1
|
Bertorello S, Cei F, Fink D, Niccolai E, Amedei A. The Future Exploring of Gut Microbiome-Immunity Interactions: From In Vivo/Vitro Models to In Silico Innovations. Microorganisms 2024; 12:1828. [PMID: 39338502 PMCID: PMC11434319 DOI: 10.3390/microorganisms12091828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 08/29/2024] [Accepted: 09/02/2024] [Indexed: 09/30/2024] Open
Abstract
Investigating the complex interactions between microbiota and immunity is crucial for a fruitful understanding progress of human health and disease. This review assesses animal models, next-generation in vitro models, and in silico approaches that are used to decipher the microbiome-immunity axis, evaluating their strengths and limitations. While animal models provide a comprehensive biological context, they also raise ethical and practical concerns. Conversely, modern in vitro models reduce animal involvement but require specific costs and materials. When considering the environmental impact of these models, in silico approaches emerge as promising for resource reduction, but they require robust experimental validation and ongoing refinement. Their potential is significant, paving the way for a more sustainable and ethical future in microbiome-immunity research.
Collapse
Affiliation(s)
- Sara Bertorello
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (S.B.); (F.C.); (D.F.); (A.A.)
| | - Francesco Cei
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (S.B.); (F.C.); (D.F.); (A.A.)
| | - Dorian Fink
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (S.B.); (F.C.); (D.F.); (A.A.)
| | - Elena Niccolai
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (S.B.); (F.C.); (D.F.); (A.A.)
- Laboratorio Congiunto MIA-LAB (Microbiome-Immunity Axis Research for a Circular Health), University of Florence, 50134 Florence, Italy
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, 50139 Florence, Italy; (S.B.); (F.C.); (D.F.); (A.A.)
- Laboratorio Congiunto MIA-LAB (Microbiome-Immunity Axis Research for a Circular Health), University of Florence, 50134 Florence, Italy
| |
Collapse
|
2
|
Anurag Anand A, Amod A, Anwar S, Sahoo AK, Sethi G, Samanta SK. A comprehensive guide on screening and selection of a suitable AMP against biofilm-forming bacteria. Crit Rev Microbiol 2024; 50:859-878. [PMID: 38102871 DOI: 10.1080/1040841x.2023.2293019] [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: 10/04/2023] [Revised: 11/27/2023] [Accepted: 11/30/2023] [Indexed: 12/17/2023]
Abstract
Lately, antimicrobial resistance (AMR) is increasing at an exponential rate making it important to search alternatives to antibiotics in order to combat multi-drug resistant (MDR) bacterial infections. Out of the several antibacterial and antibiofilm strategies being tested, antimicrobial peptides (AMPs) have shown to give better hopes in terms of a long-lasting solution to the problem. To select a desired AMP, it is important to make right use of available tools and databases that aid in identification, classification, and analysis of the physiochemical properties of AMPs. To identify the targets of these AMPs, it becomes crucial to understand their mode-of-action. AMPs can also be used in combination with other antibacterial and antibiofilm agents so as to achieve enhanced efficacy against bacteria and their biofilms. Due to concerns regarding toxicity, stability, and bioavailability, strategizing drug formulation at an early-stage becomes crucial. Although there are few concerns regarding development of bacterial resistance to AMPs, the evolution of resistance to AMPs occurs extremely slowly. This comprehensive review gives a deep insight into the selection of the right AMP, deciding the right target and combination strategy along with the type of formulation needed, and the possible resistance that bacteria can develop to these AMPs.
Collapse
Affiliation(s)
- Ananya Anurag Anand
- Department of Applied Sciences, Indian Institute of Information Technology Allahabad, Prayagraj, India
| | - Ayush Amod
- Department of Applied Sciences, Indian Institute of Information Technology Allahabad, Prayagraj, India
| | - Sarfraz Anwar
- Department of Bioinformatics, University of Allahabad, Prayagraj, India
| | - Amaresh Kumar Sahoo
- Department of Applied Sciences, Indian Institute of Information Technology Allahabad, Prayagraj, India
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Sintu Kumar Samanta
- Department of Applied Sciences, Indian Institute of Information Technology Allahabad, Prayagraj, India
| |
Collapse
|
3
|
Caselli L, Rodrigues GR, Franco OL, Malmsten M. Pulmonary delivery systems for antimicrobial peptides. Crit Rev Biotechnol 2024; 44:963-980. [PMID: 37731338 DOI: 10.1080/07388551.2023.2254932] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 07/04/2023] [Accepted: 07/17/2023] [Indexed: 09/22/2023]
Abstract
Bacterial infections of the respiratory tract cause millions of deaths annually. Several diseases exist wherein (1) bacterial infection is the main cause of disease (e.g., tuberculosis and bacterial pneumonia), (2) bacterial infection is a consequence of disease and worsens the disease prognosis (e.g., cystic fibrosis), and (3) bacteria-triggered inflammation propagates the disease (e.g., chronic obstructive pulmonary disease). Current approaches to combat infections generally include long and aggressive antibiotic treatments, which challenge patient compliance, thereby making relapses common and contributing to the development of antibiotic resistance. Consequently, the proportion of infections that cannot be treated with conventional antibiotics is rapidly increasing, and novel therapies are urgently needed. In this context, antimicrobial peptides (AMPs) have received considerable attention as they may exhibit potent antimicrobial effects against antibiotic-resistant bacterial strains but with modest toxicity. In addition, some AMPs suppress inflammation and provide other host defense functions (motivating the alternative term host defense peptides (HDPs)). However, the delivery of AMPs is complicated because they are large, positively charged, and amphiphilic. As a result of this, AMP delivery systems have recently attracted attention. For airway infections, the currently investigated delivery approaches range from aerosols and dry powders to various self-assembly and nanoparticle carrier systems, as well as their combinations. In this paper, we discuss recent developments in the field, ranging from mechanistic mode-of-action studies to the application of these systems for combating bacterial infections in the airways.
Collapse
Affiliation(s)
| | - Gisele R Rodrigues
- Centro de Análises Proteômicas e Bioquímicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Brazil
| | - Octavio L Franco
- Centro de Análises Proteômicas e Bioquímicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Brazil
- S-inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Brazil
| | - Martin Malmsten
- Physical Chemistry 1, University of Lund, Lund, Sweden
- Department of Pharmacy, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
4
|
Dasgupta P, Vinil K, Kanaujia SP. Evolutionary trends indicate a coherent organization of sap operons. Res Microbiol 2024:104228. [PMID: 38972435 DOI: 10.1016/j.resmic.2024.104228] [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: 04/15/2024] [Revised: 06/30/2024] [Accepted: 07/02/2024] [Indexed: 07/09/2024]
Abstract
Human hosts possess a complex network of immune responses against microbial pathogens. The production of antimicrobial peptides (AMPs), which target the pathogen cell membranes and inhibit them from inhabiting the hosts, is one such mechanism. However, pathogens have evolved systems that encounter these host-produced AMPs. The Sap (sensitivity to antimicrobial peptides) transporter uptakes AMPs inside the microbial cell and proteolytically degrades them. The Sap transporters comprise five subunits encoded by genes in an operon. Despite its ubiquitous nature, its subunits are not found to be in tandem with many organisms. In this study, a total of 421 Sap transporters were analyzed for their operonic arrangement. Out of 421, a total of 352 operons were found to be in consensus arrangement, while the remaining 69 show a varying arrangement of genes. The analysis of the intergenic distance between the subunits of the sap operon suggests a signature pattern with sapAB (-4), sapBC (-14), sapCD (-1), and sapDF (-4 to 1). An evolutionary analysis of these operons favors the consensus arrangement of the Sap transporter systems, substantiating its prevalence in most of the Gram-negative pathogens. Overall, this study provides insight into bacterial evolution, favoring the maintenance of the genetic organization of essential pathogenicity factors.
Collapse
Affiliation(s)
- Pratik Dasgupta
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati - 781039, Assam, India
| | - Kavya Vinil
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati - 781039, Assam, India
| | - Shankar Prasad Kanaujia
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati - 781039, Assam, India.
| |
Collapse
|
5
|
Rouhi A, Falah F, Azghandi M, Alizadeh Behbahani B, Tabatabaei-Yazdi F, Ibrahim SA, Dertli E, Vasiee A. Investigating the Effect of Melittin Peptide in Preventing Biofilm Formation, Adhesion and Expression of Virulence Genes in Listeria monocytogenes. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10318-z. [PMID: 38963508 DOI: 10.1007/s12602-024-10318-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/25/2024] [Indexed: 07/05/2024]
Abstract
Listeria monocytogenes is a notable food-borne pathogen that has the ability to create biofilms on different food processing surfaces, making it more resilient to disinfectants and posing a greater risk to human health. This study assessed melittin peptide's anti-biofilm and anti-pathogenicity effects on L. monocytogenes ATCC 19115. Melittin showed minimum inhibitory concenteration (MIC) of 100 μg/mL against this strain and scanning electron microscopy images confirmed its antimicrobial efficacy. The OD measurement demonstrated that melittin exhibited a strong proficiency in inhibiting biofilms and disrupting pre-formed biofilms at concentrations ranging from 1/8MIC to 2MIC and this amount was 92.59 ± 1.01% to 7.17 ± 0.31% and 100% to 11.50 ± 0.53%, respectively. Peptide also reduced hydrophobicity and self-aggregation of L. monocytogenes by 35.25% and 14.38% at MIC. Melittin also significantly reduced adhesion to HT-29 and Caco-2 cells by 61.33% and 59%, and inhibited invasion of HT-29 and Caco-2 cells by 49.33% and 40.66% for L. monocytogenes at the MIC value. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) revealed melittin's impact on gene expression, notably decreasing inlB (44%) and agrA (45%) gene expression in L. monocytogenes. flaA and hly genes also exhibited reduced expression. Also, significant changes were observed in sigB and prfA gene expression. These results underscore melittin's potential in combating bacterial infections and biofilm-related challenges in the food industry.
