1
|
Chen T, Wang Y, Chi X, Xiong L, Lu P, Wang X, Chen Y, Luo Q, Shen P, Xiao Y. Genetic, virulence, and antimicrobial resistance characteristics associated with distinct morphotypes in ST11 carbapenem-resistant Klebsiella pneumoniae. Virulence 2024; 15:2349768. [PMID: 38736039 PMCID: PMC11093053 DOI: 10.1080/21505594.2024.2349768] [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: 11/27/2023] [Accepted: 04/16/2024] [Indexed: 05/14/2024] Open
Abstract
ST11 is the most common lineage among carbapenem-resistant Klebsiella pneumoniae (CRKP) infections in Asia. Diverse morphotypes resulting from genetic mutations are associated with significant differences in microbial characteristics among K. pneumoniae isolates. Here, we investigated the genetic determinants and critical characteristics associated with distinct morphotypes of ST11 CRKP. An ST11-KL47 CRKP isolate carrying a pLVPK-like virulence plasmid was isolated from a patient with a bloodstream infection; the isolate had the "mcsw" morphotype. Two distinct morphotypes ("ntrd" and "msdw") were derived from this strain during in vitro passage. Whole genome sequencing was used to identify mutations that cause the distinct morphotypes of ST11 CRKP. Transmission electron microscopy, antimicrobial susceptibility tests, growth assays, biofilm formation, virulence assays, membrane permeability assays, and RNA-seq analysis were used to investigate the specific characteristics associated with different morphotypes of ST11 CRKP. Compared with the parental mcsw morphotype, the ntrd morphotype resulted from mutation of genes involved in capsular polysaccharide biosynthesis (wza, wzc, and wbaP), a result validated by gene knockout experiments. This morphotype showed capsule deficiency and lower virulence potential, but higher biofilm production. By contrast, the msdw morphotype displayed competition deficiency and increased susceptibility to chlorhexidine and polymyxin B. Further analyses indicated that these characteristics were caused by interruption of the sigma factor gene rpoN by insertion mutations and deletion of the rpoN gene, which attenuated membrane integrity presumably by downregulating the phage shock protein operon. These data expand current understanding of genetic, virulence, and antimicrobial resistance characteristics associated with distinct morphotypes in ST11 CRKP.
Collapse
Affiliation(s)
- Tao Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yuan Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiaohui Chi
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Luying Xiong
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ping Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xueting Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yunbo Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qixia Luo
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ping Shen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong, China
| | - Yonghong Xiao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong, China
| |
Collapse
|
2
|
Weerakoon D, Marzinek JK, Pedebos C, Bond PJ, Khalid S. Polymyxin B1 in the Escherichia coli inner membrane: A complex story of protein and lipopolysaccharide-mediated insertion. J Biol Chem 2024; 300:107754. [PMID: 39260694 PMCID: PMC11497408 DOI: 10.1016/j.jbc.2024.107754] [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: 02/10/2024] [Revised: 08/21/2024] [Accepted: 08/29/2024] [Indexed: 09/13/2024] Open
Abstract
The rise in multi-drug resistant Gram-negative bacterial infections has led to an increased need for "last-resort" antibiotics such as polymyxins. However, the emergence of polymyxin-resistant strains threatens to bring about a post-antibiotic era. Thus, there is a need to develop new polymyxin-based antibiotics, but a lack of knowledge of the mechanism of action of polymyxins hinders such efforts. It has recently been suggested that polymyxins induce cell lysis of the Gram-negative bacterial inner membrane (IM) by targeting trace amounts of lipopolysaccharide (LPS) localized there. We use multiscale molecular dynamics (MD), including long-timescale coarse-grained (CG) and all-atom (AA) simulations, to investigate the interactions of polymyxin B1 (PMB1) with bacterial IM models containing phospholipids (PLs), small quantities of LPS, and IM proteins. LPS was observed to (transiently) phase separate from PLs at multiple LPS concentrations, and associate with proteins in the IM. PMB1 spontaneously inserted into the IM and localized at the LPS-PL interface, where it cross-linked lipid headgroups via hydrogen bonds, sampling a wide range of interfacial environments. In the presence of membrane proteins, a small number of PMB1 molecules formed interactions with them, in a manner that was modulated by local LPS molecules. Electroporation-driven translocation of PMB1 via water-filled pores was favored at the protein-PL interface, supporting the 'destabilizing' role proteins may have within the IM. Overall, this in-depth characterization of PMB1 modes of interaction reveals how small amounts of mislocalized LPS may play a role in pre-lytic targeting and provides insights that may facilitate rational improvement of polymyxin-based antibiotics.
Collapse
Affiliation(s)
- Dhanushka Weerakoon
- School of Chemistry, University of Southampton, Southampton, UK; Bioinformatics Institute (BII), Agency for Science, Technology and Research (A∗STAR), Singapore, Republic of Singapore
| | - Jan K Marzinek
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A∗STAR), Singapore, Republic of Singapore
| | - Conrado Pedebos
- Department of Biochemistry, University of Oxford, Porto Alegre, UK; Programa de Pós-Graduação em Biociências (PPGBio), Universidade Federal de Ciências da Saudé de Porto Alegre - UFCSPA, Brazil
| | - Peter J Bond
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A∗STAR), Singapore, Republic of Singapore; Department of Biological Sciences, National University of Singapore, Singapore, Republic of Singapore.
| | - Syma Khalid
- Department of Biochemistry, University of Oxford, Porto Alegre, UK.
| |
Collapse
|
3
|
Viravathana P, Burbank LP, Jablonska B, Sun Q, Roper MC. A membrane localized RTX-like protein mediates physiochemical properties of the Pantoea stewartii subsp. stewartii cell envelope that impact surface adhesion, cell surface hydrophobicity and plant colonization. BMC Microbiol 2024; 24:369. [PMID: 39342134 PMCID: PMC11438254 DOI: 10.1186/s12866-024-03516-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 09/11/2024] [Indexed: 10/01/2024] Open
Abstract
Pantoea stewartii subsp. stewartii (Pnss), is the bacterial causal agent of Stewart's wilt of sweet corn. Disease symptoms include systemic wilting and foliar, water-soaked lesions. A Repeat-in-toxin (RTX)-like protein, RTX2, causes cell leakage and collapse in the leaf apoplast of susceptible corn varieties and is a primary mediator of water-soaked lesion formation in the P. stewartii-sweet corn pathosystem. RTX toxins comprise a large family of proteins, which are widely distributed among Gram-negative bacteria. These proteins are generally categorized as cellulolysins, but the Biofilm-Associated Proteins (Bap) subfamily of RTX toxins are implicated in surface adhesion and other biofilm behaviors. The Pnss RTX2 is most phylogenetically related to other Bap proteins suggesting that Pnss RTX2 plays a dual role in adhesion to host surfaces in addition to mediating the host cell lysis that leads to water-soaked lesion formation. Here we demonstrated that RTX2 localizes to the bacterial cell envelope and influences physiochemical properties of the bacterial cell envelope that impact bacterial cell length, cell envelope integrity and overall cellular hydrophobicity. Interestingly, the role of RTX2 as an adhesin was only evident in absence of exopolysaccharide (EPS) production suggesting that RTX2 plays a role as an adhesin early in biofilm development before EPS production is fully induced. However, deletion of rtx2 severely impacted Pnss' colonization of the xylem suggesting that the dual role of RTX2 as a cytolysin and adhesin is a mechanism that links the apoplastic water-soaked lesion phase of infection to the wilting phase of the infection in the xylem.
Collapse
Affiliation(s)
- Polrit Viravathana
- Department of Microbiology and Plant Pathology, University of California, Riverside, CA, 92521, USA
| | - Lindsey P Burbank
- USDA Agricultural Research Service, San Joaquin Valley Agricultural Sciences Center, Parlier, CA, 93648, USA
| | - Barbara Jablonska
- Department of Microbiology and Plant Pathology, University of California, Riverside, CA, 92521, USA
| | - Qiang Sun
- Department of Biology, University of Wisconsin, Stevens Point, WI, 54481, USA
| | - M Caroline Roper
- Department of Microbiology and Plant Pathology, University of California, Riverside, CA, 92521, USA.
| |
Collapse
|
4
|
Chen J, Wang W, Hu X, Yue Y, Lu X, Wang C, Wei B, Zhang H, Wang H. Medium-sized peptides from microbial sources with potential for antibacterial drug development. Nat Prod Rep 2024; 41:1235-1263. [PMID: 38651516 DOI: 10.1039/d4np00002a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
Covering: 1993 to the end of 2022As the rapid development of antibiotic resistance shrinks the number of clinically available antibiotics, there is an urgent need for novel options to fill the existing antibiotic pipeline. In recent years, antimicrobial peptides have attracted increased interest due to their impressive broad-spectrum antimicrobial activity and low probability of antibiotic resistance. However, macromolecular antimicrobial peptides of plant and animal origin face obstacles in antibiotic development because of their extremely short elimination half-life and poor chemical stability. Herein, we focus on medium-sized antibacterial peptides (MAPs) of microbial origin with molecular weights below 2000 Da. The low molecular weight is not sufficient to form complex protein conformations and is also associated to a better chemical stability and easier modifications. Microbially-produced peptides are often composed of a variety of non-protein amino acids and terminal modifications, which contribute to improving the elimination half-life of compounds. Therefore, MAPs have great potential for drug discovery and are likely to become key players in the development of next-generation antibiotics. In this review, we provide a detailed exploration of the modes of action demonstrated by 45 MAPs and offer a concise summary of the structure-activity relationships observed in these MAPs.
