1
|
Georgieva M, Stojceski F, Wüthrich F, Sosthène C, Blanco Pérez L, Grasso G, Jacquier N. Mutations in the essential outer membrane protein BamA contribute to Escherichia coli resistance to the antimicrobial peptide TAT-RasGAP 317-326. J Biol Chem 2025; 301:108018. [PMID: 39608713 PMCID: PMC11842939 DOI: 10.1016/j.jbc.2024.108018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/04/2024] [Accepted: 11/15/2024] [Indexed: 11/30/2024] Open
Abstract
Antimicrobial peptides (AMPs) are promising alternatives to classical antibiotics against antibiotic-resistant pathogens. TAT-RasGAP317-326 is an AMP with broad range antibacterial activity, but its mechanism of action is unknown. In this study, we analyzed a strain of Escherichia coli with extensive resistance to TAT-RasGAP317-326 but not to other AMPs that we obtained after twenty passages during an in vitro resistance selection experiment. This strain accumulated four mutations. One of these is a point mutation in bamA, which encodes an essential protein involved in the folding and proper insertion of outer membrane proteins. The mutation resulted in a change of charge in a surface-exposed negatively charged loop of the BamA protein. Using CRISPR-Cas9-based targeted mutagenesis, we showed that mutations lowering the negative charge of this loop decreased sensitivity of E. coli to TAT-RasGAP317-326. In silico simulations unveiled the molecular driving forces responsible for the interaction between TAT-RasGAP317-326 and BamA. These results indicated that electrostatic interactions, particularly hydrogen bonds, are involved in the stability of the molecular complex, representing a predictive fingerprint of the TAT-RasGAP317-326 - BamA interaction strength. Interestingly, BamA activity was only partially affected by TAT-RasGAP317-326, indicating that BamA may function as a specific receptor for this AMP. Our results indicate that binding and entry of TAT-RasGAP317-326 may involve different mechanisms compared to other AMPs, which is in line with limited cross-resistance observed between different AMPs. This limited cross-resistance is important for the clinical application of AMPs towards drug-resistant pathogens.
Collapse
Affiliation(s)
- Maria Georgieva
- Institute of Microbiology, University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Filip Stojceski
- Dalle Molle Institute for Artificial Intelligence, IDSIA USI-SUPSI, Lugano, Switzerland
| | - Fabian Wüthrich
- Institute of Microbiology, University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Carole Sosthène
- Institute of Microbiology, University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Laura Blanco Pérez
- Institute of Microbiology, University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Gianvito Grasso
- Dalle Molle Institute for Artificial Intelligence, IDSIA USI-SUPSI, Lugano, Switzerland
| | - Nicolas Jacquier
- Institute of Microbiology, University Hospital and University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
2
|
Maani Z, Rahbarnia L, Bahadori A, Chollou KM, Farajnia S. Spotlight on HIV-derived TAT peptide as a molecular shuttle in drug delivery. Drug Discov Today 2024; 29:104191. [PMID: 39322176 DOI: 10.1016/j.drudis.2024.104191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/08/2024] [Accepted: 09/19/2024] [Indexed: 09/27/2024]
Abstract
HIV-derived TAT peptide, with a high penetration rate into cells and its nonimmunogenic and minimally toxic nature, is an attractive tool for enhancing the biodistribution of drugs and their systemic administration. Despite the presence of numerous promising preclinical investigations illustrating its capability to specifically target distinct tissues and deliver a diverse range of pharmacological agents, the efficacy of various clinical trials incorporating TAT has been impeded by several considerable obstacles. Hence, there is much need for an in-depth investigation concerning the application of TAT in drug delivery mechanisms. In this review, we have elucidated the structure of TAT and its utility in the proficient delivery of various types of bioactive molecules.
Collapse
Affiliation(s)
- Zahra Maani
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Leila Rahbarnia
- Infectious and Tropical Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Ali Bahadori
- Department of Medical Microbiology, Sarab Faculty of Medical Sciences, Sarab, Iran
| | | | - Safar Farajnia
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
3
|
Serulla M, Anees P, Hallaj A, Trofimenko E, Kalia T, Krishnan Y, Widmann C. Plasma membrane depolarization reveals endosomal escape incapacity of cell-penetrating peptides. Eur J Pharm Biopharm 2023; 184:116-124. [PMID: 36709921 DOI: 10.1016/j.ejpb.2023.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 12/12/2022] [Accepted: 01/23/2023] [Indexed: 01/27/2023]
Abstract
Cell-penetrating peptides (CPPs) are short (<30 amino acids), generally cationic, peptides that deliver diverse cargos into cells. CPPs access the cytosol either by direct translocation through the plasma membrane or via endocytosis followed by endosomal escape. Both direct translocation and endosomal escape can occur simultaneously, making it non-trivial to specifically study endosomal escape alone. Here we depolarize the plasma membrane and showed that it inhibits the direct translocation of several CPPs but does not affect their uptake into endosomes. Despite good endocytic uptake many CPPs previously considered to access the cytosol via endosomal escape, failed to access the cytosol once direct translocation was abrogated. Even CPPs designed for enhanced endosomal escape actually showed negligible endosomal escape into the cytosol. Our data reveal that cytosolic localization of CPPs occurs mainly by direct translocation across the plasma membrane. Cell depolarization represents a simple manipulation to stringently test the endosomal escape capacity of CPPs.