Collapse
Affiliation(s)
- Arezou Rouhi
- Department of Food Science and Technology, Faculty of Agriculture, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Fereshteh Falah
- Department of Food Science and Technology, Faculty of Agriculture, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Marjan Azghandi
- Department of Animal Science, Faculty of Agriculture, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Behrooz Alizadeh Behbahani
- Department of Food Science and Technology, Faculty of Animal Science and Food Technology, Agricultural Sciences and Natural Resources University of Khuzestan, Mollasani, Iran
| | - Farideh Tabatabaei-Yazdi
- Department of Food Science and Technology, Faculty of Agriculture, Ferdowsi University of Mashhad, Mashhad, Iran.
| | - Salam A Ibrahim
- Food and Nutritional Sciences Program, North Carolina Agricultural and Technical State University, E. Market Street, 1601, Greensboro, NC, 24711, USA
| | - Enes Dertli
- Department of Food Engineering, Faculty of Chemical and Metallurgical Engineering, Yildiz Technical University, Davutpasa Campüs, Istanbul, 34210, Türkiye
| | - Alireza Vasiee
- Department of Food Safety and Quality Control, Research Institute of Food Science and Technology (RIFST), Mashhad, Iran.
| |
Collapse
|
6
|
Elbediwi M, Rolff J. Metabolic pathways and antimicrobial peptide resistance in bacteria. J Antimicrob Chemother 2024; 79:1473-1483. [PMID: 38742645 DOI: 10.1093/jac/dkae128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024] Open
Abstract
Antimicrobial resistance is a pressing concern that poses a significant threat to global public health, necessitating the exploration of alternative strategies to combat drug-resistant microbial infections. Recently, antimicrobial peptides (AMPs) have gained substantial attention as possible replacements for conventional antibiotics. Because of their pharmacodynamics and killing mechanisms, AMPs display a lower risk of bacterial resistance evolution compared with most conventional antibiotics. However, bacteria display different mechanisms to resist AMPs, and the role of metabolic pathways in the resistance mechanism is not fully understood. This review examines the intricate relationship between metabolic genes and AMP resistance, focusing on the impact of metabolic pathways on various aspects of resistance. Metabolic pathways related to guanosine pentaphosphate (pppGpp) and guanosine tetraphosphate (ppGpp) [collectively (p)ppGpp], the tricarboxylic acid (TCA) cycle, haem biosynthesis, purine and pyrimidine biosynthesis, and amino acid and lipid metabolism influence in different ways metabolic adjustments, biofilm formation and energy production that could be involved in AMP resistance. By targeting metabolic pathways and their associated genes, it could be possible to enhance the efficacy of existing antimicrobial therapies and overcome the challenges exhibited by phenotypic (recalcitrance) and genetic resistance toward AMPs. Further research in this area is needed to provide valuable insights into specific mechanisms, uncover novel therapeutic targets, and aid in the fight against antimicrobial resistance.
Collapse
Affiliation(s)
- Mohammed Elbediwi
- Evolutionary Biology, Institute for Biology, Freie Universität Berlin, 14195 Berlin, Germany
- Animal Health Research Institute, Agriculture Research Centre, 12618 Cairo, Egypt
| | - Jens Rolff
- Evolutionary Biology, Institute for Biology, Freie Universität Berlin, 14195 Berlin, Germany
- Berlin-Brandenburg Institute of Advanced Biodiversity Research (BBIB), Berlin, Germany
| |
Collapse
|
7
|
Recktenwald M, Kaur M, Benmassaoud MM, Copling A, Khanna T, Curry M, Cortes D, Fleischer G, Carabetta VJ, Vega SL. Antimicrobial Peptide Screening for Designing Custom Bactericidal Hydrogels. Pharmaceutics 2024; 16:860. [PMID: 39065557 PMCID: PMC11279943 DOI: 10.3390/pharmaceutics16070860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 06/21/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
Staphylococcus aureus (S. aureus) is an opportunistic pathogen that lives on surfaces and skin and can cause serious infections inside the body. Antimicrobial peptides (AMPs) are part of the innate immune system and can eliminate pathogens, including bacteria and viruses, and are a promising alternative to antibiotics. Although studies have reported that AMP-functionalized hydrogels can prevent bacterial adhesion and biofilm formation, AMP dosing and the combined effects of multiple AMPs are not well understood. Here, three AMPs with different antibacterial properties were synthesized and the soluble minimum inhibitory concentrations (MICs) of each AMP against methicillin-susceptible S. aureus (MSSA) and methicillin-resistant S. aureus (MRSA) were determined. Hydrogels with immobilized AMPs at their MIC (DD13-RIP 27.5 µM; indolicidin 43.8 µM; P10 120 µM) were effective in preventing MRSA adhesion and biofilm formation. Checkerboard AMP screens identified synergy between indolicidin (3.1 µM) and P10 (12.5 µM) based on soluble fractional inhibitory concentration indices (FICIs) against MRSA, and hydrogels formed with these AMPs at half of their synergistic concentrations (total peptide concentration, 7.8 µM) were highly efficacious in killing MRSA. Mammalian cells cultured atop these hydrogels were highly viable, demonstrating that these AMP hydrogels are biocompatible and selectively eradicate bacteria, based on soluble checkerboard-screening data.
Collapse
Affiliation(s)
- Matthias Recktenwald
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA; (M.R.); (M.M.B.)
| | - Muskanjot Kaur
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA; (M.K.); (M.C.); (D.C.); (G.F.)
| | - Mohammed M. Benmassaoud
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA; (M.R.); (M.M.B.)
| | - Aryanna Copling
- Department of Translational Biomedical Sciences, Rowan University, Glassboro, NJ 08028, USA;
| | - Tulika Khanna
- Department of Biological Sciences, Rowan University, Glassboro, NJ 08028, USA;
| | - Michael Curry
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA; (M.K.); (M.C.); (D.C.); (G.F.)
| | - Dennise Cortes
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA; (M.K.); (M.C.); (D.C.); (G.F.)
| | - Gilbert Fleischer
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA; (M.K.); (M.C.); (D.C.); (G.F.)
| | - Valerie J. Carabetta
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA; (M.K.); (M.C.); (D.C.); (G.F.)
| | - Sebastián L. Vega
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA; (M.R.); (M.M.B.)
- Department of Orthopedic Surgery, Cooper Medical School of Rowan University, Camden, NJ 08103, USA
| |
Collapse
|
8
|
Lichota A, Gwozdzinski K, Kowalczyk E, Kowalczyk M, Sienkiewicz M. Contribution of staphylococcal virulence factors in the pathogenesis of thrombosis. Microbiol Res 2024; 283:127703. [PMID: 38537329 DOI: 10.1016/j.micres.2024.127703] [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: 08/23/2023] [Revised: 03/20/2024] [Accepted: 03/20/2024] [Indexed: 04/17/2024]
Abstract
Staphylococci are responsible for many infections in humans, starting with skin and soft tissue infections and finishing with invasive diseases such as endocarditis, sepsis and pneumonia, which lead to high mortality. Patients with sepsis often demonstrate activated clotting pathways, decreased levels of anticoagulants, decreased fibrinolysis, activated endothelial surfaces and activated platelets. This results in disseminated intravascular coagulation and formation of a microthrombus, which can lead to a multiorgan failure. This review describes various staphylococcal virulence factors that contribute to vascular thrombosis, including deep vein thrombosis in infected patients. The article presents mechanisms of action of different factors released by bacteria in various host defense lines, which in turn can lead to formation of blood clots in the vessels.