Collapse
Affiliation(s)
- Jianwei Chen
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Wei Wang
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Xubin Hu
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yujie Yue
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Xingyue Lu
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Chenjie Wang
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Bin Wei
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Huawei Zhang
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| | - Hong Wang
- College of Pharmaceutical Science & Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Hangzhou 310014, China
| |
Collapse
|
5
|
Královič-Kanjaková N, Asi Shirazi A, Hubčík L, Klacsová M, Keshavarzi A, Martínez JC, Combet S, Teixeira J, Uhríková D. Polymyxin B-Enriched Exogenous Lung Surfactant: Thermodynamics and Structure. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:6847-6861. [PMID: 38501650 DOI: 10.1021/acs.langmuir.3c03746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/20/2024]
Abstract
The use of an exogenous pulmonary surfactant (EPS) to deliver other relevant drugs to the lungs is a promising strategy for combined therapy. We evaluated the interaction of polymyxin B (PxB) with a clinically used EPS, the poractant alfa Curosurf (PSUR). The effect of PxB on the protein-free model system (MS) composed of four phospholipids (diC16:0PC/16:0-18:1PC/16:0-18:2PC/16:0-18:1PG) was examined in parallel to distinguish the specificity of the composition of PSUR. We used several experimental techniques (differential scanning calorimetry, small- and wide-angle X-ray scattering, small-angle neutron scattering, fluorescence spectroscopy, and electrophoretic light scattering) to characterize the binding of PxB to both EPS. Electrostatic interactions PxB-EPS are dominant. The results obtained support the concept of cationic PxB molecules lying on the surface of the PSUR bilayer, strengthening the multilamellar structure of PSUR as derived from SAXS and SANS. A protein-free MS mimics a natural EPS well but was found to be less resistant to penetration of PxB into the lipid bilayer. PxB does not affect the gel-to-fluid phase transition temperature, Tm, of PSUR, while Tm increased by ∼+ 2 °C in MS. The decrease of the thickness of the lipid bilayer (dL) of PSUR upon PxB binding is negligible. The hydrophobic tail of the PxB molecule does not penetrate the bilayer as derived from SANS data analysis and changes in lateral pressure monitored by excimer fluorescence at two depths of the hydrophobic region of the bilayer. Changes in dL of protein-free MS show a biphasic dependence on the adsorbed amount of PxB with a minimum close to the point of electroneutrality of the mixture. Our results do not discourage the concept of a combined treatment with PxB-enriched Curosurf. However, the amount of PxB must be carefully assessed (less than 5 wt % relative to the mass of the surfactant) to avoid inversion of the surface charge of the membrane.
Collapse
Affiliation(s)
- Nina Královič-Kanjaková
- Department of Physical Chemistry of Drugs, Faculty of Pharmacy, Comenius University Bratislava, 832 32 Bratislava, Slovakia
| | - Ali Asi Shirazi
- Department of Physical Chemistry of Drugs, Faculty of Pharmacy, Comenius University Bratislava, 832 32 Bratislava, Slovakia
| | - Lukáš Hubčík
- Department of Physical Chemistry of Drugs, Faculty of Pharmacy, Comenius University Bratislava, 832 32 Bratislava, Slovakia
| | - Mária Klacsová
- Department of Physical Chemistry of Drugs, Faculty of Pharmacy, Comenius University Bratislava, 832 32 Bratislava, Slovakia
| | - Atoosa Keshavarzi
- Department of Physical Chemistry of Drugs, Faculty of Pharmacy, Comenius University Bratislava, 832 32 Bratislava, Slovakia
| | | | - Sophie Combet
- Laboratoire Léon-Brillouin (LLB), UMR12 CEA, CNRS, Université Paris-Saclay, F-91191 Gif-sur-Yvette CEDEX, France
| | - José Teixeira
- Laboratoire Léon-Brillouin (LLB), UMR12 CEA, CNRS, Université Paris-Saclay, F-91191 Gif-sur-Yvette CEDEX, France
| | - Daniela Uhríková
- Department of Physical Chemistry of Drugs, Faculty of Pharmacy, Comenius University Bratislava, 832 32 Bratislava, Slovakia
| |
Collapse
|
6
|
Steigenberger J, Mergen C, De Roo V, Geudens N, Martins JC, Heerklotz H. The effect of membrane thickness on the membrane permeabilizing activity of the cyclic lipopeptide tolaasin II. Front Mol Biosci 2022; 9:1064742. [PMID: 36619163 PMCID: PMC9817028 DOI: 10.3389/fmolb.2022.1064742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Accepted: 11/21/2022] [Indexed: 12/25/2022] Open
Abstract
Tolaasin II is an amphiphilic, membrane-active, cyclic lipopeptide produced by Pseudomonas tolaasii and is responsible for brown blotch disease in mushroom. To better understand the mode of action and membrane selectivity of tolaasin II and related lipopeptides, its permeabilizing effect on liposomes of different membrane thickness was characterized. An equi-activity analysis served to distinguish between the effects of membrane partitioning and the intrinsic activity of the membrane-bound peptide. It was found that thicker membranes require higher local peptide concentrations to become leaky. More specifically, the mole ratio of membrane-bound peptide per lipid needed to induce 50% leakage of calcein within 1 h, Re 50, increased monotonically with membrane thickness from 0.0016 for the 14:1 to 0.0070 for the 20:1 lipid-chains. Moreover, fast but limited leakage kinetics in the low-lipid regime were observed implying a mode of action based on membrane asymmetry stress in this time and concentration window. While the assembly of the peptide to oligomeric pores of defined length along the bilayer z-axis can in principle explain inhibition by increasing membrane thickness, it cannot account for the observed limited leakage. Therefore, reduced intrinsic membrane-permeabilizing activity with increasing membrane thickness is attributed here to the increased mechanical strength and order of thicker membranes.
Collapse
Affiliation(s)
- Jessica Steigenberger
- Department of Pharmaceutics, Institute of Pharmaceutical Sciences, University of Freiburg, Freiburg, Germany,*Correspondence: Jessica Steigenberger, ; Heiko Heerklotz,
| | - Catherine Mergen
- Department of Pharmaceutics, Institute of Pharmaceutical Sciences, University of Freiburg, Freiburg, Germany
| | - Vic De Roo
- NMR and Structure Analysis Research Group, Department of Organic and Macromolecular Chemistry, Ghent University, Ghent, Belgium
| | - Niels Geudens
- NMR and Structure Analysis Research Group, Department of Organic and Macromolecular Chemistry, Ghent University, Ghent, Belgium
| | - José C. Martins
- NMR and Structure Analysis Research Group, Department of Organic and Macromolecular Chemistry, Ghent University, Ghent, Belgium
| | - Heiko Heerklotz
- Department of Pharmaceutics, Institute of Pharmaceutical Sciences, University of Freiburg, Freiburg, Germany,Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada,Signaling Research Centers BIOSS and CIBSS, University of Freiburg, Freiburg, Germany,*Correspondence: Jessica Steigenberger, ; Heiko Heerklotz,
| |
Collapse
|
7
|
Dubashynskaya NV, Bokatyi AN, Dobrodumov AV, Kudryavtsev IV, Trulioff AS, Rubinstein AA, Aquino AD, Dubrovskii YA, Knyazeva ES, Demyanova EV, Nashchekina YA, Skorik YA. Succinyl Chitosan-Colistin Conjugates as Promising Drug Delivery Systems. Int J Mol Sci 2022; 24:ijms24010166. [PMID: 36613610 PMCID: PMC9820547 DOI: 10.3390/ijms24010166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/11/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
The growth of microbial multidrug resistance is a problem in modern clinical medicine. Chemical modification of active pharmaceutical ingredients is an attractive strategy to improve their biopharmaceutical properties by increasing bioavailability and reducing drug toxicity. Conjugation of antimicrobial drugs with natural polysaccharides provides high efficiency of these systems due to targeted delivery, controlled drug release and reduced toxicity. This paper reports a two-step synthesis of colistin conjugates (CT) with succinyl chitosan (SucCS); first, we modified chitosan with succinyl anhydride to introduce a carboxyl function into the polymer molecule, which was then used for chemical grafting with amino groups of the peptide antibiotic CT using carbodiimide chemistry. The resulting polymeric delivery systems had a degree of substitution (DS) by CT of 3-8%, with conjugation efficiencies ranging from 54 to 100% and CT contents ranging from 130-318 μg/mg. The size of the obtained particles was 100-200 nm, and the ζ-potential varied from -22 to -28 mV. In vitro release studies at pH 7.4 demonstrated ultra-slow hydrolysis of amide bonds, with a CT release of 0.1-0.5% after 12 h; at pH 5.2, the hydrolysis rate slightly increased; however, it remained extremely low (1.5% of CT was released after 12 h). The antimicrobial activity of the conjugates depended on the DS. At DS 8%, the minimum inhibitory concentration (MIC) of the conjugate was equal to the MIC of native CT (1 µg/mL); at DS of 3 and 5%, the MIC increased 8-fold. In addition, the developed systems reduced CT nephrotoxicity by 20-60%; they also demonstrated the ability to reduce bacterial lipopolysaccharide-induced inflammation in vitro. Thus, these promising CT-SucCS conjugates are prospective for developing safe and effective nanoantibiotics.
Collapse
Affiliation(s)
- Natallia V. Dubashynskaya
- Institute of Macromolecular Compounds of the Russian Academy of Sciences, Bolshoi VO 31, 199004 St. Petersburg, Russia
| | - Anton N. Bokatyi
- Institute of Macromolecular Compounds of the Russian Academy of Sciences, Bolshoi VO 31, 199004 St. Petersburg, Russia
| | - Anatoliy V. Dobrodumov
- Institute of Macromolecular Compounds of the Russian Academy of Sciences, Bolshoi VO 31, 199004 St. Petersburg, Russia
| | - Igor V. Kudryavtsev
- Institute of Experimental Medicine, Akademika Pavlova 12, 197376 St. Petersburg, Russia
| | - Andrey S. Trulioff
- Institute of Experimental Medicine, Akademika Pavlova 12, 197376 St. Petersburg, Russia
| | - Artem A. Rubinstein
- Institute of Experimental Medicine, Akademika Pavlova 12, 197376 St. Petersburg, Russia
| | - Arthur D. Aquino
- Almazov National Medical Research Centre, Akkuratova 2, 197341 St. Petersburg, Russia
| | | | - Elena S. Knyazeva
- State Research Institute of Highly Pure Biopreparations, Pudozhsakya 7, 197110 St Petersburg, Russia
| | - Elena V. Demyanova
- State Research Institute of Highly Pure Biopreparations, Pudozhsakya 7, 197110 St Petersburg, Russia
| | - Yuliya A. Nashchekina
- Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky 4, 194064 St. Petersburg, Russia
| | - Yury A. Skorik
- Institute of Macromolecular Compounds of the Russian Academy of Sciences, Bolshoi VO 31, 199004 St. Petersburg, Russia
- Correspondence:
| |
Collapse
|
8
|
Krivić H, Himbert S, Sun R, Feigis M, Rheinstädter MC. Erythro-PmBs: A Selective Polymyxin B Delivery System Using Antibody-Conjugated Hybrid Erythrocyte Liposomes. ACS Infect Dis 2022; 8:2059-2072. [PMID: 36173819 DOI: 10.1021/acsinfecdis.2c00017] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
As a result of the growing worldwide antibiotic resistance crisis, many currently existing antibiotics have become ineffective due to bacteria developing resistive mechanisms. There are a limited number of potent antibiotics that are successful at suppressing microbial growth, such as polymyxin B (PmB); however, these are often deemed as a last resort due to their toxicity. We present a novel PmB delivery system constructed by conjugating hybrid erythrocyte liposomes with antibacterial antibodies to combine a high loading efficiency with guided delivery. The retention of PmB is enhanced by incorporating negatively charged lipids into the red blood cells' cytoplasmic membrane (RBCcm). Anti-Escherichia coli antibodies are attached to these hybrid erythrocyte liposomes by the inclusion of DSPE-PEG maleimide linkers. We show that these erythro-PmBs have a loading efficiency of ∼90% and are effective in delivering PmB to E. coli, with values for the minimum inhibitory concentration (MIC) being comparable to those of free PmB. The MIC values for Klebsiella aerogenes, however, significantly increased well beyond the resistant breakpoint, indicating that the inclusion of the anti-E. coli antibodies enables the erythro-PmBs to selectively deliver antibiotics to specific targets.
