-penetrating peptide additivesSchneider, Anselm F. L.; Kithil, Marina; Cardoso, M. Cristina; Lehmann, Martin; Hackenberger, Christian P. R.Nature Chemistry (2021), 13 (6), 530-539CODEN: NCAHBB; ISSN:1755-4330. (Nature Portfolio)Abstr.: Enabling the cellular delivery and cytosolic bioavailability of functional proteins constitutes a major challenge for the life sciences. Here we demonstrate that thiol-reactive arginine-rich peptide additives can enhance the cellular uptake of protein-CPP conjugates in a non-endocytic mode, even at low micromolar concn. We show that such thiol- or HaloTag-reactive additives can result in covalently anchored CPPs on the cell surface, which are highly effective at co-delivering protein cargoes. Taking advantage of the thiol reactivity of our most effective CPP additive, we show that Cys-contg. proteins can be readily delivered into the cytosol by simple co-addn. of a slight excess of this CPP. Furthermore, we demonstrate the application of our 'CPP-additive technique' in the delivery of functional enzymes, nanobodies and full-length Ig-G antibodies. This new cellular uptake protocol greatly simplifies both the accessibility and efficiency of protein and antibody delivery, with minimal chem. or genetic engineering.
- 13Kauffman, W. B.; Guha, S.; Wimley, W. C. Synthetic molecular evolution of hybrid cell penetrating peptides. Nat. Commun. 2018, 9 (1), 2568 DOI: 10.1038/s41467-018-04874-613https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BB3c%252Fkt1Kktg%253D%253D&md5=50a72964b3069b3d25abfb5967f7b8daSynthetic molecular evolution of hybrid cell penetrating peptidesKauffman W Berkeley; Guha Shantanu; Wimley William CNature communications (2018), 9 (1), 2568 ISSN:.Peptides and analogs such as peptide nucleic acids (PNA) are promising tools and therapeutics, but the cell membrane remains a barrier to intracellular targets. Conjugation to classical cell penetrating peptides (CPPs) such as pTat48-60 (tat) and pAntp43-68 (penetratin) facilitates delivery; however, efficiencies are low. Lack of explicit design principles hinders rational improvement. Here, we use synthetic molecular evolution (SME) to identify gain-of-function CPPs with dramatically improved ability to deliver cargoes to cells at low concentration. A CPP library containing 8192 tat/penetratin hybrid peptides coupled to an 18-residue PNA is screened using the HeLa pTRE-LucIVS2 splice correction reporter system. The daughter CPPs identified are one to two orders of magnitude more efficient than the parent sequences at delivery of PNA, and also deliver a dye cargo and an anionic peptide cargo. The significant increase in performance following a single iteration of SME demonstrates the power of this approach to peptide sequence optimization.
- 14Kauffman, W. B.; Fuselier, T.; He, J.; WC, W. Mechanism matters: A taxonomy of cell penetrating peptides. Trends Biochem. Sci. 2015, 40, 739– 764, DOI: 10.1016/j.tibs.2015.10.004There is no corresponding record for this reference.
- 15Bartoš, L.; Vacha, R. Peptide translocation across asymmetric phospholipid membranes. Biophys. J. 2024, 123, 693– 702, DOI: 10.1016/j.bpj.2024.02.006There is no corresponding record for this reference.
- 16Bus, T.; Traeger, A.; Schubert, U. S. The great escape: how cationic polyplexes overcome the endosomal barrier. J. Mater. Chem. B 2018, 6 (43), 6904– 6918, DOI: 10.1039/C8TB00967H16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXhslSrsLfJ&md5=62d63fe76a4b75545afeaa170bffce43The great escape: how cationic polyplexes overcome the endosomal barrierBus, Tanja; Traeger, Anja; Schubert, Ulrich S.Journal of Materials Chemistry B: Materials for Biology and Medicine (2018), 6 (43), 6904-6918CODEN: JMCBDV; ISSN:2050-7518. (Royal Society of Chemistry)A review. The targeted and efficiency-oriented delivery of (therapeutic) nucleic acids raises hope for successful gene therapy, i.e., for the local and individual treatment of acquired and inherited genetic disorders. Despite promising achievements in the field of polymer-mediated gene delivery, the efficiency of the non-viral vectors remains orders of magnitude lower than viral-mediated ones. Several obstacles on the mol. and cellular level along the gene delivery process were identified, starting from the design and formulation of the nano-sized carriers up to the targeted release to their site of action. In particular, the efficient escape from endo-lysosomal compartments was demonstrated to be a major barrier and its exact mechanism still remains unclear. Different hypotheses and theories of the endosomal escape were postulated. The most popular one is the so-called "proton sponge" hypothesis, claiming an escape by rupture of the endosome through osmotic swelling. It was the first effort to explain the excellent transfection efficiency of poly(ethylene imine). Moreover, it was thought that a unique mechanism based on the ability to capture protons and to buffer the endosomal pH is the basis of endosomal escape. Recent theories deal with the direct interaction of the cationic polyplex or free polymer with the exoplasmic lipid leaflet causing membrane destabilization, permeability or polymer-supported nanoscale hole formation. Both escape strategies are more related to viral-mediated escape compared to the "proton sponge" effect. This review addresses the different endosomal release theories and highlights their key mechanism.
- 17Moulay, G.; Leborgne, C.; Mason, A. J.; Aisenbrey, C.; Kichler, A.; Bechinger, B. Histidine-rich designer peptides of the LAH4 family promote cell delivery of a multitude of cargo. J. Peptide Sci. 2017, 23 (4), 320– 328, DOI: 10.1002/psc.2955There is no corresponding record for this reference.
- 18Langel, U. CPP, Cell Penetrating Peptides; Springer Nature, 2019.There is no corresponding record for this reference.
- 19Wimley, W. C. Describing the mechanism of antimicrobial peptide action with the interfacial activity model. ACS Chem. Biol. 2010, 5 (10), 905– 917, DOI: 10.1021/cb100155819https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXhtVGgsLbF&md5=975166f203aa8a570841091eb8501beeDescribing the Mechanism of Antimicrobial Peptide Action with the Interfacial Activity ModelWimley, William C.ACS Chemical Biology (2010), 5 (10), 905-917CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)A review. Antimicrobial peptides (AMPs) have been studied for three decades, and yet a mol. understanding of their mechanism of action is still lacking. Here we summarize current knowledge for both synthetic vesicle expts. and microbe expts., with a focus on comparisons between the two. Microbial expts. are done at peptide to lipid ratios that are at least 4 orders of magnitude higher than vesicle-based expts. To close the gap between the two concn. regimes, we propose an "interfacial activity model", which is based on an exptl. testable mol. image of AMP-membrane interactions. The interfacial activity model may be useful in driving engineering and design of novel AMPs.