Collapse
Affiliation(s)
- Anna Lichota
- Department of Pharmaceutical Microbiology and Microbiological Diagnostics, Medical University of Lodz, Lodz, Poland.
| | | | - Edward Kowalczyk
- Department of Pharmacology and Toxicology, Medical University of Lodz, Lodz, Poland
| | | | - Monika Sienkiewicz
- Department of Pharmaceutical Microbiology and Microbiological Diagnostics, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
9
|
Tyagi JL, Gupta P, Ghate MM, Kumar D, Poluri KM. Assessing the synergistic potential of bacteriophage endolysins and antimicrobial peptides for eradicating bacterial biofilms. Arch Microbiol 2024; 206:272. [PMID: 38772980 DOI: 10.1007/s00203-024-04003-6] [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: 03/13/2024] [Accepted: 05/14/2024] [Indexed: 05/23/2024]
Abstract
Phage-encoded endolysins have emerged as a potential substitute to conventional antibiotics due to their exceptional benefits including host specificity, rapid host killing, least risk of resistance. In addition to their antibacterial potency and biofilm eradication properties, endolysins are reported to exhibit synergism with other antimicrobial agents. In this study, the synergistic potency of endolysins was dissected with antimicrobial peptides to enhance their therapeutic effectiveness. Recombinantly expressed and purified bacteriophage endolysin [T7 endolysin (T7L); and T4 endolysin (T4L)] proteins have been used to evaluate the broad-spectrum antibacterial efficacy using different bacterial strains. Antibacterial/biofilm eradication studies were performed in combination with different antimicrobial peptides (AMPs) such as colistin, nisin, and polymyxin B (PMB) to assess the endolysin's antimicrobial efficacy and their synergy with AMPs. In combination with T7L, polymyxin B and colistin effectively eradicated the biofilm of Pseudomonas aeruginosa and exhibited a synergistic effect. Further, a combination of T4L and nisin displayed a synergistic effect against Staphylococcus aureus biofilms. In summary, the obtained results endorse the theme of combinational therapy consisting of endolysins and AMPs as an effective remedy against the drug-resistant bacterial biofilms that are a serious concern in healthcare settings.
Collapse
Affiliation(s)
- Jaya Lakshmi Tyagi
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India
| | - Payal Gupta
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India
- Department of Biotechnology, Graphic Era Deemed to be University, Dehradun, Uttarakhand, 248001, India
| | - Mayur Mohan Ghate
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India
| | - Dinesh Kumar
- Centre of Bio-Medical Research, SGPGIMS, Lucknow, Uttar Pradesh, 226014, India
| | - Krishna Mohan Poluri
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India.
- Centre for Nanotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India.
| |
Collapse
|
10
|
Ostan NKH, Cole GB, Wang FZ, Reichheld SE, Moore G, Pan C, Yu R, Lai CCL, Sharpe S, Lee HO, Schryvers AB, Moraes TF. A secreted bacterial protein protects bacteria from cationic antimicrobial peptides by entrapment in phase-separated droplets. PNAS NEXUS 2024; 3:pgae139. [PMID: 38633880 PMCID: PMC11022072 DOI: 10.1093/pnasnexus/pgae139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 03/26/2024] [Indexed: 04/19/2024]
Abstract
Mammalian hosts combat bacterial infections through the production of defensive cationic antimicrobial peptides (CAPs). These immune factors are capable of directly killing bacterial invaders; however, many pathogens have evolved resistance evasion mechanisms such as cell surface modification, CAP sequestration, degradation, or efflux. We have discovered that several pathogenic and commensal proteobacteria, including the urgent human threat Neisseria gonorrhoeae, secrete a protein (lactoferrin-binding protein B, LbpB) that contains a low-complexity anionic domain capable of inhibiting the antimicrobial activity of host CAPs. This study focuses on a cattle pathogen, Moraxella bovis, that expresses the largest anionic domain of the LbpB homologs. We used an exhaustive biophysical approach employing circular dichroism, biolayer interferometry, cross-linking mass spectrometry, microscopy, size-exclusion chromatography with multi-angle light scattering coupled to small-angle X-ray scattering (SEC-MALS-SAXS), and NMR to understand the mechanisms of LbpB-mediated protection against CAPs. We found that the anionic domain of this LbpB displays an α-helical secondary structure but lacks a rigid tertiary fold. The addition of antimicrobial peptides derived from lactoferrin (i.e. lactoferricin) to the anionic domain of LbpB or full-length LbpB results in the formation of phase-separated droplets of LbpB together with the antimicrobial peptides. The droplets displayed a low rate of diffusion, suggesting that CAPs become trapped inside and are no longer able to kill bacteria. Our data suggest that pathogens, like M. bovis, leverage anionic intrinsically disordered domains for the broad recognition and neutralization of antimicrobials via the formation of biomolecular condensates.
Collapse
Affiliation(s)
- Nicholas K H Ostan
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Gregory B Cole
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Flora Zhiqi Wang
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Sean E Reichheld
- Molecular Medicine Program, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Gaelen Moore
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Chuxi Pan
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Ronghua Yu
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB T2N 4N1, Canada
| | | | - Simon Sharpe
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
- Molecular Medicine Program, Research Institute, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Hyun O Lee
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Anthony B Schryvers
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Trevor F Moraes
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
11
|
Zhao C, Yan S, Luo Y, Song Y, Xia X. Analyzing resistome in soil and Human gut: a study on the characterization and risk evaluation of antimicrobial peptide resistance. Front Microbiol 2024; 15:1352531. [PMID: 38591036 PMCID: PMC10999558 DOI: 10.3389/fmicb.2024.1352531] [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: 12/08/2023] [Accepted: 02/26/2024] [Indexed: 04/10/2024] Open
Abstract
Objective The limited existing knowledge regarding resistance to antimicrobial peptides (AMPs) is hindering their broad utilization. The aim of this study is to enhance the understanding of AMP resistance, a pivotal factor in the exploration of alternative drug development in response to the escalating challenge of antibiotic resistance. Methods We utilized metagenomic functional selection to analyze genes resistant to AMPs, with a specific focus on the microbiota in soil and the human gut. Through a combination of experimental methods and bioinformatics analyses, our investigation delved into the possibilities of the evolution of resistance to AMPs, as well as the transfer or interchange of resistance genes among the environment, the human body, and pathogens. Additionally, we examined the cross-resistance between AMPs and evaluated interactions among AMPs and conventional antibiotics. Results The presence of AMP resistance, including various resistance mechanisms, was observed in both soil and the human gut microbiota, as indicated by our findings. Significantly, the study underscored the facile evolution of AMP resistance and the potential for gene sharing or exchange among different environments. Notably, cross-resistance among AMPs was identified as a phenomenon, while cross-resistance between AMPs and antibiotics was found to be relatively infrequent. Conclusion The results of our study highlight the significance of taking a cautious stance when considering the extensive application of AMPs. It is imperative to thoroughly assess potential resistance risks, with a particular focus on the development of resistance to AMPs across diverse domains. A comprehensive grasp of these aspects is essential for making well-informed decisions and ensuring the responsible utilization of AMPs in the ongoing fight against antibiotic resistance.
Collapse
Affiliation(s)
| | | | | | - Yuzhu Song
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| | - Xueshan Xia
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
12
|
Sadecki PW, Laws GD, Morgan JJ, Wommack AJ, Nawrot R, Hicks LM. The Greater Celandine: Identification and Characterization of an Antimicrobial Peptide from Chelidonium majus. JOURNAL OF NATURAL PRODUCTS 2024; 87:544-553. [PMID: 38366995 PMCID: PMC10959680 DOI: 10.1021/acs.jnatprod.3c00939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/19/2024]
Abstract
Chelidonium majus, known as Greater Celandine, is a latex-bearing plant that has been leveraged for its anticancer and antimicrobial properties. Herein, C. majus aerial tissue is mined for the presence of antimicrobial peptides. A highly abundant cysteine-rich peptide with a length of 25 amino acids, deemed CM-AMP1, is characterized through multiple mass spectrometric approaches. Electron-activated dissociation is leveraged to differentiate between isoleucine and leucine residues and complement conventional collision-induced dissociation to gain full sequence coverage of the full-length peptide. CM-AMP1 shares little sequence similarity with any proteins in publicly available databases, highlighting the novelty of its cysteine landscape and core motif. The presence of three disulfide bonds in the native peptide confers proteolytic stability, and antimicrobial activity is greatly decreased upon the alkylation of the cysteine residues. Synthetic variants of CM-AMP1 are used to confirm the activity of the full-length sequence and the core motif. To assess the biological impact, E. coli was grown in a sublethal concentration of CM-AMP1 and quantitative proteomics was used to identify proteins produced by the bacteria under stress, ultimately suggesting a membrane lytic antimicrobial mechanism of action. This study integrates multiple analytical methods for molecular and biological characterization of a unique antimicrobial peptide identified from C. majus.