Collapse
Affiliation(s)
- Marc Serulla
- Department of Biomedical Sciences, University of Lausanne, 1005 Lausanne, Switzerland
| | - Palapuravan Anees
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA; Grossman Institute of Neuroscience, Quantitative Biology and Human Behavior, The University of Chicago, Chicago, IL 60637, USA
| | - Ali Hallaj
- Department of Biomedical Sciences, University of Lausanne, 1005 Lausanne, Switzerland
| | - Evgeniya Trofimenko
- Department of Biomedical Sciences, University of Lausanne, 1005 Lausanne, Switzerland
| | - Tara Kalia
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA
| | - Yamuna Krishnan
- Department of Chemistry, The University of Chicago, Chicago, IL 60637, USA; Grossman Institute of Neuroscience, Quantitative Biology and Human Behavior, The University of Chicago, Chicago, IL 60637, USA
| | - Christian Widmann
- Department of Biomedical Sciences, University of Lausanne, 1005 Lausanne, Switzerland.
| |
Collapse
|
4
|
Warner JM, An D, Stratton BS, O'Shaughnessy B. A hemifused complex is the hub in a network of pathways to membrane fusion. Biophys J 2023; 122:374-385. [PMID: 36463406 PMCID: PMC9892611 DOI: 10.1016/j.bpj.2022.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 06/25/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
Membrane fusion is a critical step for many essential processes, from neurotransmission to fertilization. For over 40 years, protein-free fusion driven by calcium or other cationic species has provided a simplified model of biological fusion, but the mechanisms remain poorly understood. Cation-mediated membrane fusion and permeation are essential in their own right to drug delivery strategies based on cell-penetrating peptides or cation-bearing lipid nanoparticles. Experimental studies suggest calcium drives anionic membranes to a hemifused intermediate that constitutes a hub in a network of pathways, but the pathway selection mechanism is unknown. Here we develop a mathematical model that identifies the network hub as a highly dynamic hemifusion complex. Multivalent cations drive expansion of this high-tension hemifusion interface between interacting vesicles during a brief transient. The fate of this interface determines the outcome, either fusion, dead-end hemifusion, or vesicle lysis. The model reproduces the unexplained finding that calcium-driven fusion of vesicles with planar membranes typically stalls at hemifusion, and we show the equilibrated hemifused state is a novel lens-shaped complex. Thus, membrane fusion kinetics follow a stochastic trajectory within a network of pathways, with outcome weightings set by a hemifused complex intermediate.
Collapse
Affiliation(s)
- Jason M Warner
- Department of Chemical Engineering, Columbia University, New York, New York
| | - Dong An
- Department of Chemical Engineering, Columbia University, New York, New York
| | | | - Ben O'Shaughnessy
- Department of Chemical Engineering, Columbia University, New York, New York.
| |
Collapse
|
5
|
Vizzarro G, Jacquier N. In vitro synergistic action of TAT-RasGAP 317-326 peptide with antibiotics against Gram-negative pathogens. J Glob Antimicrob Resist 2022; 31:295-303. [PMID: 36270448 DOI: 10.1016/j.jgar.2022.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/05/2022] [Accepted: 10/11/2022] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVES Multidrug-resistant (MDR) bacteria are a continuously increasing threat for medicine, causing infections recalcitrant to antibiotics. Antimicrobial peptides (AMPs) were identified as alternatives to antibiotics, being naturally occurring short peptides and part of the innate immune system of a vast majority of organisms. However, the clinical application of AMPs is limited by suboptimal pharmacokinetic properties and relatively high toxicity. Combinatorial treatments using AMPs and classical antibiotics may decrease the concentrations of AMPs required for bacterial eradication, thus lowering the side effects of these peptides. METHODS Here, we investigate the in vitro efficiency of combinations of the recently described antimicrobial peptide TAT-RasGAP317-326 with a panel of commonly used antimicrobial agents against three Gram-negative bacteria, Escherichia coli, Pseudomonas aeruginosa and Acinetobacter baumannii, using checkerboard and time-kill assays. RESULTS We identified synergistic combinations towards all three bacteria and demonstrated that these combinations had an increased bactericidal effect compared to individual drugs. Moreover, combinations were also effective against clinical isolates of A. baumannii. Finally, combination of TAT-RasGAP317-326 and meropenem had a promising antibiofilm effect towards A. baumannii. CONCLUSIONS Taken together, our results indicate that combinations of TAT-RasGAP317-326 with commonly used antimicrobial agents may lead to the development of new treatment protocols against infections caused by MDR bacteria.