- 20Marschall, A. L.; Zhang, C.; Frenzel, A.; Schirrmann, T.; Hust, M.; Perez, F.; Dubel, S. Delivery of antibodies to the cytosol: debunking the myths. MAbs 2014, 6 (4), 943– 956, DOI: 10.4161/mabs.29268There is no corresponding record for this reference.
- 21Wimley, W. C. Synthetic Molecular Evolution of Cell Penetrating Peptides. Methods Mol. Biol. 2022, 2383, 73– 89, DOI: 10.1007/978-1-0716-1752-6_5There is no corresponding record for this reference.
- 22Starr, C. G.; Ghimire, J.; Guha, S.; Hoffmann, J. P.; Wang, Y.; Sun, L.; Landreneau, B. N.; Kolansky, Z. D.; Kilanowski-Doroh, I. M.; Sammarco, M. C. Synthetic molecular evolution of host cell-compatible, antimicrobial peptides effective against drug-resistant, biofilm-forming bacteria. Proc. Natl. Acad. Sci. U.S.A. 2020, 117 (15), 8437– 8448, DOI: 10.1073/pnas.1918427117There is no corresponding record for this reference.
- 23Li, S.; Kim, S. Y.; Pittman, A. E.; King, G. M.; Wimley, W. C.; Hristova, K. Potent Macromolecule-Sized Poration of Lipid Bilayers by the Macrolittins, A Synthetically Evolved Family of Pore-Forming Peptides. J. Am. Chem. Soc. 2018, 140 (20), 6441– 6447, DOI: 10.1021/jacs.8b0302623https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1cXosVWitrs%253D&md5=f2f6ba0fc30294d7e94b957db4fc856fPotent Macromolecule-Sized Poration of Lipid Bilayers by the Macrolittins, A Synthetically Evolved Family of Pore-Forming PeptidesLi, Sijia; Kim, Sarah Y.; Pittman, Anna E.; King, Gavin M.; Wimley, William C.; Hristova, KalinaJournal of the American Chemical Society (2018), 140 (20), 6441-6447CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Pore-forming peptides with novel functions have potential utility in many biotechnol. applications. However, the sequence-structure-function relationships of pore forming peptides are not understood well enough to empower rational design. Therefore, in this work we used synthetic mol. evolution to identify a novel family of peptides that are highly potent and cause macromol. poration in synthetic lipid vesicles at low peptide concn. and at neutral pH. These unique 26-residue peptides, which we call macrolittins, release macromols. from lipid bilayer vesicles made from zwitterionic PC lipids at peptide to lipid ratios as low as 1:1000, a property that is almost unprecedented among known membrane permeabilizing peptides. The macrolittins exist as membrane-spanning α-helixes. They cause dramatic bilayer thinning and form large pores in planar supported bilayers. The high potency of these peptides is likely due to their ability to stabilize bilayer edges by a process that requires specific electrostatic interactions between peptides.
- 24Krauson, A. J.; He, J.; Hoffmann, A. R.; Wimley, A. W.; Wimley, W. C. Synthetic molecular evolution of pore-forming peptides by Iterative combinatorial library screening. ACS Chem. Biol. 2013, 8, 823– 831, DOI: 10.1021/cb300598k24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXit1Squrw%253D&md5=f0999c8059edf784b706174f62aab14aSynthetic Molecular Evolution of Pore-Forming Peptides by Iterative Combinatorial Library ScreeningKrauson, Aram J.; He, Jing; Wimley, Andrew W.; Hoffmann, Andrew R.; Wimley, William C.ACS Chemical Biology (2013), 8 (4), 823-831CODEN: ACBCCT; ISSN:1554-8929. (American Chemical Society)We previously reported the de novo design of a combinatorial peptide library that was subjected to high-throughput screening to identify membrane-permeabilizing antimicrobial peptides that have β-sheet-like secondary structure. Those peptides do not form discrete pores in membranes but instead partition into membrane interfaces and cause transient permeabilization by membrane disruption, but only when present at high concn. In this work, we used a consensus sequence from that initial screen as a template to design an iterative, second generation library. In the 24-26-residue, 16,200-member second generation library we varied six residues. Two diad repeat motifs of alternating polar and nonpolar amino acids were preserved to maintain a propensity for non-helical secondary structure. We used a new high-throughput assay to identify members that self- assemble into equil. pores in synthetic lipid bilayers. This screen was done at a very stringent peptide to lipid ratio of 1:1000 where most known membrane-permeabilizing peptides, including the template peptide, are not active. In a screen of 10,000 library members we identified 16 (∼0.2%) that are equil. pore-formers at this high stringency. These rare and highly active peptides, which share a common sequence motif, are as potent as the most active pore-forming peptides known. Furthermore, they are not α-helical, which makes them unusual, as most of the highly potent pore-forming peptides are amphipathic α-helixes. Here we demonstrate that this synthetic mol. evolution-based approach, taken together with the new high-throughput tools we have developed, enables the identification, refinement, and optimization of unique membrane active peptides.
- 25Tosteson, M. T.; Tosteson, D. C. The sting. Melittin forms channels in lipid bilayers. Biophys. J. 1981, 36 (1), 109– 116, DOI: 10.1016/S0006-3495(81)84719-4There is no corresponding record for this reference.