Collapse
Affiliation(s)
- Hannah Krivić
- Department of Physics and Astronomy, McMaster University, HamiltonL8S 4M1, Ontario, Canada.,Origins Institute, McMaster University, HamiltonL8S 4M1, Ontario, Canada
| | - Sebastian Himbert
- Department of Physics and Astronomy, McMaster University, HamiltonL8S 4M1, Ontario, Canada.,Origins Institute, McMaster University, HamiltonL8S 4M1, Ontario, Canada
| | - Ruthie Sun
- Department of Physics and Astronomy, McMaster University, HamiltonL8S 4M1, Ontario, Canada.,Origins Institute, McMaster University, HamiltonL8S 4M1, Ontario, Canada
| | - Michal Feigis
- Department of Physics and Astronomy, McMaster University, HamiltonL8S 4M1, Ontario, Canada.,Origins Institute, McMaster University, HamiltonL8S 4M1, Ontario, Canada
| | - Maikel C Rheinstädter
- Department of Physics and Astronomy, McMaster University, HamiltonL8S 4M1, Ontario, Canada.,Origins Institute, McMaster University, HamiltonL8S 4M1, Ontario, Canada
| |
Collapse
|
9
|
Synergistic Membrane Disturbance Improves the Antibacterial Performance of Polymyxin B. Polymers (Basel) 2022; 14:polym14204316. [PMID: 36297894 PMCID: PMC9611124 DOI: 10.3390/polym14204316] [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: 09/12/2022] [Revised: 09/29/2022] [Accepted: 10/09/2022] [Indexed: 01/24/2023] Open
Abstract
Drug-resistant Gram-negative bacteria pose a serious threat to public health, and polymyxin B (PMB) is clinically used as a last-line therapy for the treatment of infections caused by these pathogens. However, the appearance of PMB resistance calls for an effort to develop new approaches to improve its antibacterial performance. In this work, a new type of nanocomposite, composed of PMB molecules being chemically decorated on the surface of graphene oxide (GO) nanosheets, was designed, which showed potent antibacterial ability through synergistically and physically disturbing the bacterial membrane. The as-fabricated PMB@GO nanocomposites demonstrated an enhanced bacterial-killing efficiency, with a minimum inhibitory concentration (MIC) value half of that of free PMB (with an MIC value as low as 0.5 μg mL-1 over Escherichia coli), and a bacterial viability less than one fourth of that of PMB (with a bacterial reduction of 60% after 3 h treatment, and 90% after 6 h incubation). Furthermore, the nanocomposite displayed moderate cytotoxicity or hemolysis effect, with cellular viabilities over 85% at concentrations up to 16 times the MIC value. Studies on antibacterial mechanism revealed that the synergy between PMB molecules and GO nanosheets greatly facilitated the vertical insertion of the nanocomposite into the lipid membrane, leading to membrane disturbance and permeabilization. Our results demonstrate a physical mechanism for improving the antibacterial performance of PMB and developing advanced antibacterial agents for better clinic uses.
Collapse
|
10
|
Constitutive Phenotypic Modification of Lipid A in Clinical Acinetobacter baumannii Isolates. Microbiol Spectr 2022; 10:e0129522. [PMID: 35861511 PMCID: PMC9431647 DOI: 10.1128/spectrum.01295-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
The degree of polymyxin B (PMB) resistance was measured in 40 clinical Acinetobacter baumannii isolates obtained from health care facilities. All of the tested isolates possessed a multidrug-resistant (MDR) phenotype against four classes of antibiotics (meropenem, doxycycline, gentamicin, and erythromycin), except for PMB. The blaOXA-23 gene was detected throughout the genetic analysis and experimental assay, indicating that all of the MDR strains were carbapenem-resistant A. baumannii strains. Multilocus sequence typing-based genotyping revealed that nine selected strains belonged to the international clone II lineage. When matrix-assisted laser desorption ionization–time of flight mass spectrometry was performed, intrinsic lipid A modification by phosphoethanolamine (PEtN) incorporation was noticeable only in the PMB-resistant (PMBR) strains. However, the presence of hexa- and penta-acylated lipid A due to the loss of the laurate (C12) acyl chain was noted in all PMB-susceptible strains but not in the PMBR strains. The reduction of negative surface charges in the PMBR strains was assessed by zeta potential analysis. Fluorescence imaging using dansyl-PMB revealed that, in the PMBR strains, PMB was less likely to bind to the cell surface. IMPORTANCE The widespread presence of MDR pathogens, including A. baumannii, is causing serious hospital-acquired infections worldwide. Extensive surveillance of MDR clinical A. baumannii isolates has been conducted, but the underlying mechanisms for their development of MDR phenotypes are often neglected. Either lipid A modification or loss of lipopolysaccharide in Gram-negative bacteria leads to PMBR phenotypes. The prevalence of intrinsic lipid A modification in PMBR clinical strains was attributed to high levels of basal expression of pmrC and eptA-1. Our findings suggest that new therapeutic strategies are warranted to combat MDR pathogens due to the emergence of many PMBR clinical strains.
Collapse
|
11
|
Guo H, Zhao T, Huang C, Chen J. The Role of the Two-Component System PhoP/PhoQ in Intrinsic Resistance of Yersinia enterocolitica to Polymyxin. Front Microbiol 2022; 13:758571. [PMID: 35222323 PMCID: PMC8867023 DOI: 10.3389/fmicb.2022.758571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 01/06/2022] [Indexed: 12/02/2022] Open
Abstract
Polymyxin is the "last resort" of antibiotics. The self-induced resistance to polymyxin in Gram-negative bacteria could be mediated by lipopolysaccharide (LPS) modification, which is regulated by the two-component system, PhoP/PhoQ. Yersinia enterocolitica is a common foodborne pathogen. However, PhoP/PhoQ has not been thoroughly studied in Y. enterocolitica. In this study, the functions of PhoP/PhoQ in Y. enterocolitica intrinsic resistance were investigated. The resistance of Y. enterocolitica was found to decrease with the deletion of PhoP/PhoQ. Further, PhoP/PhoQ was found to play an important role in maintaining membrane permeability, intercellular metabolism, and reducing membrane depolarization. Based on subsequent studies, the binding ability of polymyxin to Y. enterocolitica was decreased by the modification of LPS with structures, such as L-Ara4N and palmitate. Analysis of the gene transcription levels revealed that the LPS modification genes, pagP and arn operon, were downregulated with the deletion of PhoP/PhoQ in Y. enterocolitica during exposure to polymyxin. In addition, pmrA, pmrB, and eptA were downregulated in the mutants compared with the wild-type strain. Such findings demonstrate that PhoP/PhoQ contributes to the intrinsic resistance of Y. enterocolitica toward polymyxins. LPS modification with L-Ara4N or palmitate is mainly responsible for the resistance of Y. enterocolitica to polymyxins. The transcription of genes related to LPS modification and PmrA/PmrB can be both affected by PhoP/PhoQ in Y. enterocolitica. This study adds to current knowledge regarding the role of PhoP/PhoQ in intrinsic resistance of Y. enterocolitica to polymyxin.
Collapse
Affiliation(s)
| | | | | | - Jingyu Chen
- Beijing Laboratory for Food Quality and Safety, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| |
Collapse
|
12
|
Carey AB, Ashenden A, Köper I. Model architectures for bacterial membranes. Biophys Rev 2022; 14:111-143. [PMID: 35340604 PMCID: PMC8921416 DOI: 10.1007/s12551-021-00913-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/14/2021] [Indexed: 02/06/2023] Open
Abstract
The complex composition of bacterial membranes has a significant impact on the understanding of pathogen function and their development towards antibiotic resistance. In addition to the inherent complexity and biosafety risks of studying biological pathogen membranes, the continual rise of antibiotic resistance and its significant economical and clinical consequences has motivated the development of numerous in vitro model membrane systems with tuneable compositions, geometries, and sizes. Approaches discussed in this review include liposomes, solid-supported bilayers, and computational simulations which have been used to explore various processes including drug-membrane interactions, lipid-protein interactions, host-pathogen interactions, and structure-induced bacterial pathogenesis. The advantages, limitations, and applicable analytical tools of all architectures are summarised with a perspective for future research efforts in architectural improvement and elucidation of resistance development strategies and membrane-targeting antibiotic mechanisms. Supplementary Information The online version contains supplementary material available at 10.1007/s12551-021-00913-7.
Collapse
Affiliation(s)
- Ashley B. Carey
- Institute for Nanoscale Science and Technology, College for Science and Engineering, Flinders University, Adelaide, SA 5042 Australia
| | - Alex Ashenden
- Institute for Nanoscale Science and Technology, College for Science and Engineering, Flinders University, Adelaide, SA 5042 Australia
| | - Ingo Köper
- Institute for Nanoscale Science and Technology, College for Science and Engineering, Flinders University, Adelaide, SA 5042 Australia
| |
Collapse
|
13
|
Silva KED, Rossato L, Leite AF, Simionatto S. Overview of polymyxin resistance in Enterobacteriaceae. Rev Soc Bras Med Trop 2022; 55:e0349. [PMID: 35239902 PMCID: PMC8909443 DOI: 10.1590/0037-8682-0349-2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 11/12/2021] [Indexed: 11/21/2022] Open
Abstract
Polymyxin antibiotics are disfavored owing to their potential clinical toxicity, especially nephrotoxicity. However, the dry antibiotic development pipeline, together with the increasing global prevalence of infections caused by multidrug-resistant (MDR) gram-negative bacteria, have renewed clinical interest in these polypeptide antibiotics. This review highlights the current information regarding the mechanisms of resistance to polymyxins and their molecular epidemiology. Knowledge of the resistance mechanisms and epidemiology of these pathogens is critical for the development of novel antibacterial agents and rapid treatment choices.
Collapse
|
14
|
Panjla A, Kaul G, Chopra S, Titz A, Verma S. Short Peptides and Their Mimetics as Potent Antibacterial Agents and Antibiotic Adjuvants. ACS Chem Biol 2021; 16:2731-2745. [PMID: 34779605 DOI: 10.1021/acschembio.1c00626] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Antimicrobial resistance (AMR) has been increasing unrelentingly worldwide, thus negatively impacting human health. The discovery and development of novel antibiotics is an urgent unmet need of the hour. However, it has become more challenging, requiring increasingly time-consuming efforts with increased commercial risks. Hence, alternative strategies are urgently needed to potentiate the existing antibiotics. In this context, short cationic peptides or peptide-based antimicrobials that mimic the activity of naturally occurring antimicrobial peptides (AMPs) could overcome the disadvantages of AMPs having evolved as potent antibacterial agents. Besides their potent antibacterial efficacy, short peptide conjugates have also gained attention as potent adjuvants to conventional antibiotics. Such peptide antibiotic combinations have become an increasingly cost-effective therapeutic option to tackle AMR. This Review summarizes the recent progress for peptide-based small molecules as promising antimicrobials and as adjuvants for conventional antibiotics to counter multidrug resistant (MDR) pathogens.