Collapse
Affiliation(s)
- Grazia Vizzarro
- Institute of Microbiology, University Hospital Center and University of Lausanne, Lausanne, Switzerland
| | - Nicolas Jacquier
- Institute of Microbiology, University Hospital Center and University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
6
|
Sarkar D, Mishra S, Nisal R, Majhi S, Shrivas R, Singh Y, Anusree VS, Kalia J. Site-Specific Fluorescent Labeling of the Cysteine-Rich Toxin, DkTx, for TRPV1 Ion Channel Imaging and Membrane Binding Studies. Bioconjug Chem 2022; 33:1761-1770. [PMID: 36073164 DOI: 10.1021/acs.bioconjchem.2c00355] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Peptide toxins secreted by venomous animals bind to mammalian ion channel proteins and modulate their function. The high specificity of these toxins for their target ion channels enables them to serve as powerful tools for ion channel biology. Toxins labeled with fluorescent dyes are employed for the cellular imaging of channels and also for studying toxin-channel and toxin-membrane interactions. Several of these toxins are cysteine-rich, rendering the production of properly folded fluorescently labeled toxins technically challenging. Herein, we evaluate a variety of site-specific protein bioconjugation approaches for producing fluorescently labeled double-knot toxin (DkTx), a potent TRPV1 ion channel agonist that contains an uncommonly large number of cysteines (12 out of a total of 75 amino acids present in the protein). We find that popular cysteine-mediated bioconjugation approaches are unsuccessful as the introduction of a non-native cysteine residue for thiol modification leads to the formation of misfolded toxin species. Moreover, N-terminal aldehyde-mediated bioconjugation approaches are also not suitable as the resultant labeled toxin lacks activity. In contrast to these approaches, C-terminal bioconjugation of DkTx via the sortase bioconjugation technology yields functionally active fluorescently labeled DkTx. We employ this labeled toxin for imaging rat TRPV1 heterologously expressed in Xenopus laevis oocytes, as well as for performing membrane binding studies on giant unilamellar vesicles composed of different lipid compositions. Our studies set the stage for using fluorescent DkTx as a tool for TRPV1 biology and provide an informative blueprint for labeling cysteine-rich proteins.
Collapse
Affiliation(s)
- Debayan Sarkar
- Department of Biology, Indian Institute of Science Education and Research (IISER) Pune, Dr. Homi Bhabha Road, Pashan, Pune, Maharashtra 411008, India.,Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Bhopal, Bhopal Bypass Road, Bhauri, Bhopal, Madhya Pradesh 462066, India
| | - Satyajit Mishra
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Bhopal, Bhopal Bypass Road, Bhauri, Bhopal, Madhya Pradesh 462066, India
| | - Rahul Nisal
- Department of Chemistry, Indian Institute of Science Education and Research (IISER) Pune, Dr. Homi Bhabha Road, Pashan, Pune, Maharashtra 411008, India
| | - Sumita Majhi
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Bhopal, Bhopal Bypass Road, Bhauri, Bhopal, Madhya Pradesh 462066, India
| | - Rohit Shrivas
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Bhopal, Bhopal Bypass Road, Bhauri, Bhopal, Madhya Pradesh 462066, India
| | - Yashaswi Singh
- Department of Biology, Indian Institute of Science Education and Research (IISER) Pune, Dr. Homi Bhabha Road, Pashan, Pune, Maharashtra 411008, India.,Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Bhopal, Bhopal Bypass Road, Bhauri, Bhopal, Madhya Pradesh 462066, India
| | - V S Anusree
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Bhopal, Bhopal Bypass Road, Bhauri, Bhopal, Madhya Pradesh 462066, India
| | - Jeet Kalia
- Department of Biology, Indian Institute of Science Education and Research (IISER) Pune, Dr. Homi Bhabha Road, Pashan, Pune, Maharashtra 411008, India.,Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Bhopal, Bhopal Bypass Road, Bhauri, Bhopal, Madhya Pradesh 462066, India.,Department of Chemistry, Indian Institute of Science Education and Research (IISER) Pune, Dr. Homi Bhabha Road, Pashan, Pune, Maharashtra 411008, India.,Department of Chemistry, Indian Institute of Science Education and Research (IISER) Bhopal, Bhopal Bypass Road, Bhauri, Bhopal, Madhya Pradesh 462066, India
| |
Collapse
|
7
|
The EnvZ/OmpR Two-Component System Regulates the Antimicrobial Activity of TAT-RasGAP 317-326 and the Collateral Sensitivity to Other Antibacterial Agents. Microbiol Spectr 2022; 10:e0200921. [PMID: 35579440 PMCID: PMC9241736 DOI: 10.1128/spectrum.02009-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The rapid emergence of antibiotic-resistant bacteria poses a serious threat to public health worldwide. Antimicrobial peptides (AMPs) are promising antibiotic alternatives; however, little is known about bacterial mechanisms of AMP resistance and the interplay between AMP resistance and the bacterial response to other antimicrobials. In this study, we identified Escherichia coli mutants resistant to the TAT-RasGAP317-326 antimicrobial peptide and found that resistant bacteria show collateral sensitivity to other AMPs and antibacterial agents. We determined that resistance to TAT-RasGAP317-326 peptide arises through mutations in the histidine kinase EnvZ, a member of the EnvZ/OmpR two-component system responsible