- 26Krauson, A. J.; He, J.; Wimley, W. C. Gain-of-Function Analogues of the Pore-Forming Peptide Melittin Selected by Orthogonal High-Throughput Screening. J. Am. Chem. Soc. 2012, 134 (30), 12732– 12741, DOI: 10.1021/ja304200426https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XptF2hsrY%253D&md5=e15fc7f6b1869548ec0154cbcc24c686Gain-of-function analogues of the pore-forming peptide melittin selected by orthogonal high-throughput screeningKrauson, Aram J.; He, Jing; Wimley, William C.Journal of the American Chemical Society (2012), 134 (30), 12732-12741CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)The authors recently developed an orthogonal, high-throughput assay to identify peptides that self-assemble into potent, equil. pores in synthetic lipid bilayers. Here, the authors use this assay as a high-throughput screen to select highly potent pore-forming peptides from a 7776-member rational combinatorial peptide library based on the sequence of the natural pore-forming peptide toxin melittin. In the library the authors varied ten crit. residues in the melittin sequence, chosen to test specific structural hypotheses about the mechanism of pore formation. Using the new high-throughput assay, the authors screened the library for gain-of-function sequences at a peptide to lipid ratio of 1:1000 where native melittin is not active. More than 99% of the library sequences were also inactive under these conditions. A small no. of library members (0.1%) were highly active. From these the authors identified 14 potent, gain-of-function, pore-forming sequences. These sequences differed from melittin in only 2-6 amino acids out of 26. Some native residues were highly conserved and others were consistently changed. The two factors that were essential for gain-of-function were the preservation of melittin's proline-dependent break in the middle of the helix and the improvement and extension the amphipathic nature of the α-helix. In particular the highly cationic carboxyl-terminal sequence of melittin is consistently changed in the gain-of-function variants to a sequence that it is capable of participating in an extended amphipathic α-helix. The most potent variants reside in a membrane-spanning orientation, in contrast to the parent melittin, which is predominantly surface bound. This structural information, taken together with the high-throughput tools developed for this work, enable the identification, refinement and optimization of pore-forming peptides for many potential applications.
- 27Wiedman, G.; Fuselier, T.; He, J.; Searson, P. C.; Hristova, K.; Wimley, W. C. Highly efficient macromolecule-sized poration of lipid bilayers by a synthetically evolved peptide. J. Am. Chem. Soc. 2014, 136 (12), 4724– 4731, DOI: 10.1021/ja500462s27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXjtlOlsbg%253D&md5=5accca4a4bc0e23542e62683ed1e8056Highly Efficient Macromolecule-Sized Poration of Lipid Bilayers by a Synthetically Evolved PeptideWiedman, Gregory; Fuselier, Taylor; He, Jing; Searson, Peter C.; Hristova, Kalina; Wimley, William C.Journal of the American Chemical Society (2014), 136 (12), 4724-4731CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Peptides that self-assemble, at low concn., into bilayer-spanning pores which allow the passage of macromols. would be beneficial in multiple areas of biotechnol. However, there are few, if any, natural or designed peptides that have this property. The 26-residue peptide "MelP5", a synthetically evolved gain-of-function variant of the bee venom lytic peptide melittin identified in a high-throughput screen for small mol. leakage, enables the passage of macromols. across bilayers under conditions where melittin and other pore-forming peptides do not. In surface-supported bilayers, MelP5 forms unusually high conductance, equil. pores at peptide:lipid ratios as low as 1:25000. The increase in bilayer conductance due to MelP5 is dramatically higher, per peptide, than the increase due to the parent sequence of melittin or other peptide pore formers. Here the authors also develop two novel assays for macromol. leakage from vesicles, and they use them to characterize MelP5 pores in bilayers. MelP5 allows the passage of macromols. across vesicle membranes at peptide:lipid ratios as low as 1:500, and under conditions where neither osmotic lysis nor gross vesicle destabilization occur. The macromol.-sized, equil. pores formed by MelP5 are unique as neither melittin nor other pore-forming peptides release macromols. significantly under the same conditions. MelP5 thus appears to belong to a novel functional class of peptide that could form the foundation of multiple potential biotechnol. applications.
- 28Wiedman, G.; Kim, S. Y.; Zapata-Mercado, E.; Wimley, W. C.; Hristova, K. PH-Triggered, Macromolecule-Sized Poration of Lipid Bilayers by Synthetically Evolved Peptides. J. Am. Chem. Soc. 2017, 139, 937– 945, DOI: 10.1021/jacs.6b1144728https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XitFaiurjN&md5=22f0314dfb54a347cf980012204fa247pH-Triggered, Macromolecule-Sized Poration of Lipid Bilayers by Synthetically Evolved PeptidesWiedman, Gregory; Kim, Sarah Y.; Zapata-Mercado, Elmer; Wimley, William C.; Hristova, KalinaJournal of the American Chemical Society (2017), 139 (2), 937-945CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)PH-triggered membrane-permeabilizing peptides could be exploited in a variety of applications, such as to enable cargo release from endosomes for cellular delivery, or as cancer therapeutics that selectively permeabilize the plasma membranes of malignant cells. Such peptides would be esp. useful if they could enable the movement of macromols. across membranes, a rare property in membrane-permeabilizing peptides. Here the authors approach this goal by using an orthogonal high-throughput screen of an iterative peptide library to identify peptide sequences that have the following two properties: (1) little synthetic lipid membrane permeabilization at physiol. pH 7 at high peptide concn. and (2) efficient formation of macromol.-sized defects in synthetic lipid membranes at acidic pH 5 and low peptide concn. The peptides the authors selected are remarkably potent macromol. sized pore-formers at pH 5, while having little or no activity at pH 7, as intended. The action of these peptides likely relies on tight coupling between membrane partitioning, α-helix formation, and electrostatic repulsions between acidic side chains, which collectively drive a sharp pH-triggered transition between inactive and active configurations with apparent pKa values of 5.5-5.8. This work opens new doors to developing applications that use peptides with membrane-permeabilizing activities that are triggered by physiol. relevant decreases in pH.
- 29Kim, S. Y.; Bondar, A. N.; Wimley, W. C.; Hristova, K. pH-triggered pore-forming peptides with strong composition-dependent membrane selectivity. Biophys. J. 2021, 120 (4), 618– 630, DOI: 10.1016/j.bpj.2021.01.01029https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXisVyktbc%253D&md5=80fdd94720a95e5d9c15ffd1185137fcpH-triggered pore-forming peptides with strong composition-dependent membrane selectivityKim, Sarah Y.; Bondar, Ana-Nicoleta; Wimley, William C.; Hristova, KalinaBiophysical Journal (2021), 120 (4), 618-630CODEN: BIOJAU; ISSN:0006-3495. (Cell Press)Peptides that self-assemble into nanometer-sized pores in lipid bilayers could have utility in a variety of biotechnol. and clin. applications if we can understand their phys. chem. properties and learn to control their membrane selectivity. To empower such control, we have used synthetic mol. evolution to identify the pH-dependent delivery peptides, a family of peptides that assemble into macromol.-sized pores in membranes at low peptide concn. but only at pH < ∼6. Further advancements will also require better selectivity for specific membranes. Here, we det. the effect of anionic headgroups and bilayer thickness on the mechanism of action of the pH-dependent delivery peptides by measuring binding, secondary structure, and macromol. poration. The peptide pHD15 partitions and folds equally well into zwitterionic and anionic membranes but is less potent at pore formation in phosphatidylserine-contg. membranes. The peptide also binds and folds similarly in membranes of various thicknesses, but its ability to release macromols. changes dramatically. It causes potent macromol. poration in vesicles made from phosphatidylcholine with 14 carbon acyl chains, but macromol. poration decreases sharply with increasing bilayer thickness and does not occur at any peptide concn. in fluid bilayers made from phosphatidylcholine lipids with 20-carbon acyl chains. The effects of headgroup and bilayer thickness on macromol. poration cannot be accounted for by the amt. of peptide bound but instead reflect an inherent selectivity of the peptide for inserting into the membrane-spanning pore state. Mol. dynamics simulations suggest that the effect of thickness is due to hydrophobic match/mismatch between the membrane-spanning peptide and the bilayer hydrocarbon. This remarkable degree of selectivity based on headgroup and esp. bilayer thickness is unusual and suggests ways that pore-forming peptides with exquisite selectivity for specific membranes can be designed or evolved.