Collapse
Affiliation(s)
- Apurva Panjla
- Department of Chemistry, IIT Kanpur, Kanpur-208016, Uttar Pradesh, India
| | - Grace Kaul
- Department of Microbiology, CSIR-Central Drug Research Institute, Lucknow-226031, Uttar Pradesh, India
- AcSIR: Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sidharth Chopra
- Department of Microbiology, CSIR-Central Drug Research Institute, Lucknow-226031, Uttar Pradesh, India
- AcSIR: Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Alexander Titz
- Chemical Biology of Carbohydrates (CBCH), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research, 66123 Saarbrücken, Germany
- Department of Chemistry, Saarland University, 66123 Saarbrücken, Germany
- Deutsches Zentrum für Infektionsforschung (DZIF), 38124, Standort Hannover-Braunschweig, Germany
| | - Sandeep Verma
- Department of Chemistry, IIT Kanpur, Kanpur-208016, Uttar Pradesh, India
- Center for Nanoscience, IIT Kanpur, Kanpur-208016, Uttar Pradesh, India
| |
Collapse
|
15
|
Sun Y, Deng Z, Jiang X, Yuan B, Yang K. Interactions between polymyxin B and various bacterial membrane mimics: A molecular dynamics study. Colloids Surf B Biointerfaces 2021; 211:112288. [PMID: 34942463 DOI: 10.1016/j.colsurfb.2021.112288] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 11/21/2021] [Accepted: 12/11/2021] [Indexed: 11/17/2022]
Abstract
Polymyxin B (PMB) is clinically used as a last-line therapy against life-threatening Gram-negative "superbugs". However, thorough understanding of the membrane actions of PMB at a molecular level is still lacking. In this work, a variety of bacterial membrane mimics with varying lipid compositions were built, and their interactions with PMB were systematically investigated using coarse-grained molecular dynamics simulation. PMB demonstrated characteristic preference to specific lipid species during its interaction with different membrane systems, such as the rough mutant lipipolysacchrides (Re LPS) preference in an outer membrane (OM) or the cardiolipin and POPG affinity in an inner membrane (IM). As a result of the lipid-specific actions, complicated membrane interaction states of PMB were observed, including adsorption on the OM surface. In contrast, for the IM or a mutative OM containing "impurity lipids" like POPE, POPG or lipid A, it could insert into the membrane via its acyl chain. Such actions of PMB influence the structure and lipid mobility of the membrane. In particular, the OM-bound PMB breaks the synchronous movement of Re LPS molecules in the outer leaflet and makes them diffuse more randomly, while its insertion into IM blocks the phospholipid diffusion and makes the membrane more homogeneous in the trajectory space. Our results provide insight into the action mechanism of PMB at a membrane level and a foundation for developing novel and safer polymyxin strategies for better clinical use.
Collapse
Affiliation(s)
- Yuliang Sun
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou 215006, Jiangsu, China
| | - Zhixiong Deng
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou 215006, Jiangsu, China
| | - Xukai Jiang
- National Glycoengineering Research Center, Shandong University, Qingdao 266237, Shandong, China
| | - Bing Yuan
- Songshan Lake Materials Laboratory, Dongguan 523808, Guangdong, China.
| | - Kai Yang
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou 215006, Jiangsu, China.
| |
Collapse
|
16
|
Weerakoon D, Petrov K, Pedebos C, Khalid S. Polymyxin B1 within the E. coli cell envelope: insights from molecular dynamics simulations. Biophys Rev 2021; 13:1061-1070. [PMID: 35047090 PMCID: PMC8724489 DOI: 10.1007/s12551-021-00869-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 10/22/2021] [Indexed: 11/25/2022] Open
Abstract
Polymyxins are used as last-resort antibiotics, where other treatments have been ineffectual due to antibiotic resistance. However, resistance to polymyxins has also been now reported, therefore it is instructive to characterise at the molecular level, the mechanisms of action of polymyxins. Here we review insights into these mechanisms from molecular dynamics simulations and discuss the utility of simulations as a complementary technique to experimental methodologies.
Collapse
Affiliation(s)
| | - Kamen Petrov
- Hertford College, University of Oxford, Oxford, OX1 3BW UK
| | - Conrado Pedebos
- School of Chemistry, University of Southampton, Southampton, SO17 1BJ UK
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU UK
| | - Syma Khalid
- School of Chemistry, University of Southampton, Southampton, SO17 1BJ UK
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU UK
| |
Collapse
|
17
|
Rodrigues Dos Santos Barbosa C, Feitosa Muniz D, Silvino Pereira P, Maria de Arruda Lima S, Datiane de Morais Oliveira Tintino C, Cintia Alexandrino de Souza V, Mariana Assis da Silva J, Henrique Sousa da Costa R, Cosmo Andrade Pinheiro J, Maria Lobo Soares de Matos Y, Rose Alencar Menezes I, Gonçalves da Silva T, Manoella de Souza Lima G, Cristina Leal Balbino T, Pinto Siqueira-Júnior J, Assis Bezerra da Cunha F, Douglas Melo Coutinho H, Relison Tintino S. Evaluation of Elaiophylin extracted from Streptomyces hygroscopicus as a potential inhibitor of the NorA efflux protein in Staphylococcus aureus: An in vitro and in silico approach. Bioorg Med Chem Lett 2021; 50:128334. [PMID: 34425202 DOI: 10.1016/j.bmcl.2021.128334] [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: 04/30/2021] [Revised: 08/13/2021] [Accepted: 08/16/2021] [Indexed: 10/20/2022]
Abstract
Compounds capable of inhibiting the efflux pump mechanism are a promising alternative against bacterial resistance because, when combined with antibiotics, they can increase the effectiveness of these drugs by inhibiting active efflux. Elaiophylin, derived from Streptomyces hygroscopicus, is a natural antibiotic that exhibits a variety of biological activities, including antibacterial activity. However, its potential as an inhibitor of the bacterial efflux mechanism has not been investigated. This study evaluated the ability of Elaiophylin to inhibit the NorA efflux pump in Staphylococcus aureus strains. Therefore, tests were performed to obtain the Minimum Inhibitory Concentration (MIC) and to verify the ability of Elaiophylin to potentiate the MIC of the antibiotic Norfloxacin and Ethidium Bromide (EtBr), known substrates of NorA efflux. Real-time PCR and molecular docking assays were also performed to assess the potential of Elaiophylin against NorA. The strains SA-1199 (wild type) and SA-1199B (NorA over-expressed) of S. aureus were used for this study. The results showed that Elaiophylin significantly decreased the MIC of Norfloxacin and EtBr, increasing the activity of these substrates against S. aureus, which carries the efflux protein NorA. However, Elaiophylin provided a non-significant reduction in norA gene expression, however, molecular docking demonstrated a high binding affinity between Elaiophylin and NorA efflux protein, indicating that Elaiophylin can act as a potential NorA in S. aureus.
Collapse
Affiliation(s)
| | - Débora Feitosa Muniz
- Laboratory of Microbiology and Molecular Biology (LMBM), Department of Biological Chemistry/CCBS/URCA, Brazil
| | | | | | | | | | | | | | | | | | - Irwin Rose Alencar Menezes
- Laboratory of Microbiology and Molecular Biology (LMBM), Department of Biological Chemistry/CCBS/URCA, Brazil
| | | | | | | | | | | | | | - Saulo Relison Tintino
- Laboratory of Microbiology and Molecular Biology (LMBM), Department of Biological Chemistry/CCBS/URCA, Brazil
| |
Collapse
|
18
|
Jiang X, Sun Y, Yang K, Yuan B, Velkov T, Wang L, Li J. Coarse-grained simulations uncover Gram-negative bacterial defense against polymyxins by the outer membrane. Comput Struct Biotechnol J 2021; 19:3885-3891. [PMID: 34584634 PMCID: PMC8441625 DOI: 10.1016/j.csbj.2021.06.051] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 01/12/2023] Open
Abstract
A structural model of bacterial outer membrane (OM) was developed with Ra LPS. Free energy landscape was revealed for polymyxin interactions with the OM. LPS core sugars and calcium ions confer intrinsic resistance to antibiotics.
The outer membrane (OM) of Gram-negative bacteria is a formidable barrier against antibiotics. Understanding the structure and function of the OM is essential for the discovery of novel membrane-acting agents against multidrug-resistant Gram-negative pathogens. However, it remains challenging to obtain three-dimensional structure of bacterial membranes using crystallographic approaches, which has significantly hindered the elucidation of its interaction with antibiotics. Here, we developed an asymmetric OM model consisting of rough lipopolysaccharide (LPS) and three key types of phospholipids. Using coarse-grained molecular dynamics simulations, we investigated the interaction dynamics of LPS-containing OM with the polymyxins, a last-line class of antibiotics against Gram-negative ‘superbugs’. We discovered that polymyxin molecules spontaneously penetrated the OM core sugar region where most were trapped before entering the lipid A region. Examination of the free energy profile of polymyxin penetration revealed a major free energy barrier at the LPS inner core and lipid A interface. Further analysis revealed calcium ions predominantly distributed in the inner core region and mediated extensive cross-linking interactions between LPS molecules, thereby inhibiting the penetration of polymyxins into the hydrophobic region of the OM. Collectively, our results provide novel mechanistic insights into an intrinsic defense of Gram-negative bacteria to polymyxins and may help identify new antimicrobial targets.