for osmoregulation in E. coli. In particular, we found that TAT-RasGAP317-326 binding and entry is compromised in E. coli peptide-resistant mutants. We showed that peptide resistance is associated with transcriptional regulation of a number of pathways and EnvZ-mediated resistance is dependent on the OmpR response regulator but is independent of the OmpC and OmpF outer membrane porins. Our findings provide insight into the bacterial mechanisms of TAT-RasGAP317-326 resistance and demonstrate that resistance to this AMP is associated with collateral sensitivity to other antibacterial agents. IMPORTANCE Antimicrobial peptides (AMP) are promising alternatives to classical antibiotics in the fight against antibiotic resistance. Resistance toward antimicrobial peptides can occur, but little is known about the mechanisms driving this phenomenon. Moreover, there is limited knowledge on how AMP resistance relates to the bacterial response to other antimicrobial agents. Here, we address these questions in the context of the antimicrobial peptide TAT-RasGAP317-326. We show that resistant Escherichia coli strains can be selected and do not show resistance to other antimicrobial agents. Resistance is caused by a mutation in a regulatory pathway, which lowers binding and entry of the peptide in E. coli. Our results highlight a mechanism of resistance that is specific to TAT-RasGAP317-326. Further research is required to characterize these mechanisms and to evaluate the potential of antimicrobial combinations to curb the development of antimicrobial resistance.
Collapse
|
8
|
Xian W, Hennefarth MR, Lee MW, Do T, Lee EY, Alexandrova AN, Wong GCL. Histidine-Mediated Ion Specific Effects Enable Salt Tolerance of a Pore-Forming Marine Antimicrobial Peptide. Angew Chem Int Ed Engl 2022; 61:e202108501. [PMID: 35352449 PMCID: PMC9189074 DOI: 10.1002/anie.202108501] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Indexed: 12/19/2022]
Abstract
Antimicrobial peptides (AMPs) preferentially permeate prokaryotic membranes via electrostatic binding and membrane remodeling. Such action is drastically suppressed by high salt due to increased electrostatic screening, thus it is puzzling how marine AMPs can possibly work. We examine as a model system, piscidin-1, a histidine-rich marine AMP, and show that ion-histidine interactions play unanticipated roles in membrane remodeling at high salt: Histidines can simultaneously hydrogen-bond to a phosphate and coordinate with an alkali metal ion to neutralize phosphate charge, thereby facilitating multidentate bonds to lipid headgroups in order to generate saddle-splay curvature, a prerequisite to pore formation. A comparison among Na+ , K+ , and Cs+ indicates that histidine-mediated salt tolerance is ion specific. We conclude that histidine plays a unique role in enabling protein/peptide-membrane interactions that occur in marine or other high-salt environment.
Collapse
Affiliation(s)
- Wujing Xian
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Matthew R Hennefarth
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Michelle W Lee
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Tran Do
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ernest Y Lee
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Anastassia N Alexandrova
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nano Systems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Gerard C L Wong
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA 90095, USA
- California Nano Systems Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
9
|
Bechtella L, Chalouhi E, Milán Rodríguez P, Cosset M, Ravault D, Illien F, Sagan S, Carlier L, Lequin O, Fuchs PFJ, Sachon E, Walrant A. Structural Bases for the Involvement of Phosphatidylinositol-4,5-bisphosphate in the Internalization of the Cell-Penetrating Peptide Penetratin. ACS Chem Biol 2022; 17:1427-1439. [PMID: 35608167 DOI: 10.1021/acschembio.1c00974] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Cell-penetrating peptides cross cell membranes through various parallel internalization pathways. Herein, we analyze the role of the negatively charged lipid phosphatidylinositol-4,5-bisphosphate (PI(4,5)P2) in the internalization of Penetratin. Contributions of both inner leaflet and outer leaflet pools of PI(4,5)P2 were revealed by quantifying the internalization of Penetratin in cells treated with PI(4,5)P2 binders. Studies on model systems showed that Penetratin has a strong affinity for PI(4,5)P2 and interacts selectively with this lipid, even in the presence of other negatively charged lipids, as demonstrated by affinity photo-crosslinking experiments. Differential scanning calorimetry experiments showed that Penetratin induces lateral segregation in PI(4,5)P2-containing liposomes, which was confirmed by coarse-grained molecular dynamics simulations. NMR experiments indicated that Penetratin adopts a stabilized helical conformation in the presence of PI(4,5)P2-containing membranes, with an orientation parallel to the bilayer plane, which was also confirmed by all-atom simulations. NMR and photo-crosslinking experiments also suggest a rather shallow insertion of the peptide in the membrane. Put together, our findings suggest that PI(4,5)P2 is a privileged interaction partner for Penetratin and that it plays an important role in Penetratin internalization.