- 30Kim, S. Y.; Pittman, A. E.; Zapata-Mercado, E.; King, G. M.; Wimley, W. C.; Hristova, K. Mechanism of Action of Peptides That Cause the pH-Triggered Macromolecular Poration of Lipid Bilayers. J. Am. Chem. Soc. 2019, 141 (16), 6706– 6718, DOI: 10.1021/jacs.9b0197030https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXmtFGntbs%253D&md5=2e292bc579d5cd82b71f412ba386acd1Mechanism of action of peptides that cause the pH-triggered macromolecular poration of lipid bilayersKim, Sarah Y.; Pittman, Anna E.; Zapata-Mercado, Elmer; King, Gavin M.; Wimley, William C.; Hristova, KalinaJournal of the American Chemical Society (2019), 141 (16), 6706-6718CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Using synthetic mol. evolution, we previously discovered a family of peptides that cause macromol. poration in synthetic membranes at low peptide concn. in a way that is triggered by acidic pH. To understand the mechanism of action of these "pHD peptides", here we systematically explored structure-function relationships through measurements of the effect of pH and peptide concn. on membrane binding, peptide structure, and the formation of macromol.-sized pores in membranes. Both AFM and functional assays demonstrate the peptide-induced appearance of large pores in bilayers. Pore formation has a very steep pH dependence and is also dependent on peptide concn. In vesicles, 50% leakage of 40 kDa dextrans occurs at 1 bound peptide per 1300 lipids or only 75 peptides per vesicle, an observation that holds true across a wide range of acidic pH values. The major role of pH is to regulate the amt. of peptide bound per vesicle. The phys. chem. and sequence of the pHD peptides affect their potency and pH dependence; therefore, the sequence-structure-function relationships described here can be used for the future design and optimization of membrane permeabilizing peptides for specific applications.
- 31Wiedman, G.; Wimley, W. C.; Hristova, K. Testing the limits of rational design by engineering pH sensitivity into membrane-active peptides. Biochim. Biophys. Acta 2015, 1848 (4), 951– 957, DOI: 10.1016/j.bbamem.2014.12.023There is no corresponding record for this reference.
- 32Guha, S.; Ferrie, R. P.; Ghimire, J.; Ventura, C. R.; Wu, E.; Sun, L.; Kim, S. Y.; Wiedman, G. R.; Hristova, K.; Wimley, W. C. Applications and evolution of melittin, the quintessential membrane active peptide. Biochem. Pharmacol. 2021, 193, 114769 DOI: 10.1016/j.bcp.2021.11476932https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BB3MXitFGhtrvI&md5=e16c4e779ab73131267b2942d2aa5d4eApplications and evolution of melittin, the quintessential membrane active peptideGuha, Shantanu; Ferrie, Ryan P.; Ghimire, Jenisha; Ventura, Cristina R.; Wu, Eric; Sun, Leisheng; Kim, Sarah Y.; Wiedman, Gregory R.; Hristova, Kalina; Wimley, Wimley C.Biochemical Pharmacology (Amsterdam, Netherlands) (2021), 193 (), 114769CODEN: BCPCA6; ISSN:0006-2952. (Elsevier B.V.)A review. Melittin, the main venom component of the European Honeybee, is a cationic linear peptide-amide of 26 amino acid residues with the sequence: GIGAVLKVLTTGLPALISWIKRKRQQ-NH2. Melittin binds to lipid bilayer membranes, folds into amphipathic α-helical secondary structure and disrupts the permeability barrier. Since melittin was first described, a remarkable array of activities and potential applications in biol. and medicine have been described. Melittin is also a favorite model system for biophysicists to study the structure, folding and function of peptides and proteins in membranes. Melittin has also been used as a template for the evolution of new activities in membranes. Here we overview the rich history of scientific research into the many activities of melittin and outline exciting future applications.
- 33Guha, S.; Ghimire, J.; Wu, E.; Wimley, W. C. Mechanistic Landscape of Membrane-Permeabilizing Peptides. Chem. Rev. 2019, 119, 6040– 6085, DOI: 10.1021/acs.chemrev.8b0052033https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC1MXltV2jug%253D%253D&md5=a591e43ebc009cbe483ba932a2b93feeMechanistic landscape of membrane-permeabilizing peptidesGuha, Shantanu; Ghimire, Jenisha; Wu, Eric; Wimley, William C.Chemical Reviews (Washington, DC, United States) (2019), 119 (9), 6040-6085CODEN: CHREAY; ISSN:0009-2665. (American Chemical Society)A review. Membrane-permeabilizing peptides (MPPs) are as ubiquitous as the lipid bilayer membranes they act upon. Produced by all forms of life, most MPPs are used offensively or defensively against the membranes of other organisms. Just as Nature has found many uses for them, translational scientists have worked for decades to design or optimize MPPs for applications in the lab. and in the clinic ranging from antibacterial and antiviral therapy and prophylaxis to anticancer therapeutics and drug delivery. Here, we review the field of MPPs. We discuss the diversity of their sources and structures, the systems and methods used to measure their activities, and the behaviors that are obsd. We discuss the fact that "mechanism" is not a discrete or a static entity for a MPP, but rather the result of a heterogeneous and dynamic ensemble of structural states that vary in response to many different exptl. conditions. This has led to an almost complete lack of discrete 3-dimensional active structures among the thousands of known MPPs and a lack of useful or predictive sequence-structure-function relation rules. Ultimately, we discuss how it may be more useful to think of MPP mechanisms as broad regions of a mechanistic landscape rather than discrete mol. processes.