Collapse
Affiliation(s)
- Patric W Sadecki
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Garrett D Laws
- Department of Chemistry, High Point University, High Point, North Carolina 27268, United States
| | - Johnathon J Morgan
- Department of Chemistry, High Point University, High Point, North Carolina 27268, United States
| | - Andrew J Wommack
- Department of Chemistry, High Point University, High Point, North Carolina 27268, United States
| | - Robert Nawrot
- Department of Molecular Virology, Faculty of Biology, Institute of Experimental Biology, Adam Mickiewicz University, Poznań 61-712, Poland
| | - Leslie M Hicks
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
13
|
Sawant NV, Chang SS, Pandit KA, Khekare P, Warner WR, Zimmern PE, De Nisco NJ. VesiX cetylpyridinium chloride is rapidly bactericidal and reduces uropathogenic Escherichia coli bladder epithelial cell invasion in vitro. Microbiol Spectr 2024; 12:e0271223. [PMID: 38240572 PMCID: PMC10913388 DOI: 10.1128/spectrum.02712-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 12/11/2023] [Indexed: 03/06/2024] Open
Abstract
Management of urinary tract infection (UTI) in postmenopausal women can be challenging. The recent rise in resistance to most of the available oral antibiotic options together with high recurrence rate in postmenopausal women has further complicated treatment of UTI. As such, intravesical instillations of antibiotics like gentamicin are being investigated as an alternative to oral antibiotic therapies. This study evaluates the efficacy of the candidate intravesical therapeutic VesiX, a solution containing the cationic detergent Cetylpyridinium chloride, against a broad range of uropathogenic bacterial species clinically isolated from postmenopausal women with recurrent UTI (rUTI). We also evaluate the cytotoxicity of VesiX against cultured bladder epithelial cells and find that low concentrations of 0.0063% and 0.0125% provide significant bactericidal effect toward diverse bacterial species including uropathogenic Escherichia coli (UPEC), Klebsiella pneumoniae, Enterococcus faecalis, Pseudomonas aeruginosa, and Proteus mirabilis while minimizing cytotoxic effects against cultured 5637 bladder epithelial cells. Lastly, to begin to evaluate the potential utility of using VesiX in combination therapy with existing intravesical therapies for rUTI, we investigate the combined effects of VesiX and the intravesical antibiotic gentamicin. We find that VesiX and gentamicin are not antagonistic and are able to reduce levels of intracellular UPEC in cultured bladder epithelial cells. IMPORTANCE When urinary tract infections (UTIs), which affect over 50% of women, become resistant to available antibiotic therapies dangerous complications like kidney infection and lethal sepsis can occur. New therapeutic paradigms are needed to expand our arsenal against these difficult to manage infections. Our study investigates VesiX, a Cetylpyridinium chloride (CPC)-based therapeutic, as a candidate broad-spectrum antimicrobial agent for use in bladder instillation therapy for antibiotic-resistant UTI. CPC is a cationic surfactant that is FDA-approved for use in mouthwashes and is used as a food additive but has not been extensively evaluated as a UTI therapeutic. Our study is the first to investigate its rapid bactericidal kinetics against diverse uropathogenic bacterial species isolated from postmenopausal women with recurrent UTI and host cytotoxicity. We also report that together with the FDA-approved bladder-instillation agent gentamicin, VesiX was able to significantly reduce intracellular populations of uropathogenic bacteria in cultured bladder epithelial cells.
Collapse
Affiliation(s)
- Namrata V. Sawant
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, Texas, USA
| | - Samuel S. Chang
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, Texas, USA
| | - Krutika A. Pandit
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, Texas, USA
| | - Prachi Khekare
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, Texas, USA
| | | | - Philippe E. Zimmern
- Department of Urology, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Nicole J. De Nisco
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, Texas, USA
- Department of Urology, The University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
14
|
Murtha AN, Kazi M, Kim E, Rosch KM, Torres F, Dörr T. Multiple resistance factors collectively promote inoculum-dependent dynamic survival during antimicrobial peptide exposure in Enterobacter cloacae. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.03.583169. [PMID: 38463991 PMCID: PMC10925329 DOI: 10.1101/2024.03.03.583169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Antimicrobial peptides (AMPs) are a promising tool with which to fight rising antibiotic resistance. However, pathogenic bacteria are equipped with several AMP defense mechanisms, whose contributions to AMP resistance are often poorly defined. Here, we evaluate the genetic determinants of resistance to an insect AMP, cecropin B, in the opportunistic pathogen Enterobacter cloacae. Single-cell analysis of E. cloacae's response to cecropin revealed marked heterogeneity in cell survival, phenotypically reminiscent of heteroresistance (the ability of a subpopulation to grow in the presence of supra-MIC concentration of antimicrobial). The magnitude of this response was highly dependent on initial E. cloacae inoculum. We identified 3 genetic factors which collectively contribute to E. cloacae resistance in response to the AMP cecropin: The PhoPQ-two-component system, OmpT-mediated proteolytic cleavage of cecropin, and Rcs-mediated membrane stress response. Altogether, this evidence suggests that multiple, independent mechanisms contribute to AMP resistance in E. cloacae.
Collapse
Affiliation(s)
- Andrew N. Murtha
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
- Department of Microbiology, Cornell University, Ithaca, NY 14853, USA
| | - Misha Kazi
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
| | - Eileen Kim
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
| | - Kelly M. Rosch
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
| | - Facundo Torres
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
- Department of Microbiology, Cornell University, Ithaca, NY 14853, USA
| | - Tobias Dörr
- Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
- Department of Microbiology, Cornell University, Ithaca, NY 14853, USA
- Cornell Institute of Host-Microbe Interactions and Disease, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
15
|
Rajabloo A, Karimi J, Mehrabadi M. Differential induction of NF-κB pathways by non-pathogenic and pathogenic bacteria in Helicoverpa armigera is critical for an efficient immune response and survival. J Invertebr Pathol 2024; 203:108049. [PMID: 38159795 DOI: 10.1016/j.jip.2023.108049] [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: 10/12/2023] [Revised: 12/05/2023] [Accepted: 12/27/2023] [Indexed: 01/03/2024]
Abstract
Following pathogen infection in a host, extensive changes occur in the host's gene expression pattern to suppress infection and increase the chance of host survival. Likewise, many pathogens have evolved to evade/suppress host immunity and increase their survival within the host. In this study, we assessed the NF-κB (Imd and Toll) essential gene expression response of Helicoverpa armigera to an entomopathogenic Serratia marcescens and non-pathogenic Escherichia coli. Bacterial cells of S. marcescens or E. coli were injected into the haemocoel of fifth-instar larvae of H. armigera, whereas distilled water was injected into control insects. Our results showed that the expression levels of the Imd and Toll pathway genes (i.e., Relish, imd, spätzle and dif) and the antimicrobial peptides (i.e., gloverin, transferin, gallerimycin, and galiomicin) were differentially expressed following the bacterial injections while control larvae showed no differences. The E. coli injection induced higher and longer-lasted gene expression than the S. marcescens injected larvae, in which the gene expressions were diminished from 24 h post injection. Transcript Knockdown of Relish increased the replication rates of S. marcescens and E. coli, and lowered the infected larvae survival rates. These results showed that H. armigera NF-κB immunity pathways (particularly Imd pathway) play a vital role in immunity against bacterial infections, and S. marcescens might modulate these pathways to survive and replicate in the host.
Collapse
Affiliation(s)
- Akram Rajabloo
- Department of Agricultural Biotechnology, Faculty of Agriculture, Shahed University, Tehran, Iran
| | - Jaber Karimi
- Department of Plant Protection, Faculty of Agriculture, Shahed University, Tehran, Iran.
| | - Mohammad Mehrabadi
- Department of Entomology, Faculty of Agriculture, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
16
|
Do H, Li ZR, Tripathi PK, Mitra S, Guerra S, Dash A, Weerasekera D, Makthal N, Shams S, Aggarwal S, Singh BB, Gu D, Du Y, Olsen RJ, LaRock C, Zhang W, Kumaraswami M. Engineered probiotic overcomes pathogen defences using signal interference and antibiotic production to treat infection in mice. Nat Microbiol 2024; 9:502-513. [PMID: 38228859 PMCID: PMC10847043 DOI: 10.1038/s41564-023-01583-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 12/11/2023] [Indexed: 01/18/2024]
Abstract
Probiotic supplements are suggested to promote human health by preventing pathogen colonization. However, the mechanistic bases for their efficacy in vivo are largely uncharacterized. Here using metabolomics and bacterial genetics, we show that the human oral probiotic Streptococcus salivarius K12 (SAL) produces salivabactin, an antibiotic that effectively inhibits pathogenic Streptococcus pyogenes (GAS) in vitro and in mice. However, prophylactic dosing with SAL enhanced GAS colonization in mice and ex vivo in human saliva. We showed that, on co-colonization, GAS responds to a SAL intercellular peptide signal that controls SAL salivabactin production. GAS produces a secreted protease, SpeB, that targets SAL-derived salivaricins and enhances GAS survival. Using this knowledge, we re-engineered probiotic SAL to prevent signal eavesdropping by GAS and potentiate SAL antimicrobials. This engineered probiotic demonstrated superior efficacy in preventing GAS colonization in vivo. Our findings show that knowledge of interspecies interactions can identify antibiotic- and probiotic-based strategies to combat infection.