Collapse
Affiliation(s)
- Leïla Bechtella
- Laboratoire des Biomolécules, LBM, Sorbonne Université, École normale supérieure, PSL University, CNRS, 75005 Paris, France
| | - Edward Chalouhi
- Laboratoire des Biomolécules, LBM, Sorbonne Université, École normale supérieure, PSL University, CNRS, 75005 Paris, France
| | - Paula Milán Rodríguez
- Laboratoire des Biomolécules, LBM, Sorbonne Université, École normale supérieure, PSL University, CNRS, 75005 Paris, France
| | - Marine Cosset
- Laboratoire des Biomolécules, LBM, Sorbonne Université, École normale supérieure, PSL University, CNRS, 75005 Paris, France
| | - Delphine Ravault
- Laboratoire des Biomolécules, LBM, Sorbonne Université, École normale supérieure, PSL University, CNRS, 75005 Paris, France
| | - Françoise Illien
- Laboratoire des Biomolécules, LBM, Sorbonne Université, École normale supérieure, PSL University, CNRS, 75005 Paris, France
| | - Sandrine Sagan
- Laboratoire des Biomolécules, LBM, Sorbonne Université, École normale supérieure, PSL University, CNRS, 75005 Paris, France
| | - Ludovic Carlier
- Laboratoire des Biomolécules, LBM, Sorbonne Université, École normale supérieure, PSL University, CNRS, 75005 Paris, France
| | - Olivier Lequin
- Laboratoire des Biomolécules, LBM, Sorbonne Université, École normale supérieure, PSL University, CNRS, 75005 Paris, France
| | - Patrick F. J. Fuchs
- Laboratoire des Biomolécules, LBM, Sorbonne Université, École normale supérieure, PSL University, CNRS, 75005 Paris, France
- Université de Paris, UFR Sciences du Vivant, 75013 Paris, France
| | - Emmanuelle Sachon
- Laboratoire des Biomolécules, LBM, Sorbonne Université, École normale supérieure, PSL University, CNRS, 75005 Paris, France
- Sorbonne Université, Mass Spectrometry Sciences Sorbonne Université, MS3U platform, UFR 926, UFR 927, Paris 75005, France
| | - Astrid Walrant
- Laboratoire des Biomolécules, LBM, Sorbonne Université, École normale supérieure, PSL University, CNRS, 75005 Paris, France
| |
Collapse
|
10
|
Xian W, Hennefarth MR, Lee MW, Do T, Lee EY, Alexandrova AN, Wong GCL. Histidine‐Mediated Ion Specific Effects Enable Salt Tolerance of a Pore‐Forming Marine Antimicrobial Peptide. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202108501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Wujing Xian
- Department of Bioengineering University of California, Los Angeles Los Angeles CA 90095 USA
| | - Matthew R. Hennefarth
- Department of Chemistry and Biochemistry University of California, Los Angeles Los Angeles CA 90095 USA
| | - Michelle W. Lee
- Department of Bioengineering University of California, Los Angeles Los Angeles CA 90095 USA
| | - Tran Do
- Department of Bioengineering University of California, Los Angeles Los Angeles CA 90095 USA
| | - Ernest Y. Lee
- Department of Bioengineering University of California, Los Angeles Los Angeles CA 90095 USA
| | - Anastassia N. Alexandrova
- Department of Chemistry and Biochemistry University of California, Los Angeles Los Angeles CA 90095 USA
- California Nano Systems Institute University of California, Los Angeles Los Angeles CA 90095 USA
| | - Gerard C. L. Wong
- Department of Bioengineering University of California, Los Angeles Los Angeles CA 90095 USA
- California Nano Systems Institute University of California, Los Angeles Los Angeles CA 90095 USA
| |
Collapse
|
11
|
Trofimenko E, Homma Y, Fukuda M, Widmann C. The endocytic pathway taken by cationic substances requires Rab14 but not Rab5 and Rab7. Cell Rep 2021; 37:109945. [PMID: 34731620 DOI: 10.1016/j.celrep.2021.109945] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 08/23/2021] [Accepted: 10/13/2021] [Indexed: 02/01/2023] Open
Abstract
Endocytosis and endosome dynamics are controlled by proteins of the small GTPase Rab family. Besides possible recycling routes to the plasma membrane and various organelles, previously described endocytic pathways (e.g., clathrin-mediated endocytosis, macropinocytosis, CLIC/GEEC pathway) all appear to funnel the endocytosed material to Rab5-positive early endosomes that then mature into Rab7-positive late endosomes/lysosomes. By studying the uptake of a series of cell-penetrating peptides (CPPs), we identify an endocytic pathway that moves material to nonacidic Lamp1-positive late endosomes. Trafficking via this endocytic route is fully independent of Rab5 and Rab7 but requires the Rab14 protein. The pathway taken by CPPs differs from the conventional Rab5-dependent endocytosis at the stage of vesicle formation already, as it is not affected by a series of compounds that inhibit macropinocytosis or clathrin-mediated endocytosis. The Rab14-dependent pathway is also used by physiological cationic molecules such as polyamines and homeodomains found in homeoproteins.