- 34Sun, L.; Hristova, K.; Bondar, A.-N.; Wimley, W. C. Structural Determinants of Peptide Nanopore Formation. ACS Nano 2024, 18, 15831– 15854, DOI: 10.1021/acsnano.4c02824There is no corresponding record for this reference.
- 35Michalska, M.; Wolf, P. Pseudomonas Exotoxin A: optimized by evolution for effective killing. Front. Microbiol. 2015, 6, 963, DOI: 10.3389/fmicb.2015.0096335https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC283ps1akug%253D%253D&md5=20c632c00c9c4e16292642a971b6ad47Pseudomonas Exotoxin A: optimized by evolution for effective killingMichalska Marta; Wolf PhilippFrontiers in microbiology (2015), 6 (), 963 ISSN:1664-302X.Pseudomonas Exotoxin A (PE) is the most toxic virulence factor of the pathogenic bacterium Pseudomonas aeruginosa. This review describes current knowledge about the intoxication pathways of PE. Moreover, PE represents a remarkable example for pathoadaptive evolution, how bacterial molecules have been structurally and functionally optimized under evolutionary pressure to effectively impair and kill their host cells.
- 36Deng, Q.; Barbieri, J. T. Molecular mechanisms of the cytotoxicity of ADP-ribosylating toxins. Annu. Rev. Microbiol. 2008, 62, 271– 288, DOI: 10.1146/annurev.micro.62.081307.16284836https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXht1Gls73N&md5=248e3f5084b0338ecfb1af1df7b31ad8Molecular mechanisms of the cytotoxicity of ADP-ribosylating toxinsDeng, Qing; Barbieri, Joseph T.Annual Review of Microbiology (2008), 62 (), 271-288CODEN: ARMIAZ; ISSN:0066-4227. (Annual Reviews Inc.)A review. Bacterial pathogens utilize toxins to modify or kill host cells. The bacterial ADP-ribosyltransferases are a family of protein toxins that covalently transfer the ADP-ribose portion of NAD to host proteins. Each bacterial ADP-ribosyltransferase toxin modifies a specific host protein(s) that yields a unique pathol. These toxins possess the capacity to enter a host cell or to use a bacterial type III app. for delivery into the host cell. Advances in the understanding of bacterial toxin action parallel the development of biophys. and structural biol. as well as an understanding of the mammalian cell. Bacterial toxins have been utilized as vaccines, as tools to dissect host cell physiol., and more recently for the development of novel therapies to treat human disease.
- 37Moss, D. L.; Park, H. W.; Mettu, R. R.; Landry, S. J. Deimmunizing substitutions in Pseudomonas exotoxin domain III perturb antigen processing without eliminating T-cell epitopes. J. Biol. Chem. 2019, 294 (12), 4667– 4681, DOI: 10.1074/jbc.RA118.006704There is no corresponding record for this reference.
- 38Wedekind, J. E.; Trame, C. B.; Dorywalska, M.; Koehl, P.; Raschke, T. M.; McKee, M.; FitzGerald, D.; Collier, R. J.; McKay, D. B. Refined crystallographic structure of Pseudomonas aeruginosa exotoxin A and its implications for the molecular mechanism of toxicity. J. Mol. Biol. 2001, 314 (4), 823– 837, DOI: 10.1006/jmbi.2001.5195There is no corresponding record for this reference.
- 39Marks, J. R.; Placone, J.; Hristova, K.; Wimley, W. C. Spontaneous membrane-translocating peptides by orthogonal high-throughput screening. J. Am. Chem. Soc. 2011, 133 (23), 8995– 9004, DOI: 10.1021/ja201741639https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXmtlWrtrk%253D&md5=c47082603ff99de9b48c0f6777249ae9Spontaneous Membrane-Translocating Peptides by Orthogonal High-Throughput ScreeningMarks, Jessica R.; Placone, Jesse; Hristova, Kalina; Wimley, William C.Journal of the American Chemical Society (2011), 133 (23), 8995-9004CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)Combinatorial peptide chem. and orthogonal high-throughput screening were used to select peptides that spontaneously translocate across synthetic lipid bilayer membranes without permeabilization. A conserved sequence motif was identified that contains several cationic residues in conserved positions in an otherwise hydrophobic sequence. This 9-residue motif rapidly translocates across synthetic multibilayer vesicles and into cells while carrying a large polar dye as a "cargo" moiety. The extraordinary ability of this family of peptides to spontaneously translocate across bilayers without an energy source of any kind is distinctly different from the behavior of the well-known, highly cationic cell-penetrating peptides, such as the HIV tat peptide, which do not translocate across synthetic bilayers, and enter cells mostly by active endocytosis. Peptides that translocate spontaneously across membranes have the potential to transform the field of drug design by enabling the delivery of otherwise membrane-impermeant polar drugs into cells and tissues. Here we describe the chem. tools needed to rapidly identify spontaneous membrane translocating peptides.
- 40He, J.; Kauffman, W. B.; Fuselier, T.; Naveen, S. K.; Voss, T. G.; Hristova, K.; Wimley, W. C. Direct Cytosolic Delivery of Polar Cargo to Cells by Spontaneous Membrane-translocating Peptides. J. Biol. Chem. 2013, 288 (41), 29974– 29986, DOI: 10.1074/jbc.M113.488312There is no corresponding record for this reference.
- 41Armstrong, J. K.; Wenby, R. B.; Meiselman, H. J.; Fisher, T. C. The hydrodynamic radii of macromolecules and their effect on red blood cell aggregation. Biophys. J. 2004, 87 (6), 4259– 4270, DOI: 10.1529/biophysj.104.04774641https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXhtVOmur7L&md5=1426af6c85786a5eead545b528518fc3The hydrodynamic radii of macromolecules and their effect on red blood cell aggregationArmstrong, J. K.; Wenby, R. B.; Meiselman, H. J.; Fisher, T. C.Biophysical Journal (2004), 87 (6), 4259-4270CODEN: BIOJAU; ISSN:0006-3495. (Biophysical Society)The effects of nonionic polymers on human red blood cell (RBC) aggregation were investigated. The hydrodynamic radius (Rh) of individual samples of dextran, polyvinylpyrrolidone, and polyoxyethylene over a range of mol. wts. (1500-2,000,000) were calcd. from their intrinsic viscosities using the Einstein viscosity relation and directly measured by quasi-elastic light scattering, and the effect of each polymer sample on RBC aggregation was studied by nephelometry and low-shear viscometry. For all three polymers, despite their different structures, samples with Rh <4 nm were found to inhibit aggregation, whereas those with Rh >4 nm enhanced aggregation. Inhibition increased with Rh and was maximal at ∼3 nm; above 4 nm, the pro-aggregant effect increased with Rh. For comparison, the Rh of 12 plasma proteins were calcd. from literature values of intrinsic viscosity or diffusion coeff. Each protein known to promote RBC aggregation had Rh >4 nm, whereas those with Rh <4 nm either inhibited or had no effect on aggregation. These results suggest that the influence of a nonionic polymer or plasma protein on RBC aggregation is simply a consequence of its size in an aq. environment and that the specific type of macromol. is of minor importance.