Collapse
Affiliation(s)
- Hackwon Do
- Center for Molecular and Translational Human Infectious Diseases Research, Houston Methodist Research Institute, Houston, TX, USA
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, USA
- Research unit of cryogenic novel material, Korea Polar Research Institute, Incheon, South Korea
| | - Zhong-Rui Li
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, USA
| | - Praveen Kumar Tripathi
- Center for Molecular and Translational Human Infectious Diseases Research, Houston Methodist Research Institute, Houston, TX, USA
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Sonali Mitra
- Center for Molecular and Translational Human Infectious Diseases Research, Houston Methodist Research Institute, Houston, TX, USA
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Stephanie Guerra
- Department of Microbiology and Immunology, Emory School of Medicine, Atlanta, GA, USA
| | - Ananya Dash
- Department of Microbiology and Immunology, Emory School of Medicine, Atlanta, GA, USA
| | - Dulanthi Weerasekera
- Center for Molecular and Translational Human Infectious Diseases Research, Houston Methodist Research Institute, Houston, TX, USA
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Nishanth Makthal
- Center for Molecular and Translational Human Infectious Diseases Research, Houston Methodist Research Institute, Houston, TX, USA
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Syed Shams
- Center for Molecular and Translational Human Infectious Diseases Research, Houston Methodist Research Institute, Houston, TX, USA
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Shifu Aggarwal
- Center for Molecular and Translational Human Infectious Diseases Research, Houston Methodist Research Institute, Houston, TX, USA
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Bharat Bhushan Singh
- Center for Molecular and Translational Human Infectious Diseases Research, Houston Methodist Research Institute, Houston, TX, USA
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, USA
| | - Di Gu
- Department of Chemistry, University of California, Berkeley, CA, USA
| | - Yongle Du
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, USA
| | - Randall J Olsen
- Center for Molecular and Translational Human Infectious Diseases Research, Houston Methodist Research Institute, Houston, TX, USA
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, USA
- Department of Pathology and Laboratory Medicine, Weill Medical College of Cornell University, New York, NY, USA
| | - Christopher LaRock
- Department of Microbiology and Immunology, Emory School of Medicine, Atlanta, GA, USA
- Department of Medicine, Division of Infectious Diseases, Emory School of Medicine, Atlanta, GA, USA
- Emory Antibiotic Resistance Center, Emory School of Medicine, Atlanta, GA, USA
| | - Wenjun Zhang
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| | - Muthiah Kumaraswami
- Center for Molecular and Translational Human Infectious Diseases Research, Houston Methodist Research Institute, Houston, TX, USA.
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, TX, USA.
| |
Collapse
|
17
|
Fleitas O, Fontes W, De Souza CM, Da Costa MC, Cardoso MH, Castro MS, Sousa MV, Ricart CAO, Ramada MHS, Duque HM, Porto WF, Silva ON, Franco OL. A proteomic perspective on the resistance response of Klebsiella pneumoniae to antimicrobial peptide PaDBS1R1. J Antimicrob Chemother 2024; 79:112-122. [PMID: 37966053 DOI: 10.1093/jac/dkad354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 10/31/2023] [Indexed: 11/16/2023] Open
Abstract
BACKGROUND The synthetic antimicrobial peptide, PaDBS1R1, has been reported as a powerful anti-Klebsiella pneumoniae antimicrobial. However, there is only scarce knowledge about whether K. pneumoniae could develop resistance against PaDBS1R1 and which resistance mechanisms could be involved. OBJECTIVES Identify via label-free shotgun proteomics the K. pneumoniae resistance mechanisms developed against PaDBS1R1. METHODS An adaptive laboratory evolution experiment was performed to obtain a PaDBS1R1-resistant K. pneumoniae lineage. Antimicrobial susceptibility was determined through microdilution assay. Modifications in protein abundances between the resistant and sensitive lineages were measured via label-free quantitative shotgun proteomics. Enriched Gene Ontology terms and KEGG pathways were identified through over-representation analysis. Data are available via ProteomeXchange with identifier PXD033020. RESULTS K. pneumoniae ATCC 13883 parental strain challenged with increased subinhibitory PaDBS1R1 concentrations allowed the PaDBS1R1-resistant K. pneumoniae lineage to emerge. Proteome comparisons between PaDBS1R1-resistant K. pneumoniae and PaDBS1R1-sensitive K. pneumoniae under PaDBS1R1-induced stress conditions enabled the identification and quantification of 1702 proteins, out of which 201 were differentially abundant proteins (DAPs). The profiled DAPs comprised 103 up-regulated proteins (adjusted P value < 0.05, fold change ≥ 2) and 98 down-regulated proteins (adjusted P value < 0.05, fold change ≤ 0.5). The enrichment analysis suggests that PhoPQ-guided LPS modifications and CpxRA-dependent folding machinery could be relevant resistance mechanisms against PaDBS1R1. CONCLUSIONS Based on experimental evolution and a label-free quantitative shotgun proteomic approach, we showed that K. pneumoniae developed resistance against PaDBS1R1, whereas PhoPQ-guided LPS modifications and CpxRA-dependent folding machinery appear to be relevant resistance mechanisms against PaDBS1R1.
Collapse
Affiliation(s)
- Osmel Fleitas
- Programa de Pós-Graduação em Patologia Molecular, Universidade de Brasília, Brasília, Brazil
- Centro de Análises Proteômicas e Bioquímicas, Universidade Católica de Brasília, Brasília, Brazil
| | - Wagner Fontes
- Programa de Pós-Graduação em Patologia Molecular, Universidade de Brasília, Brasília, Brazil
- Laboratório de Bioquímica e Química de Proteínas, Universidade de Brasília, Brasília, Brazil
| | - Camila M De Souza
- Centro de Análises Proteômicas e Bioquímicas, Universidade Católica de Brasília, Brasília, Brazil
- Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Brazil
| | - Mylena C Da Costa
- Centro de Análises Proteômicas e Bioquímicas, Universidade Católica de Brasília, Brasília, Brazil
| | - Marlon H Cardoso
- Centro de Análises Proteômicas e Bioquímicas, Universidade Católica de Brasília, Brasília, Brazil
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Brazil
- Instituto de Biociências (INBIO), Universidade Federal de Mato Grosso do Sul, Cidade Universitária, 79070900, Campo Grande, Mato Grosso do Sul, Brazil
| | - Mariana S Castro
- Programa de Pós-Graduação em Patologia Molecular, Universidade de Brasília, Brasília, Brazil
- Laboratório de Bioquímica e Química de Proteínas, Universidade de Brasília, Brasília, Brazil
| | - Marcelo V Sousa
- Programa de Pós-Graduação em Patologia Molecular, Universidade de Brasília, Brasília, Brazil
- Laboratório de Bioquímica e Química de Proteínas, Universidade de Brasília, Brasília, Brazil
| | - Carlos A O Ricart
- Programa de Pós-Graduação em Patologia Molecular, Universidade de Brasília, Brasília, Brazil
- Laboratório de Bioquímica e Química de Proteínas, Universidade de Brasília, Brasília, Brazil
| | - Marcelo H S Ramada
- Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Brazil
- Programa de Pós-Graduação em Gerontologia, Universidade Católica de Brasília, Brasília, Brazil
| | - Harry M Duque
- Centro de Análises Proteômicas e Bioquímicas, Universidade Católica de Brasília, Brasília, Brazil
| | - William F Porto
- Centro de Análises Proteômicas e Bioquímicas, Universidade Católica de Brasília, Brasília, Brazil
| | - Osmar N Silva
- Programa de Pós-graduação em Ciências Farmacêuticas, Universidade Evangélica de Anapólis, University City, 75083-515 Anápolis-GO, Brazil
| | - Octávio L Franco
- Programa de Pós-Graduação em Patologia Molecular, Universidade de Brasília, Brasília, Brazil
- Centro de Análises Proteômicas e Bioquímicas, Universidade Católica de Brasília, Brasília, Brazil
- Programa de Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Brazil
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Brazil
| |
Collapse
|
18
|
Savitskaya A, Masso-Silva J, Haddaoui I, Enany S. Exploring the arsenal of antimicrobial peptides: Mechanisms, diversity, and applications. Biochimie 2023; 214:216-227. [PMID: 37499896 DOI: 10.1016/j.biochi.2023.07.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 07/09/2023] [Accepted: 07/24/2023] [Indexed: 07/29/2023]
Abstract
Antimicrobial peptides (AMPs) are essential for defence against pathogens in all living organisms and possessed activities against bacteria, fungi, viruses, parasites and even cancer cells. AMPs are short peptides containing 12-100 amino acids conferring a net positive charge and an amphiphilic property in most cases. Although, anionic AMPs also exist. AMPs can be classified based on the types of secondary structures, charge, hydrophobicity, amino acid composition, length, etc. Their mechanism of action usually includes a membrane disruption process through pore formation (three different models have been described, barrel-stave, toroidal or carpet model) but AMPs can also penetrate and impair intracellular functions. Besides their activity against pathogens, they have also shown immunomodulatory properties in complex scenarios through many different interactions. The aim of this review to summarize knowledge about AMP's and discuss the potential application of AMPs as therapeutics, the challenges due to their limitations, including their susceptibility to degradation, the potential generation of AMP resistance, cost, etc. We also discuss the current FDA-approved drugs based on AMPs and strategies to circumvent natural AMPs' limitations.