Collapse
Affiliation(s)
- Xukai Jiang
- National Glycoengineering Research Center, Shandong University, Qingdao, China.,Biomedicine Discovery Institute, Infection & Immunity Program, Department of Microbiology, Monash University, Melbourne, Australia
| | - Yuliang Sun
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou, China
| | - Kai Yang
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou, China
| | - Bing Yuan
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou, China
| | - Tony Velkov
- Department of Pharmacology and Therapeutics, The University of Melbourne, Melbourne, Australia
| | - Lushan Wang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Jian Li
- Biomedicine Discovery Institute, Infection & Immunity Program, Department of Microbiology, Monash University, Melbourne, Australia
| |
Collapse
|
19
|
Goode A, Yeh V, Bonev BB. Interactions of polymyxin B with lipopolysaccharide-containing membranes. Faraday Discuss 2021; 232:317-329. [PMID: 34550139 PMCID: PMC8704168 DOI: 10.1039/d1fd00036e] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Bacterial resistance to antibiotics constantly remodels the battlefront between infections and antibiotic therapy. Polymyxin B, a cationic peptide with an anti-Gram-negative spectrum of activity is re-entering use as a last resort measure and as an adjuvant. We use fluorescence dequenching to investigate the role of the rough chemotype bacterial lipopolysaccharide from E. coli BL21 as a molecular facilitator of membrane disruption by LPS. The minimal polymyxin B/lipid ratio required for leakage onset increased from 5.9 × 10−4 to 1.9 × 10−7 in the presence of rLPS. We confirm polymyxin B activity against E. coli BL21 by the agar diffusion method and determined a MIC of 291 μg ml−1. Changes in lipid membrane stability and dynamics in response to polymyxin and the role of LPS are investigated by 31P NMR and high resolution 31P MAS NMR relaxation is used to monitor selective molecular interactions between polymyxin B and rLPS within bilayer lipid membranes. We observe a strong facilitating effect from rLPS on the membrane lytic properties of polymyxin B and a specific, pyrophosphate-mediated process of molecular recognition of LPS by polymyxin B. Polymyxin B uses bacterial LPS as docking receptor to cross the outer membrane.![]()
Collapse
Affiliation(s)
- Alice Goode
- School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, NG7 2UH, UK.
| | - Vivien Yeh
- School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, NG7 2UH, UK.
| | - Boyan B Bonev
- School of Life Sciences, University of Nottingham, Queen's Medical Centre, Nottingham, NG7 2UH, UK.
| |
Collapse
|
20
|
Mohamed Z, Shin JH, Ghosh S, Sharma AK, Pinnock F, Bint E Naser Farnush S, Dörr T, Daniel S. Clinically Relevant Bacterial Outer Membrane Models for Antibiotic Screening Applications. ACS Infect Dis 2021; 7:2707-2722. [PMID: 34227387 DOI: 10.1021/acsinfecdis.1c00217] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Antibiotic resistance is a growing global health concern that has been increasing in prevalence over the past few decades. In Gram-negative bacteria, the outer membrane is an additional barrier through which antibiotics must traverse to kill the bacterium. In addition, outer membrane features and properties, like membrane surface charge, lipopolysaccharide (LPS) length, and membrane porins, can be altered in response to antibiotics and therefore, further mediate resistance. Model membranes have been used to mimic bacterial membranes to study antibiotic-induced membrane changes but often lack the compositional complexity of the actual outer membrane. Here, we developed a surface-supported membrane platform using outer membrane vesicles (OMVs) from clinically relevant Gram-negative bacteria and use it to characterize membrane biophysical properties and investigate its interaction with antibacterial compounds. We demonstrate that this platform maintains critical features of outer membranes, like fluidity, while retaining complex membrane components, like OMPs and LPS, which are central to membrane-mediated antibiotic resistance. This platform offers a non-pathogenic, cell-free surface to study such phenomena that is compatible with advanced microscopy and surface characterization tools like quartz crystal microbalance. We confirm these OMV bilayers recapitulate membrane interactions (or lack thereof) with the antibiotic compounds polymyxin B, bacitracin, and vancomycin, validating their use as representative models for the bacterial surface. By forming OMV bilayers from different strains, we envision that this platform could be used to investigate underlying biophysical differences in outer membranes leading to resistance, to screen and identify membrane-active antibiotics, or for the development of phage technologies targeting a particular membrane surface component.
Collapse
Affiliation(s)
- Zeinab Mohamed
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York United States
| | - Jung-Ho Shin
- Weill Institute for Cell and Molecular Biology and Department of Microbiology, Cornell University, Ithaca, New York United States
| | - Surajit Ghosh
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York United States
| | - Abhishek K. Sharma
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York United States
| | - Ferra Pinnock
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York United States
| | - Samavi Bint E Naser Farnush
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York United States
| | - Tobias Dörr
- Weill Institute for Cell and Molecular Biology and Department of Microbiology, Cornell University, Ithaca, New York United States
| | - Susan Daniel
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York United States
- Robert F. Smith School of Chemical and Biomolecular Engineering, Cornell University, Ithaca, New York United States
| |
Collapse
|
21
|
Jiang X, Yang K, Yuan B, Han M, Zhu Y, Roberts KD, Patil NA, Li J, Gong B, Hancock REW, Velkov T, Schreiber F, Wang L, Li J. Molecular dynamics simulations informed by membrane lipidomics reveal the structure-interaction relationship of polymyxins with the lipid A-based outer membrane of Acinetobacter baumannii. J Antimicrob Chemother 2021; 75:3534-3543. [PMID: 32911540 DOI: 10.1093/jac/dkaa376] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 08/04/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND MDR bacteria represent an urgent threat to human health globally. Polymyxins are a last-line therapy against life-threatening Gram-negative 'superbugs', including Acinetobacter baumannii. Polymyxins exert antimicrobial activity primarily via permeabilizing the bacterial outer membrane (OM); however, the mechanism of interaction between polymyxins and the OM remains unclear at the atomic level. METHODS We constructed a lipid A-based OM model of A. baumannii using quantitative membrane lipidomics data and employed all-atom molecular dynamics simulations with umbrella sampling techniques to elucidate the structure-interaction relationship and thermodynamics governing the penetration of polymyxins [B1 and E1 (i.e. colistin A) representing the two clinically used polymyxins] into the OM. RESULTS Polymyxin B1 and colistin A bound to the A. baumannii OM by the initial electrostatic interactions between the Dab residues of polymyxins and the phosphates of lipid A, competitively displacing the cations from the headgroup region of the OM. Both polymyxin B1 and colistin A formed a unique folded conformation upon approaching the hydrophobic centre of the OM, consistent with previous experimental observations. Polymyxin penetration induced reorientation of the headgroups of the OM lipids near the penetration site and caused local membrane disorganization, thereby significantly increasing membrane permeability and promoting the subsequent penetration of polymyxin molecules into the OM and periplasmic space. CONCLUSIONS The thermodynamics governing the penetration of polymyxins through the outer leaflet of the A. baumannii OM were examined and novel structure-interaction relationship information was obtained at the atomic and membrane level. Our findings will facilitate the discovery of novel polymyxins against MDR Gram-negative pathogens.
Collapse
Affiliation(s)
- Xukai Jiang
- Biomedicine Discovery Institute, Infection & Immunity Program and Department of Microbiology, Monash University, Melbourne, Australia
| | - Kai Yang
- Centre for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou, China
| | - Bing Yuan
- Centre for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou, China
| | - Meiling Han
- Biomedicine Discovery Institute, Infection & Immunity Program and Department of Microbiology, Monash University, Melbourne, Australia
| | - Yan Zhu
- Biomedicine Discovery Institute, Infection & Immunity Program and Department of Microbiology, Monash University, Melbourne, Australia
| | - Kade D Roberts
- Biomedicine Discovery Institute, Infection & Immunity Program and Department of Microbiology, Monash University, Melbourne, Australia
| | - Nitin A Patil
- Biomedicine Discovery Institute, Infection & Immunity Program and Department of Microbiology, Monash University, Melbourne, Australia
| | - Jingliang Li
- Institute for Frontier Materials, Deakin University, Geelong, Victoria, Australia
| | - Bin Gong
- School of Computer Science and Technology, Shandong University, Jinan, China
| | - Robert E W Hancock
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada
| | - Tony Velkov
- Department of Pharmacology & Therapeutics, University of Melbourne, Melbourne, Australia
| | - Falk Schreiber
- Department of Computer and Information Science, University of Konstanz, Konstanz, Germany
| | - Lushan Wang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Jian Li
- Biomedicine Discovery Institute, Infection & Immunity Program and Department of Microbiology, Monash University, Melbourne, Australia
| |
Collapse
|
22
|
Kassem N, Araya-Secchi R, Bugge K, Barclay A, Steinocher H, Khondker A, Wang Y, Lenard AJ, Bürck J, Sahin C, Ulrich AS, Landreh M, Pedersen MC, Rheinstädter MC, Pedersen PA, Lindorff-Larsen K, Arleth L, Kragelund BB. Order and disorder-An integrative structure of the full-length human growth hormone receptor. SCIENCE ADVANCES 2021; 7:7/27/eabh3805. [PMID: 34193419 PMCID: PMC8245047 DOI: 10.1126/sciadv.abh3805] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 05/18/2021] [Indexed: 05/13/2023]
Abstract
Because of its small size (70 kilodalton) and large content of structural disorder (>50%), the human growth hormone receptor (hGHR) falls between the cracks of conventional high-resolution structural biology methods. Here, we study the structure of the full-length hGHR in nanodiscs with small-angle x-ray scattering (SAXS) as the foundation. We develop an approach that combines SAXS, x-ray diffraction, and NMR spectroscopy data obtained on individual domains and integrate these through molecular dynamics simulations to interpret SAXS data on the full-length hGHR in nanodiscs. The hGHR domains reorient freely, resulting in a broad structural ensemble, emphasizing the need to take an ensemble view on signaling of relevance to disease states. The structure provides the first experimental model of any full-length cytokine receptor in a lipid membrane and exemplifies how integrating experimental data from several techniques computationally may access structures of membrane proteins with long, disordered regions, a widespread phenomenon in biology.