- 42Bohrer, M. P.; Deen, W. M.; Robertson, C. R.; Troy, J. L.; Brenner, B. M. Influence of molecular configuration on the passage of macromolecules across glomerular capillary wall. J. Gen. Physiol 1979, 74, 583– 593, DOI: 10.1085/jgp.74.5.583There is no corresponding record for this reference.
- 43Loret, C.; Chaufer, B.; Sebille, B.; Hamelin, M.; Blain, Y.; Le Hir, A. Characterization and hydrodynamic behaviour of modified gelatin: ii. Characterization by high performance size exclusion chromatography comparison with dextrans and proteins. Int. J. Biol. Macromol. 1988, 10, 366– 372, DOI: 10.1016/0141-8130(88)90031-1There is no corresponding record for this reference.
- 44Wu, E.; Jenschke, R. M.; Hristova, K.; Wimley, W. C. Rational Modulation of pH-Triggered Macromolecular Poration by Peptide Acylation and Dimerization. J. Phys. Chem. B 2020, 124, 8835– 8843, DOI: 10.1021/acs.jpcb.0c05363There is no corresponding record for this reference.
- 45Johnson, W. C. Protein secondary structure and circular dichroism: A practical guide. Proteins 1990, 7, 205– 214, DOI: 10.1002/prot.34007030245https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaK3cXksFGitbY%253D&md5=f1316896de77d441523c349575dbaaabProtein secondary structure and circular dichroism: a practical guideJohnson, W. Curtis, Jr.Proteins: Structure, Function, and Genetics (1990), 7 (3), 205-14CODEN: PSFGEY; ISSN:0887-3585.A review, with 24 refs., of CD spectroscopy in the study of protein secondary structure.
- 46Lakowicz, J. R. Principles of Fluorescence Spectroscopy; Plenum Press, 1983.There is no corresponding record for this reference.
- 47Ladokhin, A. S.; Jayasinghe, S.; White, S. H. How to measure and analyze tryptophan fluorescence in membranes properly, and why bother?. Anal. Biochem. 2000, 285 (2), 235– 245, DOI: 10.1006/abio.2000.477347https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD3cXmvFOksL0%253D&md5=99c2776187f13c33656bd1f4e59e2142How to Measure and Analyze Tryptophan Fluorescence in Membranes Properly, and Why Bother?Ladokhin, Alexey S.; Jayasinghe, Sajith; White, Stephen H.Analytical Biochemistry (2000), 285 (2), 235-245CODEN: ANBCA2; ISSN:0003-2697. (Academic Press)Tryptophan fluorescence is a powerful tool for studying protein structure and function, esp. membrane-active proteins and peptides. It is arguably the most frequently used tool for examg. the interactions of proteins and peptides with vesicular unilamellar model membranes. However, high light scattering assocd. with vesicular membrane systems presents special challenges. Because of their reduced light scattering compared to large unilamellar vesicles (LUV), small unilamellar vesicles (SUV) produced by sonication are widely used membrane models. Unfortunately, SUV, unlike LUV, are metastable and consequently unsuitable for equil. thermodn. measurements. We present simple and easily implemented exptl. procedures for the accurate detn. of tryptophan (Trp) fluorescence in either LUV or SUV. Specifically, we show that Trp spectra can be obtained in the presence of up to 6 mM LUV that are virtually identical to spectra obtained in buffer alone, which obviates the use of SUV. We show how the widths and peak positions of such spectra can be used to evaluate the heterogeneity of the membrane conformation and penetration of peptides. Finally, we show how to use a ref. fluorophore for the correction of intensity measurements so that the energetics of peptide partitioning into membranes can be accurately detd. (c) 2000 Academic Press.
- 48Pittman, A. E.; Marsh, B. P.; King, G. M. Conformations and Dynamic Transitions of a Melittin Derivative That Forms Macromolecule-Sized Pores in Lipid Bilayers. Langmuir 2018, 34 (28), 8393– 8399, DOI: 10.1021/acs.langmuir.8b00804There is no corresponding record for this reference.
- 49Sun, L.; Hristova, K.; Wimley, W. C. Membrane-selective nanoscale pores in liposomes by a synthetically evolved peptide: implications for triggered release. Nanoscale 2021, 13 (28), 12185– 12197, DOI: 10.1039/D1NR03084AThere is no corresponding record for this reference.
- 50Di, L. Strategic approaches to optimizing peptide ADME properties. AAPS J. 2015, 17 (1), 134– 143, DOI: 10.1208/s12248-014-9687-350https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2cXhvFSnurvJ&md5=530597d3719d65509105096f433fee0aStrategic Approaches to Optimizing Peptide ADME PropertiesDi, LiAAPS Journal (2015), 17 (1), 134-143CODEN: AJAOB6; ISSN:1550-7416. (Springer)Development of peptide drugs is challenging but also quite rewarding. Five blockbuster peptide drugs are currently on the market, and six new peptides received first marketing approval as new mol. entities in 2012. Although peptides only represent 2% of the drug market, the market is growing twice as quickly and might soon occupy a larger niche. Natural peptides typically have poor absorption, distribution, metab., and excretion (ADME) properties with rapid clearance, short half-life, low permeability, and sometimes low soly. Strategies have been developed to improve peptide drugability through enhancing permeability, reducing proteolysis and renal clearance, and prolonging half-life. In vivo, in vitro, and in silico tools are available to evaluate ADME properties of peptides, and structural modification strategies are in place to improve peptide developability.
- 51Brunsveld, L.; Waldmann, H.; Huster, D. Membrane binding of lipidated Ras peptides and proteins--the structural point of view. Biochim. Biophys. Acta 2009, 1788 (1), 273– 288, DOI: 10.1016/j.bbamem.2008.08.006There is no corresponding record for this reference.