Collapse
Affiliation(s)
- Evgeniya Trofimenko
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Yuta Homma
- Laboratory of Membrane Trafficking Mechanisms, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Mitsunori Fukuda
- Laboratory of Membrane Trafficking Mechanisms, Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Miyagi, Japan
| | - Christian Widmann
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
12
|
Trofimenko E, Grasso G, Heulot M, Chevalier N, Deriu MA, Dubuis G, Arribat Y, Serulla M, Michel S, Vantomme G, Ory F, Dam LC, Puyal J, Amati F, Lüthi A, Danani A, Widmann C. Genetic, cellular, and structural characterization of the membrane potential-dependent cell-penetrating peptide translocation pore. eLife 2021; 10:69832. [PMID: 34713805 PMCID: PMC8639150 DOI: 10.7554/elife.69832] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 10/28/2021] [Indexed: 12/11/2022] Open
Abstract
Cell-penetrating peptides (CPPs) allow intracellular delivery of bioactive cargo molecules. The mechanisms allowing CPPs to enter cells are ill-defined. Using a CRISPR/Cas9-based screening, we discovered that KCNQ5, KCNN4, and KCNK5 potassium channels positively modulate cationic CPP direct translocation into cells by decreasing the transmembrane potential (Vm). These findings provide the first unbiased genetic validation of the role of Vm in CPP translocation in cells. In silico modeling and live cell experiments indicate that CPPs, by bringing positive charges on the outer surface of the plasma membrane, decrease the Vm to very low values (–150 mV or less), a situation we have coined megapolarization that then triggers formation of water pores used by CPPs to enter cells. Megapolarization lowers the free energy barrier associated with CPP membrane translocation. Using dyes of varying dimensions in CPP co-entry experiments, the diameter of the water pores in living cells was estimated to be 2 (–5) nm, in accordance with the structural characteristics of the pores predicted by in silico modeling. Pharmacological manipulation to lower transmembrane potential boosted CPP cellular internalization in zebrafish and mouse models. Besides identifying the first proteins that regulate CPP translocation, this work characterized key mechanistic steps used by CPPs to cross cellular membranes. This opens the ground for strategies aimed at improving the ability of cells to capture CPP-linked cargos in vitro and in vivo. Before a drug can have its desired effect, it must reach its target tissue or organ, and enter its cells. This is not easy because cells are surrounded by the plasma membrane, a fat-based barrier that separates the cell from its external environment. The plasma membrane contains proteins that act as channels, shuttling specific molecules in and out of the cell, and it also holds charge, with its inside surface being more negatively charged than its outside surface. Cell-penetrating peptides are short sequences of amino acids (the building blocks that form proteins) that carry positive charges. These positive charges allow them to cross the membrane easily, but it is not well understood how. To find out how cell-penetrating peptides cross the membrane, Trofimenko et al. attached them to dyes of different sizes. This revealed that the cell-penetrating peptides enter the cell through temporary holes called water pores, which measure about two nanometres across. The water pores form when the membrane becomes ‘megapolarized’, this is, when the difference in charge between the inside and the outside of the membrane becomes greater than normal. This can happen when the negative charge on the inside surface or the positive charge on the outer surface of the membrane increase. Megapolarization depends on potassium channels, which transport positive potassium ions outside the cell, making the outside of the membrane positive. When cell-penetrating peptides arrive at the outer surface of the cell near potassium channels, they make it even more positive. This increases the charge difference between the inside and the outside of the cell, allowing water pores to form. Once the peptides pass through the pores, the charge difference between the inside and the outside of the cell membrane dissipates, and the pores collapse. Drug developers are experimenting with attaching cell-penetrating peptides to drugs to help them get inside their target cells. Currently there are several experimental medications of this kind in clinical trials. Understanding how these peptides gain entry, and what size of molecule they could carry with them, provides solid ground for further drug development.