Collapse
Affiliation(s)
- Anna Savitskaya
- Institute of Bioorganic Chemistry of Russian Academy of Science, Moscow, Russian Federation
| | - Jorge Masso-Silva
- Division of Pulmonary, Critical Care, Sleep Medicine and Physiology, University of California San Diego, La Jolla, CA, USA
| | - Imen Haddaoui
- National Research Institute of Rural Engineering, Water and Forestry, University of Carthage, LR Valorization of Unconventional Waters, Ariana, Tunisia
| | - Shymaa Enany
- Microbiology and Immunology Department, Faculty of Pharmacy, Suez Canal University, Ismailia, Egypt; Biomedical Research Department, Armed Force College of Medicine, Cairo, Egypt.
| |
Collapse
|
19
|
Yang S, Duncan GA. Synthetic mucus biomaterials for antimicrobial peptide delivery. J Biomed Mater Res A 2023; 111:1616-1626. [PMID: 37199137 PMCID: PMC10524183 DOI: 10.1002/jbm.a.37559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/25/2023] [Accepted: 05/08/2023] [Indexed: 05/19/2023]
Abstract
Despite the promise of antimicrobial peptides (AMPs) as treatments for antibiotic-resistant infections, their therapeutic efficacy is limited due to the rapid degradation and low bioavailability of AMPs. To address this, we have developed and characterized a synthetic mucus (SM) biomaterial capable of delivering LL37 AMPs and enhancing their therapeutic effect. LL37 is an AMP that exhibits a wide range of antimicrobial activity against bacteria, including Pseudomonas aeruginosa. LL37 loaded SM hydrogels demonstrated controlled release with 70%-95% of loaded LL37 over 8 h due to charge-mediated interactions between mucins and LL37 AMPs. Compared to treatment with LL37 alone where antimicrobial activity was reduced after 3 h, LL37-SM hydrogels inhibited P. aeruginosa (PAO1) growth over 12 h. LL37-SM hydrogel treatment reduced PAO1 viability over 6 h whereas a rebound in bacterial growth was observed when treated with LL37 only. These data demonstrate LL37-SM hydrogels enhance antimicrobial activity by preserving LL37 AMP activity and bioavailability. Overall, this work establishes SM biomaterials as a platform for enhanced AMP delivery for antimicrobial applications.
Collapse
Affiliation(s)
- Sydney Yang
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
| | - Gregg A Duncan
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
| |
Collapse
|
20
|
Gill CP, Phan C, Platt V, Worrell D, Andl T, Roy H. The MprF homolog LysX synthesizes lysyl-diacylglycerol contributing to antibiotic resistance and virulence. Microbiol Spectr 2023; 11:e0142923. [PMID: 37768052 PMCID: PMC10580965 DOI: 10.1128/spectrum.01429-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/10/2023] [Indexed: 09/29/2023] Open
Abstract
Lysyl-diacylglycerol (Lys-DAG) was identified three decades ago in Mycobacterium phlei, but the biosynthetic pathway and function of this aminoacylated lipid have since remained uncharacterized. Combining genetic methods, mass spectrometry, and biochemical approaches, we show that the multiple peptide resistance factor (MprF) homolog LysX from Corynebacterium pseudotuberculosis and two mycobacterial species is responsible for Lys-DAG synthesis. LysX is conserved in most Actinobacteria and was previously implicated in the synthesis of another modified lipid, lysyl-phosphatidylglycerol (Lys-PG), in Mycobacterium tuberculosis. Although we detected low levels of Lys-PG in the membrane of C. pseudotuberculosis, our data suggest that Lys-PG is not directly synthesized by LysX and may require an additional downstream pathway, which is as yet undefined. Our results show that LysX in C. pseudotuberculosis is a major factor of resistance against a variety of positively charged antibacterial agents, including cationic antimicrobial peptides (e.g., human peptide LL-37 and polymyxin B) and aminoglycosides (e.g., gentamycin and apramycin). Deletion of lysX caused an increase in cellular membrane permeability without dissipation of the membrane potential, suggesting that loss of the protein does not result in mechanical damage to the cell membrane. Furthermore, lysX-deficient cells exhibited an attenuated virulence phenotype in a Galleria mellonella infection model, supporting a role for LysX during infection. Altogether, Lys-DAG represents a novel molecular determinant for antimicrobial resistance and virulence that may be widespread in Actinobacteria and points to a richer landscape than previously realized of lipid components contributing to overall membrane physiology in this important bacterial phylum. IMPORTANCE In the past two decades, tRNA-dependent modification of membrane phosphatidylglycerol has been implicated in altering the biochemical properties of the cell surface, thereby enhancing the antimicrobial resistance and virulence of various bacterial pathogens. Here, we show that in several Actinobacteria, the multifunctional protein LysX attaches lysine to diacylglycerol instead of phosphatidylglycerol. We found that lysyl-diacylglycerol (Lys-DAG) confers high levels of resistance against various cationic antimicrobial peptides and aminoglycosides and also enhances virulence. Our data show that Lys-DAG is a lipid commonly found in important actinobacterial pathogens, including Mycobacterium and Corynebacterium species.
Collapse
Affiliation(s)
- Cameron P. Gill
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Christopher Phan
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Vivien Platt
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Danielle Worrell
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Thomas Andl
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| | - Hervé Roy
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, USA
| |
Collapse
|
21
|
Cheung GYC, Otto M. Virulence Mechanisms of Staphylococcal Animal Pathogens. Int J Mol Sci 2023; 24:14587. [PMID: 37834035 PMCID: PMC10572719 DOI: 10.3390/ijms241914587] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 09/20/2023] [Accepted: 09/23/2023] [Indexed: 10/15/2023] Open
Abstract
Staphylococci are major causes of infections in mammals. Mammals are colonized by diverse staphylococcal species, often with moderate to strong host specificity, and colonization is a common source of infection. Staphylococcal infections of animals not only are of major importance for animal well-being but have considerable economic consequences, such as in the case of staphylococcal mastitis, which costs billions of dollars annually. Furthermore, pet animals can be temporary carriers of strains infectious to humans. Moreover, antimicrobial resistance is a great concern in livestock infections, as there is considerable antibiotic overuse, and resistant strains can be transferred to humans. With the number of working antibiotics continuously becoming smaller due to the concomitant spread of resistant strains, alternative approaches, such as anti-virulence, are increasingly being investigated to treat staphylococcal infections. For this, understanding the virulence mechanisms of animal staphylococcal pathogens is crucial. While many virulence factors have similar functions in humans as animals, there are increasingly frequent reports of host-specific virulence factors and mechanisms. Furthermore, we are only beginning to understand virulence mechanisms in animal-specific staphylococcal pathogens. This review gives an overview of animal infections caused by staphylococci and our knowledge about the virulence mechanisms involved.
Collapse
Affiliation(s)
| | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 50 South Drive, Bethesda, MD 20814, USA;
| |
Collapse
|
22
|
Guryanova SV, Balandin SV, Belogurova-Ovchinnikova OY, Ovchinnikova TV. Marine Invertebrate Antimicrobial Peptides and Their Potential as Novel Peptide Antibiotics. Mar Drugs 2023; 21:503. [PMID: 37888438 PMCID: PMC10608444 DOI: 10.3390/md21100503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/14/2023] [Accepted: 09/21/2023] [Indexed: 10/28/2023] Open
Abstract
Marine invertebrates constantly interact with a wide range of microorganisms in their aquatic environment and possess an effective defense system that has enabled their existence for millions of years. Their lack of acquired immunity sets marine invertebrates apart from other marine animals. Invertebrates could rely on their innate immunity, providing the first line of defense, survival, and thriving. The innate immune system of marine invertebrates includes various biologically active compounds, and specifically, antimicrobial peptides. Nowadays, there is a revive of interest in these peptides due to the urgent need to discover novel drugs against antibiotic-resistant bacterial strains, a pressing global concern in modern healthcare. Modern technologies offer extensive possibilities for the development of innovative drugs based on these compounds, which can act against bacteria, fungi, protozoa, and viruses. This review focuses on structural peculiarities, biological functions, gene expression, biosynthesis, mechanisms of antimicrobial action, regulatory activities, and prospects for the therapeutic use of antimicrobial peptides derived from marine invertebrates.
Collapse
Affiliation(s)
- Svetlana V. Guryanova
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (S.V.G.); (S.V.B.)
- Medical Institute, Peoples’ Friendship University of Russia, 117198 Moscow, Russia
| | - Sergey V. Balandin
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (S.V.G.); (S.V.B.)
| | | | - Tatiana V. Ovchinnikova
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia; (S.V.G.); (S.V.B.)
- Phystech School of Biological and Medical Physics, Moscow Institute of Physics and Technology, 141701 Dolgoprudny, Russia;
- Department of Biotechnology, I.M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| |
Collapse
|
23
|
Rangel K, Lechuga GC, Provance DW, Morel CM, De Simone SG. An Update on the Therapeutic Potential of Antimicrobial Peptides against Acinetobacter baumannii Infections. Pharmaceuticals (Basel) 2023; 16:1281. [PMID: 37765087 PMCID: PMC10537560 DOI: 10.3390/ph16091281] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 08/09/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023] Open
Abstract
The rise in antibiotic-resistant strains of clinically important pathogens is a major threat to global health. The World Health Organization (WHO) has recognized the urgent need to develop alternative treatments to address the growing list of priority pathogens. Antimicrobial peptides (AMPs) rank among the suggested options with proven activity and high potential to be developed into effective drugs. Many AMPs are naturally produced by living organisms protecting the host against pathogens as a part of their innate immunity. Mechanisms associated with AMP actions include cell membrane disruption, cell wall weakening, protein synthesis inhibition, and interference in nucleic acid dynamics, inducing apoptosis and necrosis. Acinetobacter baumannii is a critical pathogen, as severe clinical implications have developed from isolates resistant to current antibiotic treatments and conventional control procedures, such as UV light, disinfectants, and drying. Here, we review the natural AMPs representing primary candidates for new anti-A. baumannii drugs in post-antibiotic-era and present computational tools to develop the next generation of AMPs with greater microbicidal activity and reduced toxicity.