- 52Zhang, L.; Bulaj, G. Converting peptides into drug leads by lipidation. Curr. Med. Chem. 2012, 19 (11), 1602– 1618, DOI: 10.2174/09298671279994500352https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XmsF2jt7s%253D&md5=a9f289aea1cea75a566370a1b2c06583Converting peptides into drug leads by lipidationZhang, L.; Bulaj, G.Current Medicinal Chemistry (2012), 19 (11), 1602-1618CODEN: CMCHE7; ISSN:0929-8673. (Bentham Science Publishers Ltd.)A review. Lipidation is a posttranslational modification of proteins that has also found its use in designing peptide drugs. The presence of a lipid group in peptides modulates their hydrophobicity, secondary structures and self-assembling propensities while retaining their abilities to bind to target receptors. Lipidation improves peptides' metabolic stability, membrane permeability, bioavailability, and changes peptides' pharmacokinetic and pharmacodynamic properties. Herein, we review the applications of various lipidation strategies in peptide drug design, the effects of the chain length and anchor position of fatty acids in peptide lipidation, the physicochem. and biol. properties of selected lipidated peptides and the synthesis strategies for peptide lipidation.
- 53Ezzat, K.; Andaloussi, S. E.; Zaghloul, E. M.; Lehto, T.; Lindberg, S.; Moreno, P. M.; Viola, J. R.; Magdy, T.; Abdo, R.; Guterstam, P. PepFect 14, a novel cell-penetrating peptide for oligonucleotide delivery in solution and as solid formulation. Nucleic Acids Res. 2011, 39 (12), 5284– 5298, DOI: 10.1093/nar/gkr07253https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXoslOgtbw%253D&md5=64e66305efb772c25386977d53802340PepFect 14, a novel cell-penetrating peptide for oligonucleotide delivery in solution and as solid formulationEzzat, Kariem; El Andaloussi, Samir; Zaghloul, Eman M.; Lehto, Taavi; Lindberg, Staffan; Moreno, Pedro M. D.; Viola, Joana R.; Magdy, Tarek; Abdo, Rania; Guterstam, Peter; Sillard, Rannar; Hammond, Suzan M.; Wood, Matthew J. A.; Arzumanov, Andrey A.; Gait, Michael J.; Smith, C. I. Edvard; Haellbrink, Mattias; Langel, UeloNucleic Acids Research (2011), 39 (12), 5284-5298CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)Numerous human genetic diseases are caused by mutations that give rise to aberrant alternative splicing. Recently, several of these debilitating disorders have been shown to be amenable for splice-correcting oligonucleotides (SCOs) that modify splicing patterns and restore the phenotype in exptl. models. However, translational approaches are required to transform SCOs into usable drug products. In this study, we present a new cell-penetrating peptide, PepFect14 (PF14), which efficiently delivers SCOs to different cell models including HeLa pLuc705 and mdx mouse myotubes; a cell culture model of Duchenne's muscular dystrophy (DMD). Non-covalent PF14-SCO nanocomplexes induce splice-correction at rates higher than the com. available lipid-based vector Lipofectamine 2000 (LF2000) and remain active in the presence of serum. Furthermore, we demonstrate the feasibility of incorporating this delivery system into solid formulations that could be suitable for several therapeutic applications. Solid dispersion technique is utilized and the formed solid formulations are as active as the freshly prepd. nanocomplexes in soln. even when stored at an elevated temps. for several weeks. In contrast, LF2000 drastically loses activity after being subjected to same procedure. This shows that using PF14 is a very promising translational approach for the delivery of SCOs in different pharmaceutical forms.
- 54Gatto, E.; Mazzuca, C.; Stella, L.; Venanzi, M.; Toniolo, C.; Pispisa, B. Effect of peptide lipidation on membrane perturbing activity: a comparative study on two trichogin analogues. J. Phys. Chem. B 2006, 110 (45), 22813– 22818, DOI: 10.1021/jp064580j54https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD28XhtVyntbnN&md5=76fb933226981c03757eb547119aee16Effect of Peptide Lipidation on Membrane Perturbing Activity: A Comparative Study on Two Trichogin AnaloguesGatto, Emanuela; Mazzuca, Claudia; Stella, Lorenzo; Venanzi, Mariano; Toniolo, Claudio; Pispisa, BasilioJournal of Physical Chemistry B (2006), 110 (45), 22813-22818CODEN: JPCBFK; ISSN:1520-6106. (American Chemical Society)The effect of lipidation on the membrane perturbing activity of peptaibol antibiotics was investigated by performing a comparative study on two synthetic analogs of the natural peptide trichogin GA IV. Both analogs were labeled with a hydrophobic fluorescent probe, but one of them lacked the N-terminal n-octanoyl chain, present in the natural peptide. Spectroscopic studies show that the fatty acyl chain produces two opposite effects: It increases the affinity of the monomeric peptide for the membrane phase, but, at the same time, it favors peptide aggregation in water, thus inhibiting membrane binding by reducing the effective monomer concn. In the membrane phase the two analogs exhibit the same aggregation and orientation behavior, indicating that the n-octanoyl chain plays no specific role in detg. their orientation or membrane perturbing activity. Indeed, the dependence of peptide-induced membrane leakage on total peptide concn. is basically the same for the two analogs, because the aforementioned opposite effects, caused by peptide lipidation, tend to balance. These findings make questionable the use of lipidation as a general method for increasing the peptide membrane-perturbing activity, as its validity seems to be restricted to parent compds. of limited overall hydrophobicity.