Collapse
Affiliation(s)
- Evgeniya Trofimenko
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Gianvito Grasso
- Dalle Molle Institute for Artificial Intelligence Research, Università della Svizzera italiana, Scuola Universitaria Professionale della Svizzera Italiana, Lugano, Switzerland
| | - Mathieu Heulot
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Nadja Chevalier
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Marco A Deriu
- PolitoBIOMed Lab Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Torino, Italy
| | - Gilles Dubuis
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Yoan Arribat
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Marc Serulla
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Sebastien Michel
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Gil Vantomme
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Florine Ory
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Linh Chi Dam
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Julien Puyal
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland.,CURML (University Center of Legal Medicine), Lausanne University Hospital, Lausanne, Switzerland
| | - Francesca Amati
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Anita Lüthi
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Andrea Danani
- Dalle Molle Institute for Artificial Intelligence Research, Università della Svizzera italiana, Scuola Universitaria Professionale della Svizzera Italiana, Lugano, Switzerland
| | - Christian Widmann
- Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
13
|
Georgieva M, Heinonen T, Vitale A, Hargraves S, Causevic S, Pillonel T, Eberl L, Widmann C, Jacquier N. Bacterial surface properties influence the activity of the TAT-RasGAP 317-326 antimicrobial peptide. iScience 2021; 24:102923. [PMID: 34430812 PMCID: PMC8365389 DOI: 10.1016/j.isci.2021.102923] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/05/2021] [Accepted: 07/27/2021] [Indexed: 12/13/2022] Open
Abstract
Antibiotic resistance is an increasing threat for public health, underscoring the need for new antibacterial agents. Antimicrobial peptides (AMPs) represent an alternative to classical antibiotics. TAT-RasGAP317-326 is a recently described AMP effective against a broad range of bacteria, but little is known about the conditions that may influence its activity. Using RNA-sequencing and screening of mutant libraries, we show that Escherichia coli and Pseudomonas aeruginosa respond to TAT-RasGAP317-326 by regulating metabolic and stress response pathways, possibly implicating two-component systems. Our results also indicate that bacterial surface properties, in particular integrity of the lipopolysaccharide layer, influence peptide binding and entry. Finally, we found differences between bacterial species with respect to their rate of resistance emergence against this peptide. Our findings provide the basis for future investigation on the mode of action of TAT-RasGAP317-326, which may help developing antimicrobial treatments based on this peptide.
Collapse
Affiliation(s)
- Maria Georgieva
- Department of Biomedical Sciences, University of Lausanne, Lausanne 1005, Switzerland
| | - Tytti Heinonen
- Institute of Microbiology, Lausanne University Hospital and University of Lausanne, Lausanne 1011, Switzerland
| | - Alessandra Vitale
- Department of Plant and Microbial Biology, University of Zurich, Zurich 8008, Switzerland
| | - Simone Hargraves
- Institute of Microbiology, Lausanne University Hospital and University of Lausanne, Lausanne 1011, Switzerland
| | - Senka Causevic
- Institute of Microbiology, Lausanne University Hospital and University of Lausanne, Lausanne 1011, Switzerland
| | - Trestan Pillonel
- Institute of Microbiology, Lausanne University Hospital and University of Lausanne, Lausanne 1011, Switzerland
| | - Leo Eberl
- Department of Plant and Microbial Biology, University of Zurich, Zurich 8008, Switzerland
| | - Christian Widmann
- Department of Biomedical Sciences, University of Lausanne, Lausanne 1005, Switzerland
- Corresponding author
| | - Nicolas Jacquier
- Institute of Microbiology, Lausanne University Hospital and University of Lausanne, Lausanne 1011, Switzerland
- Corresponding author
| |
Collapse
|
14
|
Conde-Rubio MDC, Mylonas R, Widmann C. The proteolytic landscape of cells exposed to non-lethal stresses is shaped by executioner caspases. Cell Death Discov 2021; 7:164. [PMID: 34226511 PMCID: PMC8257705 DOI: 10.1038/s41420-021-00539-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/26/2021] [Accepted: 05/29/2021] [Indexed: 02/06/2023] Open
Abstract
Cells are in constant adaptation to environmental changes to insure their proper functioning. When exposed to stresses, cells activate specific pathways to elicit adaptive modifications. Those changes can be mediated by selective modulation of gene and protein expression as well as by post-translational modifications, such as phosphorylation and proteolytic processing. Protein cleavage, as a controlled and limited post-translational modification, is involved in diverse physiological processes such as the maintenance of protein homeostasis, activation of repair pathways, apoptosis and the regulation of proliferation. Here we assessed by quantitative proteomics the proteolytic landscape in two cell lines subjected to low cisplatin concentrations used as a mild non-lethal stress paradigm. This landscape was compared to the one obtained in the same cells stimulated with cisplatin concentrations inducing apoptosis. These analyses were performed in wild-type cells and in cells lacking the two main executioner caspases: caspase-3 and caspase-7. Ninety-two proteins were found to be cleaved at one or a few sites (discrete cleavage) in low stress conditions compared to four hundred and fifty-three in apoptotic cells. Many of the cleaved proteins in stressed cells were also found to be cleaved in apoptotic conditions. As expected, ~90% of the cleavage events were dependent on caspase-3/caspase-7 in apoptotic cells. Strikingly, upon exposure to non-lethal stresses, no discrete cleavage was detected in cells lacking caspase-3 and caspase-7. This indicates that the proteolytic landscape in stressed viable cells fully depends on the activity of executioner caspases. These results suggest that the so-called executioner caspases fulfill important stress adaptive responses distinct from their role in apoptosis. Mass spectrometry data are available via ProteomeXchange with identifier PXD023488.