Collapse
Affiliation(s)
- Noah Kassem
- Structural Biology and NMR Laboratory, Department of Biology, University of Copenhagen, Ole Maaløes vej 5, 2200 Copenhagen N, Denmark
| | - Raul Araya-Secchi
- X-ray and Neutron Science, The Niels Bohr Institute, University of Copenhagen, Copenhagen, Denmark
| | - Katrine Bugge
- Structural Biology and NMR Laboratory, Department of Biology, University of Copenhagen, Ole Maaløes vej 5, 2200 Copenhagen N, Denmark
| | - Abigail Barclay
- X-ray and Neutron Science, The Niels Bohr Institute, University of Copenhagen, Copenhagen, Denmark
| | - Helena Steinocher
- Structural Biology and NMR Laboratory, Department of Biology, University of Copenhagen, Ole Maaløes vej 5, 2200 Copenhagen N, Denmark
| | - Adree Khondker
- Department of Physics and Astronomy, McMaster University, Hamilton, ON, Canada
| | - Yong Wang
- Structural Biology and NMR Laboratory, Department of Biology, University of Copenhagen, Ole Maaløes vej 5, 2200 Copenhagen N, Denmark
| | - Aneta J Lenard
- Structural Biology and NMR Laboratory, Department of Biology, University of Copenhagen, Ole Maaløes vej 5, 2200 Copenhagen N, Denmark
| | - Jochen Bürck
- Institute of Biological Interfaces (IBG-2), Karlsruhe Institute of Technology (KIT), POB 3640, 76021 Karlsruhe, Germany
| | - Cagla Sahin
- Department of Microbiology, Tumor, and Cell Biology, Karolinska Institutet, Stockholm 171 65, Sweden
| | - Anne S Ulrich
- Institute of Biological Interfaces (IBG-2), Karlsruhe Institute of Technology (KIT), POB 3640, 76021 Karlsruhe, Germany
| | - Michael Landreh
- Department of Microbiology, Tumor, and Cell Biology, Karolinska Institutet, Stockholm 171 65, Sweden
| | - Martin Cramer Pedersen
- X-ray and Neutron Science, The Niels Bohr Institute, University of Copenhagen, Copenhagen, Denmark
| | | | - Per Amstrup Pedersen
- Department of Biology, University of Copenhagen, Universitetsparken 13, DK-2100 Copenhagen, Denmark
| | - Kresten Lindorff-Larsen
- Structural Biology and NMR Laboratory, Department of Biology, University of Copenhagen, Ole Maaløes vej 5, 2200 Copenhagen N, Denmark.
| | - Lise Arleth
- X-ray and Neutron Science, The Niels Bohr Institute, University of Copenhagen, Copenhagen, Denmark.
| | - Birthe B Kragelund
- Structural Biology and NMR Laboratory, Department of Biology, University of Copenhagen, Ole Maaløes vej 5, 2200 Copenhagen N, Denmark.
| |
Collapse
|
23
|
The Pseudomonas aeruginosa substrate-binding protein Ttg2D functions as a general glycerophospholipid transporter across the periplasm. Commun Biol 2021; 4:448. [PMID: 33837253 PMCID: PMC8035174 DOI: 10.1038/s42003-021-01968-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 03/04/2021] [Indexed: 11/19/2022] Open
Abstract
In Pseudomonas aeruginosa, Ttg2D is the soluble periplasmic phospholipid-binding component of an ABC transport system thought to be involved in maintaining the asymmetry of the outer membrane. Here we use the crystallographic structure of Ttg2D at 2.5 Å resolution to reveal that this protein can accommodate four acyl chains. Analysis of the available structures of Ttg2D orthologs shows that they conform a new substrate-binding-protein structural cluster. Native and denaturing mass spectrometry experiments confirm that Ttg2D, produced both heterologously and homologously and isolated from the periplasm, can carry two diacyl glycerophospholipids as well as one cardiolipin. Binding is notably promiscuous, allowing the transport of various molecular species. In vitro binding assays coupled to native mass spectrometry show that binding of cardiolipin is spontaneous. Gene knockout experiments in P. aeruginosa multidrug-resistant strains reveal that the Ttg2 system is involved in low-level intrinsic resistance against certain antibiotics that use a lipid-mediated pathway to permeate through membranes. Yero et al. elucidate the function of Ttg2D, a Pseudomonas aeruginosa periplasmic protein, in maintaining phospholipid asymmetry between the outer and inner membrane. Gram negative bacteria have inner and outer membranes that differ in phospholipd composition. Using X-ray crystallography and mass spectrometry, the authors show that Ttg2D can carry two diacyl glycerophospholipids or a cardiolipin. The authors also identify a role for Ttg2D in resistance against antibiotics that use a lipid-mediated pathway into the cell.
Collapse
|
24
|
Investigating the mechanism of action of aggregation-inducing antimicrobial Pept-ins. Cell Chem Biol 2021; 28:524-536.e4. [PMID: 33434517 DOI: 10.1016/j.chembiol.2020.12.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 11/20/2020] [Accepted: 12/16/2020] [Indexed: 12/24/2022]
Abstract
Aggregation can be selectively induced by aggregation-prone regions (APRs) contained in the target proteins. Aggregation-inducing antimicrobial peptides (Pept-ins) contain sequences homologous to APRs of target proteins and exert their bactericidal effect by causing aggregation of a large number of proteins. To better understand the mechanism of action of Pept-ins and the resistance mechanisms, we analyzed the phenotypic, lipidomic, and transcriptomic as well as genotypic changes in laboratory-derived Pept-in-resistant E. coli mutator cells. The analysis showed that the Pept-in resistance mechanism is dominated by a decreased Pept-in uptake, in both laboratory-derived mutator cells and clinical isolates. Our data indicate that Pept-in uptake involves an electrostatic attraction between the Pept-in and the bacterial membrane and follows a complex mechanism potentially involving many transporters. Furthermore, it seems more challenging for bacteria to become resistant toward Pept-ins that are less dependent on electrostatic attraction for uptake, suggesting that future Pept-ins should be selected for this property.
Collapse
|
25
|
Dou Y, Li W, Xia Y, Chen Z, Wu Z, Ge Y, Lin Z, Zhang M, Yang K, Yuan B, Kang Z. Photo-Voltage Transients for Real-Time Analysis of the Interactions between Molecules and Membranes. ACS APPLIED BIO MATERIALS 2020. [DOI: 10.1021/acsabm.0c01180] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Yujiang Dou
- College of Electronics and Information, Soochow University, Suzhou 215006, Jiangsu, China
- Suzhou Weimu Intelligent System Co. Ltd., Suzhou 215163, Jiangsu, China
| | - Wenwen Li
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou 215006, Jiangsu, China
| | - Yu Xia
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou 215006, Jiangsu, China
| | - Zhonglan Chen
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou 215006, Jiangsu, China
| | - Zhenyu Wu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, Jiangsu, China
| | - Yuke Ge
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou 215006, Jiangsu, China
| | - Zhao Lin
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou 215006, Jiangsu, China
| | - Mengling Zhang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, Jiangsu, China
| | - Kai Yang
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou 215006, Jiangsu, China
| | - Bing Yuan
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou 215006, Jiangsu, China
| | - Zhenhui Kang
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, Jiangsu, China
- Institute of Advanced Materials, Northeast Normal University, 5268 Renmin Street, Changchun 130024, Jilin, China
| |
Collapse
|
26
|
Jiang X, Yang K, Han ML, Yuan B, Li J, Gong B, Velkov T, Schreiber F, Wang L, Li J. Outer Membranes of Polymyxin-Resistant Acinetobacter baumannii with Phosphoethanolamine-Modified Lipid A and Lipopolysaccharide Loss Display Different Atomic-Scale Interactions with Polymyxins. ACS Infect Dis 2020; 6:2698-2708. [PMID: 32871077 DOI: 10.1021/acsinfecdis.0c00330] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Resistance to the last-line polymyxins is increasingly reported in multidrug-resistant Gram-negative pathogens, including Acinetobacter baumannii, which develops resistance via either lipid A modification (e.g., with phosphoethanolamine [pEtN]) or even lipopolysaccharide (LPS) loss in the outer membrane (OM). Considering these two different mechanisms, quantitative membrane lipidomics data were utilized to develop three OM models representing polymyxin-susceptible and -resistant A. baumannii strains. Through all-atom molecular simulations with enhanced sampling techniques, the effect of lipid A-pEtN modification and LPS loss on the action of colistin (i.e., polymyxin E) was examined for the first time, with a focus on the dynamics and energetics of colistin penetration into these OMs. Lipid A-pEtN modification improved the OM stability, impeding the penetration of colistin into the OM; this differed from the current literature that lipid A-pEtN modification confers resistance by diminishing the initial interaction with polymyxins. In contrast, the LPS deficiency significantly reduced the negative charges on the OM surface, diminishing the binding of colistin. Moreover, both lipid A-pEtN modification and LPS loss also constituted colistin resistance through disturbing the conformational transitions of the colistin molecule. Collectively, atomic-scale interactions between polymyxins and different bacterial OMs are very different and the findings may facilitate the discovery of new-generation polymyxins against Gram-negative 'superbugs'.
Collapse
Affiliation(s)
- Xukai Jiang
- Biomedicine Discovery Institute, Infection & Immunity Program, Department of Microbiology, Monash University, Melbourne, VIC 3800, Australia
| | - Kai Yang
- Center for Soft Condensed Matter Physics and Interdisciplinary Research, School of Physical Science and Technology, Soochow University, Suzhou 215006, China
| | - Mei-Ling Han
- Biomedicine Discovery Institute, Infection & Immunity Program, Department of Microbiology, Monash University, Melbourne, VIC 3800, Australia
| | - Bing Yuan
- Center for Soft Condensed Matter Physics and Interdisciplinary Research, School of Physical Science and Technology, Soochow University, Suzhou 215006, China
| | - Jingliang Li
- Institute for Frontier Materials, Deakin University, Geelong, VIC 3217, Australia
| | - Bin Gong
- School of Software, Shandong University, Jinan 250101, China
| | - Tony Velkov
- Department of Pharmacology & Therapeutics, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Falk Schreiber
- Department of Computer and Information Science, University of Konstanz, Konstanz 78467, Germany
| | - Lushan Wang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Jian Li
- Biomedicine Discovery Institute, Infection & Immunity Program, Department of Microbiology, Monash University, Melbourne, VIC 3800, Australia
| |
Collapse
|
27
|
Jiang X, Zhang S, Azad MAK, Roberts KD, Wan L, Gong B, Yang K, Yuan B, Uddin H, Li J, Thompson PE, Velkov T, Fu J, Wang L, Li J. Structure-Interaction Relationship of Polymyxins with the Membrane of Human Kidney Proximal Tubular Cells. ACS Infect Dis 2020; 6:2110-2119. [PMID: 32619094 PMCID: PMC7485602 DOI: 10.1021/acsinfecdis.0c00190] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Multidrug-resistant Gram-negative bacteria are a serious global threat to human health. Polymyxins are increasingly used in patients as a last-line therapy to treat infections caused by these life-threatening 'superbugs'. Unfortunately, polymyxin-induced nephrotoxicity is the major dose-limiting factor and understanding its mechanism is crucial for the development of novel, safer polymyxins. Here, we undertook the first all-atom molecular dynamics simulations of the interaction between four naturally occurring polymyxins A1, B1, M1 and colistin A (representative structural variations of the polymyxin core structure) and the membrane of human kidney proximal tubular cells. All polymyxins inserted spontaneously into the hydrophobic region of the membrane where they were retained, although their insertion abilities varied. Polymyxin A1 completely penetrated into the hydrophobic region of the membrane with a unique folded conformation, whereas the other three polymyxins only inserted their fatty acyl tails into this region. Furthermore, local membrane defects and increased water penetration were induced by each polymyxin, which may represent the initial stage of cellular membrane damage. Finally, the structure-interaction relationship of polymyxins was investigated based on atomic interactions at the cell membrane level. The hydrophobicity at positions 6/7 and stereochemistry at position 3 regulated the interactions of polymyxins with the cell membrane. Collectively, our results provide new mechanistic insights into polymyxin-induced nephrotoxicity at the atomic level and will facilitate the development of new-generation polymyxins.