- 55Müller, A.; Wenzel, M.; Strahl, H.; Grein, F.; Saaki, T. N. V.; Kohl, B.; Siersma, T.; Bandow, J. E.; Sahl, H. G.; Schneider, T.; Hamoen, L. W. Daptomycin inhibits cell envelope synthesis by interfering with fluid membrane microdomains. Proc. Natl. Acad. Sci. U.S.A. 2016, 113 (45), E7077– E7086, DOI: 10.1073/pnas.161117311355https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhslalsLrL&md5=900e3e7b23f443b37e0a0a1ba6ec590fDaptomycin inhibits cell envelope synthesis by interfering with fluid membrane microdomainsMueller, Anna; Wenzel, Michaela; Strahl, Henrik; Grein, Fabian; Saaki, Terrens N. V.; Kohl, Bastian; Siersma, Tjalling; Bandow, Julia E.; Sahl, Hans-Georg; Schneider, Tanja; Hamoen, Leendert W.Proceedings of the National Academy of Sciences of the United States of America (2016), 113 (45), E7077-E7086CODEN: PNASA6; ISSN:0027-8424. (National Academy of Sciences)Daptomycin is a highly efficient last-resort antibiotic that targets the bacterial cell membrane. Despite its clin. importance, the exact mechanism by which daptomycin kills bacteria is not fully understood. Different expts. have led to different models, including (i) blockage of cell wall synthesis, (ii) membrane pore formation, and (iii) the generation of altered membrane curvature leading to aberrant recruitment of proteins. To det. which model is correct, the authors carried out a comprehensive mode-of-action study using the model organism Bacillus subtilis and different assays, including proteomics, ionomics, and fluorescence light microscopy. The authors found that daptomycin causes a gradual decrease in membrane potential but does not form discrete membrane pores. Although the authors found no evidence for altered membrane curvature, the authors confirmed that daptomycin inhibits cell wall synthesis. Interestingly, using different fluorescent lipid probes, the authors showed that binding of daptomycin led to a drastic rearrangement of fluid lipid domains, affecting overall membrane fluidity. Importantly, these changes resulted in the rapid detachment of the membrane-assocd. lipid II synthase MurG and the phospholipid synthase PlsX. Both proteins preferentially colocalize with fluid membrane microdomains. Delocalization of these proteins presumably is a key reason why daptomycin blocks cell wall synthesis. Finally, clustering of fluid lipids by daptomycin likely causes hydrophobic mismatches between fluid and more rigid membrane areas. This mismatch can facilitate proton leakage and may explain the gradual membrane depolarization obsd. with daptomycin. Targeting of fluid lipid domains has not been described before for antibiotics and adds another dimension to the authors' understanding of membrane-active antibiotics.
- 56Lin, B. F.; Missirlis, D.; Krogstad, D. V.; Tirrell, M. Structural effects and lipid membrane interactions of the pH-responsive GALA peptide with fatty acid acylation. Biochemistry 2012, 51 (23), 4658– 4668, DOI: 10.1021/bi300314h56https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XntFSgsLs%253D&md5=0d8f3bc422caab96b4f5b8f373ef21f2Structural Effects and Lipid Membrane Interactions of the pH-Responsive GALA Peptide with Fatty Acid AcylationLin, Brian F.; Missirlis, Dimitris; Krogstad, Daniel V.; Tirrell, MatthewBiochemistry (2012), 51 (23), 4658-4668CODEN: BICHAW; ISSN:0006-2960. (American Chemical Society)GALA is a pH-responsive, membrane-perturbing peptide designed to fold from a random coil at physiol. pH to an amphipathic α-helix under mildly acidic conditions. Because of its pH-activated function, GALA has been sought-after as a component of intracellular drug delivery systems that could actively propel endosomal escape. In this study, we conjugated GALA with lauryl and palmitoyl fatty acid tails as model hydrophobic moieties and examd. the physicochem. characteristics and activities of the resulting peptide amphiphiles (PAs). The fatty acid variants of GALA exhibited distinctly different membrane perturbing mechanisms at pH 7.5 and 5.5. At physiol. pH, the PAs ruptured liposomes through a surfactant-like mechanism. At pH 5.5, lauryl-GALA was shown to form transmembrane pores with a higher potency as compared to its unmodified peptide counterpart; however, after prolonged exposure it also caused liposome lysis. The lytic activity of fatty acid-conjugated GALA did not impair cell viability. Lauryl-GALA was tolerated well by SJSA-1 osteocarcinoma cells and enhanced cell internalization of the PA was obsd. Our findings are discussed with the overarching goal of developing efficient therapeutic delivery systems.
- 57Chu-Kung, A. F.; Nguyen, R.; Bozzelli, K. N.; Tirrell, M. Chain length dependence of antimicrobial peptide-fatty acid conjugate activity. J. Colloid Interface Sci. 2010, 345 (2), 160– 167, DOI: 10.1016/j.jcis.2009.11.05757https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXkt1OktbY%253D&md5=1b283ed963ec56a1255b0706d3767253Chain length dependence of antimicrobial peptide-fatty acid conjugate activityChu-Kung, Alexander F.; Nguyen, Rose; Bozzelli, Kristen N.; Tirrell, MatthewJournal of Colloid and Interface Science (2010), 345 (2), 160-167CODEN: JCISA5; ISSN:0021-9797. (Elsevier B.V.)The rise of resistant bacteria has prompted the search for new antimicrobial agents. Antimicrobial membrane lytic peptides have potential as future microbial agents due to their novel mode of action. Recently conjugation of a fatty acid to antimicrobial peptides has been explored as a method to modulate the activity and selectivity of the peptide. Our work further explores these phenomena by testing two peptides, YGAAKKAAKAAKKAAKAA (AKK) and LKKLLKLLKLLKL (LKK), conjugated to fatty acids of varying length for their activity, structure, soln. assembly properties and the ability to bind model membranes. We found that increasing the length of fatty acids conjugated to peptide AKK, up to a 16 carbons in length, increases the antimicrobial activity. Peptide AKK appears to lose activity when the minimal active concn. is higher than the crit. micelle concn. (CMC) of the mol. Thus, if the CMC of the peptide conjugate is too low the activity is lost. Peptide LKK has no activity when conjugated to lauric acid and appears to aggregate at very low concns. Conjugation of AKK with a fatty acid increases its affinity to model supported lipid membranes. It appears that the increased hydrophobic interaction imparted by the fatty acid increases the affinity of the peptide to the surface thus increasing its activity. At concns. above the CMC, soln. self-assembly inhibits binding of the peptide to cell membranes.
- 58Sweatt, A. J.; Griffiths, C. D.; Groves, S. M.; Paudel, B. B.; Wang, L.; Kashatus, D. F.; Janes, K. A. Proteome-wide copy-number estimation from transcriptomics. Mol. Syst. Biol. 2024, 20 (11), 1230– 1256, DOI: 10.1038/s44320-024-00064-3There is no corresponding record for this reference.
Supporting Information
Supporting Information
The Supporting Information is available free of charge at https://pubs.acs.org/doi/10.1021/acsnano.4c07525.
Confocal microscopy images of uptake and delivery; quantitation of protein delivery; quantitation of antibody fragment delivery; model calculation for uptake and delivery (PDF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.