Collapse
Affiliation(s)
- Karyne Rangel
- Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation in Neglected Population Diseases (INCT-IDPN), Oswaldo Cruz Institut, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil; (K.R.); (G.C.L.); (D.W.P.J.); (C.M.M.)
- Epidemiology and Molecular Systematics Laboratory (LEMS), Oswaldo Cruz Institut, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil
| | - Guilherme Curty Lechuga
- Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation in Neglected Population Diseases (INCT-IDPN), Oswaldo Cruz Institut, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil; (K.R.); (G.C.L.); (D.W.P.J.); (C.M.M.)
- Epidemiology and Molecular Systematics Laboratory (LEMS), Oswaldo Cruz Institut, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil
| | - David W. Provance
- Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation in Neglected Population Diseases (INCT-IDPN), Oswaldo Cruz Institut, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil; (K.R.); (G.C.L.); (D.W.P.J.); (C.M.M.)
- Epidemiology and Molecular Systematics Laboratory (LEMS), Oswaldo Cruz Institut, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil
| | - Carlos M. Morel
- Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation in Neglected Population Diseases (INCT-IDPN), Oswaldo Cruz Institut, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil; (K.R.); (G.C.L.); (D.W.P.J.); (C.M.M.)
| | - Salvatore G. De Simone
- Center for Technological Development in Health (CDTS), National Institute of Science and Technology for Innovation in Neglected Population Diseases (INCT-IDPN), Oswaldo Cruz Institut, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil; (K.R.); (G.C.L.); (D.W.P.J.); (C.M.M.)
- Epidemiology and Molecular Systematics Laboratory (LEMS), Oswaldo Cruz Institut, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil
- Program of Post-Graduation on Science and Biotechnology, Department of Molecular and Cellular Biology, Biology Institute, Federal Fluminense University, Niterói 22040-036, RJ, Brazil
- Program of Post-Graduation on Parasitic Biology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-900, RJ, Brazil
| |
Collapse
|
24
|
Arias-Rojas A, Frahm D, Hurwitz R, Brinkmann V, Iatsenko I. Resistance to host antimicrobial peptides mediates resilience of gut commensals during infection and aging in Drosophila. Proc Natl Acad Sci U S A 2023; 120:e2305649120. [PMID: 37639605 PMCID: PMC10483595 DOI: 10.1073/pnas.2305649120] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 08/04/2023] [Indexed: 08/31/2023] Open
Abstract
Resilience to short-term perturbations, like inflammation, is a fundamental feature of microbiota, yet the underlying mechanisms of microbiota resilience are incompletely understood. Here, we show that Lactiplantibacillus plantarum, a major Drosophila commensal, stably colonizes the fruit fly gut during infection and is resistant to Drosophila antimicrobial peptides (AMPs). By transposon screening, we identified L. plantarum mutants sensitive to AMPs. These mutants were impaired in peptidoglycan O-acetylation or teichoic acid D-alanylation, resulting in increased negative cell surface charge and higher affinity to cationic AMPs. AMP-sensitive mutants were cleared from the gut after infection and aging-induced gut inflammation in wild-type, but not in AMP-deficient flies, suggesting that resistance to host AMPs is essential for commensal resilience in an inflamed gut environment. Thus, our work reveals that in addition to the host immune tolerance to the microbiota, commensal-encoded resilience mechanisms are necessary to maintain the stable association between host and microbiota during inflammation.
Collapse
Affiliation(s)
- Aranzazu Arias-Rojas
- Research group Genetics of host–microbe interactions, Max Planck Institute for Infection Biology, Berlin10117, Germany
- Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin, Berlin14195, Germany
| | - Dagmar Frahm
- Research group Genetics of host–microbe interactions, Max Planck Institute for Infection Biology, Berlin10117, Germany
| | - Robert Hurwitz
- Protein Purification Core Facility, Max Planck Institute for Infection Biology, Berlin10117, Germany
| | - Volker Brinkmann
- Microscopy Core Facility, Max Planck Institute for Infection Biology, Berlin10117, Germany
| | - Igor Iatsenko
- Research group Genetics of host–microbe interactions, Max Planck Institute for Infection Biology, Berlin10117, Germany
| |
Collapse
|
25
|
Ladewig L, Gloy L, Langfeldt D, Pinnow N, Weiland-Bräuer N, Schmitz RA. Antimicrobial Peptides Originating from Expression Libraries of Aurelia aurita and Mnemiopsis leidyi Prevent Biofilm Formation of Opportunistic Pathogens. Microorganisms 2023; 11:2184. [PMID: 37764028 PMCID: PMC10537229 DOI: 10.3390/microorganisms11092184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/17/2023] [Accepted: 08/26/2023] [Indexed: 09/29/2023] Open
Abstract
The demand for novel antimicrobial compounds is rapidly growing due to the rising appearance of antibiotic resistance in bacteria; accordingly, alternative approaches are urgently needed. Antimicrobial peptides (AMPs) are promising, since they are a naturally occurring part of the innate immune system and display remarkable broad-spectrum activity and high selectivity against various microbes. Marine invertebrates are a primary resource of natural AMPs. Consequently, cDNA expression (EST) libraries from the Cnidarian moon jellyfish Aurelia aurita and the Ctenophore comb jelly Mnemiopsis leidyi were constructed in Escherichia coli. Cell-free size-fractionated cell extracts (<3 kDa) of the two libraries (each with 29,952 clones) were consecutively screened for peptides preventing the biofilm formation of opportunistic pathogens using the crystal violet assay. The 3 kDa fraction of ten individual clones demonstrated promising biofilm-preventing activities against Klebsiella oxytoca and Staphylococcus epidermidis. Sequencing the respective activity-conferring inserts allowed for the identification of small ORFs encoding peptides (10-22 aa), which were subsequently chemically synthesized to validate their inhibitory potential. Although the peptides are likely artificial products from a random translation of EST inserts, the biofilm-preventing effects against K. oxytoca, Pseudomonas aeruginosa, S. epidermidis, and S. aureus were verified for five synthetic peptides in a concentration-dependent manner, with peptide BiP_Aa_5 showing the strongest effects. The impact of BiP_Aa_2, BiP_Aa_5, and BiP_Aa_6 on the dynamic biofilm formation of K. oxytoca was further validated in microfluidic flow cells, demonstrating a significant reduction in biofilm thickness and volume by BiP_Aa_2 and BiP_Aa_5. Overall, the structural characteristics of the marine invertebrate-derived AMPs, their physicochemical properties, and their promising antibiofilm effects highlight them as attractive candidates for discovering new antimicrobials.
Collapse
Affiliation(s)
- Lisa Ladewig
- General Microbiology, Kiel University, Am Botanischen Garten 1-9, 24118 Kiel, Germany
| | - Leon Gloy
- General Microbiology, Kiel University, Am Botanischen Garten 1-9, 24118 Kiel, Germany
| | - Daniela Langfeldt
- General Microbiology, Kiel University, Am Botanischen Garten 1-9, 24118 Kiel, Germany
- Institute of Clinical Molecular Biology (IKMB), Kiel University, Am Botanischen Garten 11, 24118 Kiel, Germany
| | - Nicole Pinnow
- General Microbiology, Kiel University, Am Botanischen Garten 1-9, 24118 Kiel, Germany
| | - Nancy Weiland-Bräuer
- General Microbiology, Kiel University, Am Botanischen Garten 1-9, 24118 Kiel, Germany
| | - Ruth A. Schmitz
- General Microbiology, Kiel University, Am Botanischen Garten 1-9, 24118 Kiel, Germany
| |
Collapse
|
26
|
Akintayo SO, Hosseini B, Vahidinasab M, Messmer M, Pfannstiel J, Bertsche U, Hubel P, Henkel M, Hausmann R, Voegele RT, Lilge L. Characterization ofantifungal properties of lipopeptide-producing Bacillus velezensis strains and their proteome-based response to the phytopathogens, Diaporthe spp. Front Bioeng Biotechnol 2023; 11:1228386. [PMID: 37609113 PMCID: PMC10440741 DOI: 10.3389/fbioe.2023.1228386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 07/28/2023] [Indexed: 08/24/2023] Open
Abstract
Introduction: B. velezensis strains are of interest in agricultural applications due to their beneficial interactions with plants, notable through their antimicrobial activity. The biocontrol ability of two new lipopeptides-producing B. velezensis strains ES1-02 and EFSO2-04, against fungal phytopathogens of Diaporthe spp., was evaluated and compared with reference strains QST713 and FZB42. All strains were found to be effective against the plant pathogens, with the new strains showing comparable antifungal activity to QST713 and slightly lower activity than FZB42. Methods: Lipopeptides and their isoforms were identified by high-performance thin-layer chromatography (HPTLC) and mass spectrometric measurements. The associated antifungal influences were determined in direct in vitro antagonistic dual culture assays, and the inhibitory growth effects on Diaporthe spp. as representatives of phytopathogenic fungi were determined. The effects on bacterial physiology of selected B. velezensis strains were analyzed by mass spectrometric proteomic analyses using nano-LC-MS/MS. Results and Discussion: Lipopeptide production analysis revealed that all strains produced surfactin, and one lipopeptide of the iturin family, including bacillomycin L by ES1-02 and EFSO2-04, while QST713 and FZB42 produced iturin A and bacillomycin D, respectively. Fengycin production was however only detected in the reference strains. As a result of co-incubation of strain ES1-02 with the antagonistic phytopathogen D. longicolla, an increase in surfactin production of up to 10-fold was observed, making stress induction due to competitors an attractive strategy for surfactin bioproduction. An associated global proteome analysis showed a more detailed overview about the adaptation and response mechanisms of B. velezensis, including an increased abundance of proteins associated with the biosynthesis of antimicrobial compounds. Furthermore, higher abundance was determined for proteins associated with oxidative, nitrosative, and general stress response. In contrast, proteins involved in phosphate uptake, amino acid transport, and translation were decreased in abundance. Altogether, this study provides new insights into the physiological adaptation of lipopeptide-producing B. velezensis strains, which show the potential for use as biocontrol agents with respect to phytopathogenic fungi.