Collapse
Affiliation(s)
| | - Roman Mylonas
- Protein Analysis Facility, University of Lausanne, Génopode, Lausanne, Switzerland.,SIB Swiss Institute of Bioinformatics, Amphipole, Lausanne, Switzerland
| | - Christian Widmann
- Department of Biomedical Sciences, University of Lausanne, Bugnon 7, Lausanne, Switzerland.
| |
Collapse
|
15
|
|
16
|
Kong X, Xu J, Yang X, Zhai Y, Ji J, Zhai G. Progress in tumour-targeted drug delivery based on cell-penetrating peptides. J Drug Target 2021; 30:46-60. [PMID: 33944641 DOI: 10.1080/1061186x.2021.1920026] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Since the discovery of cell-penetrating peptides (CPP) in the 1980s, they have played a unique role in various fields owing to their excellent and unique cell membrane penetration function. In particular, in the treatment of tumours, CPPS have been used to deliver several types of 'cargos' to cancer cells. To address the insufficient targeting ability, non-selectivity, and blood instability, activatable cell-penetrating peptides, which can achieve targeted drug delivery in tumour treatment, enhance curative effects, and reduce toxicity have been developed. This study reviews the application of different cell-penetrating peptides in tumour-targeted delivery, overcoming multidrug resistance, organelle targeting, tumour imaging, and diagnosis, and summarises the different mechanisms of activatable cell-penetrating peptides in detail.
Collapse
Affiliation(s)
- Xinru Kong
- Key Laboratory of Chemical Biology, Department of Pharmaceutics, School of Pharmaceutical Sciences, Ministry of Education, Shandong University, Jinan, China
| | - Jiangkang Xu
- Key Laboratory of Chemical Biology, Department of Pharmaceutics, School of Pharmaceutical Sciences, Ministry of Education, Shandong University, Jinan, China
| | - Xiaoye Yang
- Key Laboratory of Chemical Biology, Department of Pharmaceutics, School of Pharmaceutical Sciences, Ministry of Education, Shandong University, Jinan, China
| | - Yujia Zhai
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, UT, USA
| | - Jianbo Ji
- Key Laboratory of Chemical Biology, Department of Pharmaceutics, School of Pharmaceutical Sciences, Ministry of Education, Shandong University, Jinan, China
| | - Guangxi Zhai
- Key Laboratory of Chemical Biology, Department of Pharmaceutics, School of Pharmaceutical Sciences, Ministry of Education, Shandong University, Jinan, China
| |
Collapse
|
17
|
Heinonen T, Hargraves S, Georgieva M, Widmann C, Jacquier N. The antimicrobial peptide TAT-RasGAP 317-326 inhibits the formation and expansion of bacterial biofilms in vitro. J Glob Antimicrob Resist 2021; 25:227-231. [PMID: 33852935 DOI: 10.1016/j.jgar.2021.03.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 03/03/2021] [Accepted: 03/11/2021] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVES Biofilms are structured aggregates of bacteria embedded in a self-produced matrix that develop in diverse ecological niches. Pathogenic bacteria can form biofilms on surfaces and in tissues, causing nosocomial and chronic infections that are difficult to treat. While antibiotics are largely inefficient in limiting biofilm formation and expansion, antimicrobial peptides (AMPs) are emerging as alternative antibiofilm treatments. In this study, we explore the effect of the newly described AMP TAT-RasGAP317-326 on Acinetobacter baumannii, Pseudomonas aeruginosa and Staphylococcus aureus biofilms. METHODS Efficiency of TAT-RasGAP317-326 on biofilms was tested in vitro. Both viability of bacteria contained in the biofilm as well as biomass of the biofilm were quantified using resazurin and crystal violet staining, respectively. The antibiofilm effect of TAT-RasGAP317-326 was compared with a selection of classical antibiotics and AMPs. RESULTS We observe that TAT-RasGAP317-326 inhibits biofilm formation at concentrations equivalent or two times greater than the minimum inhibitory concentration (MIC) of planktonic bacteria. Moreover, TAT-RasGAP317-326 limits the expansion of A. baumannii and P. aeruginosa established biofilms at twice the concentration inhibiting biofilm formation. CONCLUSION These results underscore the potential use of TAT-RasGAP317-326 against biofilms and encourage further studies in the development of AMPs to treat biofilm-related infections.
Collapse
Affiliation(s)
- Tytti Heinonen
- Institute of Microbiology, Lausanne University Hospital and University of Lausanne, Rue du Bugnon 48, 1011 Lausanne, Switzerland
| | - Simone Hargraves
- Institute of Microbiology, Lausanne University Hospital and University of Lausanne, Rue du Bugnon 48, 1011 Lausanne, Switzerland
| | - Maria Georgieva
- Department of Biomedical Sciences, University of Lausanne, Rue du Bugnon 7, 1005 Lausanne, Switzerland
| | - Christian Widmann
- Department of Biomedical Sciences, University of Lausanne, Rue du Bugnon 7, 1005 Lausanne, Switzerland
| | - Nicolas Jacquier
- Institute of Microbiology, Lausanne University Hospital and University of Lausanne, Rue du Bugnon 48, 1011 Lausanne, Switzerland.
| |
Collapse
|