Collapse
Affiliation(s)
- Xukai Jiang
- Biomedicine Discovery Institute, Infection & Immunity Program and Department of Microbiology, Monash University, Clayton, Melbourne, Victoria 3800, Australia
| | - Shuo Zhang
- Department of Mechanical and Aerospace Engineering, Monash University, Melbourne, Victoria 3800, Australia
| | - Mohammad A. K. Azad
- Biomedicine Discovery Institute, Infection & Immunity Program and Department of Microbiology, Monash University, Clayton, Melbourne, Victoria 3800, Australia
| | - Kade D. Roberts
- Biomedicine Discovery Institute, Infection & Immunity Program and Department of Microbiology, Monash University, Clayton, Melbourne, Victoria 3800, Australia
| | - Lin Wan
- School of Software, Shandong University, Jinan 250101, China
| | - Bin Gong
- School of Software, Shandong University, Jinan 250101, China
| | - Kai Yang
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou 215006, China
| | - Bing Yuan
- Center for Soft Condensed Matter Physics and Interdisciplinary Research & School of Physical Science and Technology, Soochow University, Suzhou 215006, China
| | - Hemayet Uddin
- Melbourne Centre for Nanofabrication, Clayton, Melbourne, Victoria 3168, Australia
| | - Jingliang Li
- Institute for Frontier Materials, Deakin University, Geelong, Victoria 3216, Australia
| | - Philip E. Thompson
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Parkville, Melbourne, Victoria 3052, Australia
| | - Tony Velkov
- Department of Pharmacology & Therapeutics, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Jing Fu
- Department of Mechanical and Aerospace Engineering, Monash University, Melbourne, Victoria 3800, Australia
| | - Lushan Wang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China
| | - Jian Li
- Biomedicine Discovery Institute, Infection & Immunity Program and Department of Microbiology, Monash University, Clayton, Melbourne, Victoria 3800, Australia
| |
Collapse
|
28
|
Ayoub Moubareck C. Polymyxins and Bacterial Membranes: A Review of Antibacterial Activity and Mechanisms of Resistance. MEMBRANES 2020; 10:membranes10080181. [PMID: 32784516 PMCID: PMC7463838 DOI: 10.3390/membranes10080181] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 07/30/2020] [Accepted: 07/31/2020] [Indexed: 12/19/2022]
Abstract
Following their initial discovery in the 1940s, polymyxin antibiotics fell into disfavor due to their potential clinical toxicity, especially nephrotoxicity. However, the dry antibiotic development pipeline, together with the rising global prevalence of infections caused by multidrug-resistant (MDR) Gram-negative bacteria have both rejuvenated clinical interest in these polypeptide antibiotics. Parallel to the revival of their use, investigations into the mechanisms of action and resistance to polymyxins have intensified. With an initial known effect on biological membranes, research has uncovered the detailed molecular and chemical interactions that polymyxins have with Gram-negative outer membranes and lipopolysaccharide structure. In addition, genetic and epidemiological studies have revealed the basis of resistance to these agents. Nowadays, resistance to polymyxins in MDR Gram-negative pathogens is well elucidated, with chromosomal as well as plasmid-encoded, transferrable pathways. The aims of the current review are to highlight the important chemical, microbiological, and pharmacological properties of polymyxins, to discuss their mechanistic effects on bacterial membranes, and to revise the current knowledge about Gram-negative acquired resistance to these agents. Finally, recent research, directed towards new perspectives for improving these old agents utilized in the 21st century, to combat drug-resistant pathogens, is summarized.
Collapse
|
29
|
Li Z, Chakraborty P, de Vries RH, Song C, Zhao X, Roelfes G, Scheffers DJ, Kuipers OP. Characterization of two relacidines belonging to a novel class of circular lipopeptides that act against Gram-negative bacterial pathogens. Environ Microbiol 2020; 22:5125-5136. [PMID: 32608161 PMCID: PMC7818431 DOI: 10.1111/1462-2920.15145] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/28/2020] [Indexed: 12/18/2022]
Abstract
The development of sustainable agriculture and the increasing antibiotic resistance of human pathogens call for novel antimicrobial compounds. Here, we describe the extraction and characterization of a class of cationic circular lipopeptides, for which we propose the name relacidines, from the soil bacterium Brevibacillus laterosporus MG64. Relacidines are composed of a fatty acid side chain (4‐methylhexanoic acid) and 13 amino acid residues. A lactone ring is formed by the last five amino acid residues and three positively charged ornithines are located in the linear fragment. Relacidines selectively combat Gram‐negative pathogens, including phytopathogens and human pathogens. Further investigation of the mode of action revealed that relacidine B binds to the lipopolysaccharides but does not form pores in the cell membrane. We also provide proof to show that relacidine B does not affect the biosynthesis of the cell wall and RNA. Instead, it affects the oxidative phosphorylation process of cells and diminishes the biosynthesis of ATP. Transcription of relacidines is induced by plant pathogens, which strengthens the potential of B. laterosporus MG64 to be used as a biocontrol agent. Thus, we identified a new group of potent antibiotic compounds for combating Gram‐negative pathogens of plants or animals.
Collapse
Affiliation(s)
- Zhibo Li
- Department of Molecular Genetics, University of Groningen, Groningen, the Netherlands
| | - Parichita Chakraborty
- Department of Molecular Microbiology, University of Groningen, Groningen, the Netherlands
| | - Reinder H de Vries
- Stratingh Institute for Chemistry, University of Groningen, Groningen, the Netherlands
| | - Chunxu Song
- College of Resources and Environmental Science, National Academy of Agriculture Green Development, Key Laboratory of Plant-Soil Interaction, Ministry of Education, China Agricultural University, Beijing, 100193, China
| | - Xinghong Zhao
- Department of Molecular Genetics, University of Groningen, Groningen, the Netherlands
| | - Gerard Roelfes
- Stratingh Institute for Chemistry, University of Groningen, Groningen, the Netherlands
| | - Dirk-Jan Scheffers
- Department of Molecular Microbiology, University of Groningen, Groningen, the Netherlands
| | - Oscar P Kuipers
- Department of Molecular Genetics, University of Groningen, Groningen, the Netherlands
| |
Collapse
|
30
|
Di Marino D, Bruno A, Grimaldi M, Scrima M, Stillitano I, Amodio G, Della Sala G, Romagnoli A, De Santis A, Moltedo O, Remondelli P, Boccia G, D'Errico G, D'Ursi AM, Limongelli V. Binding of the Anti-FIV Peptide C8 to Differently Charged Membrane Models: From First Docking to Membrane Tubulation. Front Chem 2020; 8:493. [PMID: 32676493 PMCID: PMC7333769 DOI: 10.3389/fchem.2020.00493] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 05/13/2020] [Indexed: 12/11/2022] Open
Abstract
Gp36 is the virus envelope glycoproteins catalyzing the fusion of the feline immunodeficiency virus with the host cells. The peptide C8 is a tryptophan-rich peptide corresponding to the fragment 770W-I777 of gp36 exerting antiviral activity by binding the membrane cell and inhibiting the virus entry. Several factors, including the membrane surface charge, regulate the binding of C8 to the lipid membrane. Based on the evidence that imperceptible variation of membrane charge may induce a dramatic effect in several critical biological events, in the present work we investigate the effect induced by systematic variation of charge in phospholipid bilayers on the aptitude of C8 to interact with lipid membranes, the tendency of C8 to assume specific conformational states and the re-organization of the lipid bilayer upon the interaction with C8. Accordingly, employing a bottom-up multiscale protocol, including CD, NMR, ESR spectroscopy, atomistic molecular dynamics simulations, and confocal microscopy, we studied C8 in six membrane models composed of different ratios of zwitterionic/negatively charged phospholipids. Our data show that charge content modulates C8-membrane binding with significant effects on the peptide conformations. C8 in micelle solution or in SUV formed by DPC or DOPC zwitterionic phospholipids assumes regular β-turn structures that are progressively destabilized as the concentration of negatively charged SDS or DOPG phospholipids exceed 40%. Interaction of C8 with zwitterionic membrane surface is mediated by Trp1 and Trp4 that are deepened in the membrane, forming H-bonds and cation-π interactions with the DOPC polar heads. Additional stabilizing salt bridge interactions involve Glu2 and Asp3. MD and ESR data show that the C8-membrane affinity increases as the concentration of zwitterionic phospholipid increases. In the lipid membrane characterized by an excess of zwitterionic phospholipids, C8 is adsorbed at the membrane interface, inducing a stiffening of the outer region of the DOPC bilayer. However, the bound of C8 significantly perturbs the whole organization of lipid bilayer resulting in membrane remodeling. These events, measurable as a variation of the bilayer thickness, are the onset mechanism of the membrane fusion and vesicle tubulation observed in confocal microscopy by imaging zwitterionic MLVs in the presence of C8 peptide.
Collapse
Affiliation(s)
- Daniele Di Marino
- Department of Life and Environmental Sciences, New York-Marche Structural Biology Center (NY-MaSBiC), Polytechnic University of Marche, Ancona, Italy
| | - Agostino Bruno
- Department of Pharmacy, University of Naples "Federico II", Naples, Italy
| | | | - Mario Scrima
- Department of Pharmacy, University of Salerno, Fisciano, Italy
| | | | - Giuseppina Amodio
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Italy
| | - Grazia Della Sala
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Alice Romagnoli
- Department of Life and Environmental Sciences, New York-Marche Structural Biology Center (NY-MaSBiC), Polytechnic University of Marche, Ancona, Italy
| | - Augusta De Santis
- Department of Chemical Science, University of Naples Federico II, Naples, Italy
| | - Ornella Moltedo
- Department of Pharmacy, University of Salerno, Fisciano, Italy
| | - Paolo Remondelli
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Italy
| | - Giovanni Boccia
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi, Italy
| | - Gerardino D'Errico
- Department of Chemical Science, University of Naples Federico II, Naples, Italy
| | | | - Vittorio Limongelli
- Department of Pharmacy, University of Naples "Federico II", Naples, Italy.,Faculty of Biomedical Sciences, Institute of Computational Science, Università della Svizzera italiana (USI), Lugano, Switzerland
| |
Collapse
|
31
|
Rowen RJ. Ozone and oxidation therapies as a solution to the emerging crisis in infectious disease management: a review of current knowledge and experience. Med Gas Res 2020; 9:232-237. [PMID: 31898609 PMCID: PMC7802416 DOI: 10.4103/2045-9912.273962] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Medicine faces crisis with emerging “super bugs,” lethal viruses (Ebola), and stealth pathogens such as tick-borne infections. Thousands are dying worldwide of once easily treatable diseases. Ozone therapy, extensively studied, may be a valuable adjunctive or stand-alone therapy. Ebola again ravages Africa with over 2000 already dead, carrying a 65% mortality rate. The world desperately needs safe, inexpensive and effective anti-infective therapy to which microbes will not develop resistance. Oxidation therapies have shown an extremely high safety profile, lacking credible reports of significant injury beyond vein irritation. Ozone therapy, the most studied and least expensive to perform, is in itself a germicide, not an antibiotic, and improves several physiological parameters essential for infection defense. Recent reports indicate very favorable responses to both bacterial and viral disease, inclusive of Ebola. Despite lack of commercial profitability (not patentable), medicine would do well to revisit its pre-antibiotic era oxidation therapy roots, especially ozone in the current crisis.
Collapse
|
32
|
Ahmed R, Huang J, Weber DK, Gopinath T, Veglia G, Akimoto M, Khondker A, Rheinstädter MC, Huynh V, Wylie RG, Bozelli JC, Epand RM, Melacini G. Molecular Mechanism for the Suppression of Alpha Synuclein Membrane Toxicity by an Unconventional Extracellular Chaperone. J Am Chem Soc 2020; 142:9686-9699. [DOI: 10.1021/jacs.0c01894] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Rashik Ahmed
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton ON L8S 4M1, Canada
| | - Jinfeng Huang
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton ON L8S 4M1, Canada
| | - Daniel K. Weber
- Department of Biochemistry, Chemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Tata Gopinath
- Department of Biochemistry, Chemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Gianluigi Veglia
- Department of Biochemistry, Chemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Madoka Akimoto
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton ON L8S 4M1, Canada
| | - Adree Khondker
- Department of Physics and Astronomy, McMaster University, Hamilton ON L8S 4M1, Canada
| | | | - Vincent Huynh
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton ON L8S 4M1, Canada
| | - Ryan G. Wylie
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton ON L8S 4M1, Canada
| | - José C. Bozelli
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton ON L8S 4M1, Canada
| | - Richard M. Epand
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton ON L8S 4M1, Canada
| | - Giuseppe Melacini
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton ON L8S 4M1, Canada
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton ON L8S 4M1, Canada
| |
Collapse
|
33
|
Dubashynskaya NV, Skorik YA. Polymyxin Delivery Systems: Recent Advances and Challenges. Pharmaceuticals (Basel) 2020; 13:E83. [PMID: 32365637 PMCID: PMC7281078 DOI: 10.3390/ph13050083] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 04/26/2020] [Accepted: 04/27/2020] [Indexed: 02/07/2023] Open
Abstract
Polymyxins are vital antibiotics for the treatment of multiresistant Gram-negative ESKAPE pathogen infections. However, their clinical value is limited by their high nephrotoxicity and neurotoxicity, as well as their poor permeability and absorption in the gastrointestinal tract. This review focuses on various polymyxin delivery systems that improve polymyxin bioavailability and reduce drug toxicity through targeted and controlled release. Currently, the most suitable systems for improving oral, inhalation, and parenteral polymyxin delivery are polymer particles, liposomes, and conjugates, while gels, polymer fibers, and membranes are attractive materials for topical administration of polymyxin for the treatment of infected wounds and burns. In general, the application of these systems protects polymyxin molecules from the negative effects of both physiological and pathological factors while achieving higher concentrations at the target site and reducing dosage and toxicity. Improving the properties of polymyxin will be of great interest to researchers who are focused on developing antimicrobial drugs that show increased efficacy and safety.
Collapse
Affiliation(s)
| | - Yury A. Skorik
- Institute of Macromolecular Compounds of the Russian Academy of Sciences, Bolshoy pr. V.O. 31, St. Petersburg 199004, Russia;
| |
Collapse
|
34
|
Saem S, Shahid O, Khondker A, Moran-Hidalgo C, Rheinstädter MC, Moran-Mirabal J. Benchtop-fabricated lipid-based electrochemical sensing platform for the detection of membrane disrupting agents. Sci Rep 2020; 10:4595. [PMID: 32165701 PMCID: PMC7067837 DOI: 10.1038/s41598-020-61561-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 02/28/2020] [Indexed: 11/22/2022] Open
Abstract
There are increasing concerns about the danger that water-borne pathogens and pollutants pose to the public. Of particular importance are those that disrupt the plasma membrane, since loss of membrane integrity can lead to cell death. Currently, quantitative assays to detect membrane-disrupting (lytic) agents are done offsite, leading to long turnaround times and high costs, while existing colorimetric point-of-need solutions often sacrifice sensitivity. Thus, portable and highly sensitive solutions are needed to detect lytic agents for health and environmental monitoring. Here, a lipid-based electrochemical sensing platform is introduced to rapidly detect membrane-disrupting agents. The platform combines benchtop fabricated microstructured electrodes (MSEs) with lipid membranes. The sensing mechanism of the lipid-based platform relies on stacked lipid membranes serving as passivating layers that when disrupted generate electrochemical signals proportional to the membrane damage. The MSE topography, membrane casting and annealing conditions were optimized to yield the most reproducible and sensitive devices. We used the sensors to detect membrane-disrupting agents sodium dodecyl sulfate and Polymyxin-B within minutes and with limits of detection in the ppm regime. This study introduces a platform with potential for the integration of complex membranes on MSEs towards the goal of developing Membrane-on-Chip sensing devices.
Collapse
Affiliation(s)
- Sokunthearath Saem
- McMaster University, Department of Chemistry and Chemical Biology, Hamilton, L8S 4L8, Canada
| | - Osama Shahid
- McMaster University, Department of Chemistry and Chemical Biology, Hamilton, L8S 4L8, Canada
| | - Adree Khondker
- McMaster University, Department of Physics and Astronomy, Hamilton, L8S 4L8, Canada
| | - Camila Moran-Hidalgo
- McMaster University, Department of Chemistry and Chemical Biology, Hamilton, L8S 4L8, Canada
| | | | - Jose Moran-Mirabal
- McMaster University, Department of Chemistry and Chemical Biology, Hamilton, L8S 4L8, Canada.
| |
Collapse
|
35
|
Khondker A, Rheinstädter MC. How do bacterial membranes resist polymyxin antibiotics? Commun Biol 2020; 3:77. [PMID: 32066819 PMCID: PMC7026071 DOI: 10.1038/s42003-020-0803-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 01/09/2020] [Indexed: 01/17/2023] Open
Abstract
In our recent Communications Biology article, we reported on the biophysical mechanism of resistance for polymyxin antibiotics in bacterial membranes. The emergence of plasmid-borne colistin resistance poses a threat to our last line of defense against many pathogens. Here, we outline the current understanding of mcr-1 -mediated polymyxin resistance, and propose future directions for membrane-targeting antibiotic research. Adree Khondker and Maikel Rheinstadter discuss how bacteria escape being killed by polymyxin antibiotics. Touching on their recent Communications Biology paper, they elaborate on the mechanism by which the bacterial membrane becomes resistant and on future directions to take in order to understand this phenomenon.
Collapse
Affiliation(s)
- Adree Khondker
- Department of Physics and Astronomy, McMaster University, Hamilton, ON, Canada
- Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Maikel C Rheinstädter
- Department of Physics and Astronomy, McMaster University, Hamilton, ON, Canada.
- Origins Institute, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
36
|
Fu L, Wan M, Zhang S, Gao L, Fang W. Polymyxin B Loosens Lipopolysaccharide Bilayer but Stiffens Phospholipid Bilayer. Biophys J 2019; 118:138-150. [PMID: 31812355 DOI: 10.1016/j.bpj.2019.11.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/31/2019] [Accepted: 11/08/2019] [Indexed: 12/17/2022] Open
Abstract
Multidrug-resistant Gram-negative bacteria have increased the prevalence of a variety of serious diseases in modern times. Polymyxins are used as the last-line therapeutic options for the treatment of infections. However, the mechanism of action of polymyxins remains in dispute. In this work, we used a coarse-grained molecular dynamics simulation to investigate the mechanism of the cationic antimicrobial peptide polymyxin B (PmB) interacting with both the inner and outer membrane models of bacteria. Our results show that the binding of PmB disturbs the outer membrane by displacing the counterions, decreasing the orientation order of the lipopolysaccharide tail, and creating more lipopolysaccharide packing defects. Upon binding onto the inner membrane, in contrast to the traditional killing mechanism that antimicrobial peptides usually use to induce holes in the membrane, PmBs do not permeabilize the inner membrane but stiffen it by filling up the lipid packing defect, increasing the lipid tail order and the membrane bending rigidity as well as restricting the lipid diffusion. PmBs also mediate intermembrane contact and adhesion. These joint effects suggest that PmBs deprive the biological activity of Gram-negative bacteria by sterilizing the cell.
Collapse
Affiliation(s)
- Lei Fu
- Key Laboratory of Theoretical and Computational Photochemistry, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, China
| | - Mingwei Wan
- Key Laboratory of Theoretical and Computational Photochemistry, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, China
| | - Shan Zhang
- Key Laboratory of Theoretical and Computational Photochemistry, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, China
| | - Lianghui Gao
- Key Laboratory of Theoretical and Computational Photochemistry, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, China.
| | - Weihai Fang
- Key Laboratory of Theoretical and Computational Photochemistry, Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, China
| |
Collapse
|
37
|
Yokosawa T, Yamada M, Noguchi T, Suzuki S, Hirata Y, Matsuzawa A. Pro-caspase-3 protects cells from polymyxin B-induced cytotoxicity by preventing ROS accumulation. J Antibiot (Tokyo) 2019; 72:848-852. [PMID: 31371783 DOI: 10.1038/s41429-019-0216-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 07/05/2019] [Accepted: 07/07/2019] [Indexed: 12/21/2022]
Abstract
Polymyxin B (PMB), a last-line antibiotic used against antibiotic-resistant superbugs, causes undesirable cytotoxic side effects. However, its mechanisms remain unknown. In this study, we unexpectedly found that caspase-3, a main executor of apoptosis, plays a protective role in PMB-induced cytotoxicity. Caspase-3 knockout (KO) cells exhibited higher susceptibility to PMB-induced cytotoxicity compared with wild-type (WT) cells, accompanied by increased levels of reactive oxygen species (ROS). Interestingly, co-treatment with the antioxidant N-acetylcysteine (NAC) rescued cell viability to a similar extent as WT cells. Furthermore, PMB failed to facilitate the processing of inactive caspase-3 (pro-caspase-3) into active forms, suggesting that pro-caspase-3 nonenzymatically suppresses PMB-driven ROS accumulation and its cytotoxicity. Thus, our findings that demonstrate the potential ability of PMB to stimulate ROS generation, but which is normally masked by pro-caspase-3-dependent mechanisms, may provide novel insights into the mechanisms of PMB-induced side effects.
Collapse
Affiliation(s)
- Takumi Yokosawa
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Mayuka Yamada
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Takuya Noguchi
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan.
| | - Saki Suzuki
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Yusuke Hirata
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Atsushi Matsuzawa
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan.
| |
Collapse
|