Collapse
Affiliation(s)
- Stephen Olusanmi Akintayo
- Department of Bioprocess Engineering, Institute of Food Science and Biotechnology, University of Hohenheim, Stuttgart, Germany
| | - Behnoush Hosseini
- Department of Phytopathology, Institute of Phytomedicine, University of Hohenheim, Stuttgart, Germany
| | - Maliheh Vahidinasab
- Department of Bioprocess Engineering, Institute of Food Science and Biotechnology, University of Hohenheim, Stuttgart, Germany
| | - Marc Messmer
- Department of Bioprocess Engineering, Institute of Food Science and Biotechnology, University of Hohenheim, Stuttgart, Germany
| | - Jens Pfannstiel
- Core Facility Hohenheim, Mass Spectrometry Core Facility, University of Hohenheim, Stuttgart, Germany
| | - Ute Bertsche
- Core Facility Hohenheim, Mass Spectrometry Core Facility, University of Hohenheim, Stuttgart, Germany
| | - Philipp Hubel
- Core Facility Hohenheim, Mass Spectrometry Core Facility, University of Hohenheim, Stuttgart, Germany
| | - Marius Henkel
- Cellular Agriculture, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Rudolf Hausmann
- Department of Bioprocess Engineering, Institute of Food Science and Biotechnology, University of Hohenheim, Stuttgart, Germany
| | - Ralf T. Voegele
- Department of Phytopathology, Institute of Phytomedicine, University of Hohenheim, Stuttgart, Germany
| | - Lars Lilge
- Department of Bioprocess Engineering, Institute of Food Science and Biotechnology, University of Hohenheim, Stuttgart, Germany
- Department of Molecular Genetics, University of Groningen, Groningen, Netherlands
| |
Collapse
|
27
|
Zhou L, Ma L, Liu L, Sun S, Jing X, Lu Z. The Effects of Diet on the Immune Responses of the Oriental Armyworm Mythimna separata. INSECTS 2023; 14:685. [PMID: 37623395 PMCID: PMC10455674 DOI: 10.3390/insects14080685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/31/2023] [Accepted: 07/31/2023] [Indexed: 08/26/2023]
Abstract
Nutrients can greatly affect host immune defenses against infection. Possessing a simple immune system, insects have been widely used as models to address the relationships between nutrition and immunity. The effects of high versus low protein-to-carbohydrate ratio (P:C) diets on insect immune responses vary in different studies. To reveal the dietary manipulation of immune responses in the polyphagous agricultural pest oriental armyworm, we examined immune gene expression, phenoloxidase (PO) activity, and phagocytosis to investigate the immune traits of bacteria-challenged oriental armyworms, which were fed different P:C ratio diets. We found the oriental armyworms that were fed a 35:7 (P:C) diet showed higher phenoloxidase (PO) activity and stronger melanization, and those reared on a 28:14 (P:C) diet showed higher antimicrobial activity. However, different P:C diets had no apparent effect on the hemocyte number and phagocytosis. These results overall indicate that high P:C diets differently optimize humoral immune defense responses in oriental armyworms, i.e., PO-mediated melanization and antimicrobial peptide synthesis in response to bacteria challenge.
Collapse
Affiliation(s)
- Lizhen Zhou
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling 712100, China; (L.Z.); (L.M.); (L.L.); (S.S.); (X.J.)
| | - Li Ma
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling 712100, China; (L.Z.); (L.M.); (L.L.); (S.S.); (X.J.)
| | - Lu Liu
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling 712100, China; (L.Z.); (L.M.); (L.L.); (S.S.); (X.J.)
| | - Shaolei Sun
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling 712100, China; (L.Z.); (L.M.); (L.L.); (S.S.); (X.J.)
| | - Xiangfeng Jing
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling 712100, China; (L.Z.); (L.M.); (L.L.); (S.S.); (X.J.)
- Key Laboratory of Plant Protection Resources and Pest Management of Ministry of Education, Northwest A&F University, Yangling 712100, China
| | - Zhiqiang Lu
- Department of Entomology, College of Plant Protection, Northwest A&F University, Yangling 712100, China; (L.Z.); (L.M.); (L.L.); (S.S.); (X.J.)
- Key Laboratory of Plant Protection Resources and Pest Management of Ministry of Education, Northwest A&F University, Yangling 712100, China
- State Key Laboratory of Crop Stress Biology for Arid Areas, Northwest A&F University, Yangling 712100, China
- Key Laboratory of Integrated Pest Management on Loess Plateau of Ministry of Agriculture and Rural Affairs, Northwest A&F University, Yangling 712100, China
| |
Collapse
|
28
|
Jakubec M, Rylandsholm FG, Rainsford P, Silk M, Bril'kov M, Kristoffersen T, Juskewitz E, Ericson JU, Svendsen JSM. Goldilocks Dilemma: LPS Works Both as the Initial Target and a Barrier for the Antimicrobial Action of Cationic AMPs on E. coli. Biomolecules 2023; 13:1155. [PMID: 37509189 PMCID: PMC10377513 DOI: 10.3390/biom13071155] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Antimicrobial peptides (AMPs) are generally membrane-active compounds that physically disrupt bacterial membranes. Despite extensive research, the precise mode of action of AMPs is still a topic of great debate. This work demonstrates that the initial interaction between the Gram-negative E. coli and AMPs is driven by lipopolysaccharides (LPS) that act as kinetic barriers for the binding of AMPs to the bacterial membrane. A combination of SPR and NMR experiments provide evidence suggesting that cationic AMPs first bind to the negatively charged LPS before reaching a binding place in the lipid bilayer. In the event that the initial LPS-binding is too strong (corresponding to a low dissociation rate), the cationic AMPs cannot effectively get from the LPS to the membrane, and their antimicrobial potency will thus be diminished. On the other hand, the AMPs must also be able to effectively interact with the membrane to exert its activity. The ability of the studied cyclic hexapeptides to bind LPS and to translocate into a lipid membrane is related to the nature of the cationic charge (arginine vs. lysine) and to the distribution of hydrophobicity along the molecule (alternating vs. clumped tryptophan).
Collapse
Affiliation(s)
- Martin Jakubec
- Department of Chemistry, Faculty of Science and Technology, UiT the Arctic University of Norway, 9019 Tromsø, Norway
| | - Fredrik G Rylandsholm
- Department of Chemistry, Faculty of Science and Technology, UiT the Arctic University of Norway, 9019 Tromsø, Norway
| | - Philip Rainsford
- Department of Chemistry, Faculty of Science and Technology, UiT the Arctic University of Norway, 9019 Tromsø, Norway
| | - Mitchell Silk
- Department of Chemistry, Faculty of Science and Technology, UiT the Arctic University of Norway, 9019 Tromsø, Norway
| | - Maxim Bril'kov
- Department of Pharmacy, Faculty of Health Sciences, UiT the Arctic University of Norway, 9019 Tromsø, Norway
| | - Tone Kristoffersen
- Department of Chemistry, Faculty of Science and Technology, UiT the Arctic University of Norway, 9019 Tromsø, Norway
| | - Eric Juskewitz
- Department of Medical Biology, Faculty of Health Sciences, UiT the Arctic University of Norway, 9019 Tromsø, Norway
| | - Johanna U Ericson
- Department of Medical Biology, Faculty of Health Sciences, UiT the Arctic University of Norway, 9019 Tromsø, Norway
| | - John Sigurd M Svendsen
- Department of Chemistry, Faculty of Science and Technology, UiT the Arctic University of Norway, 9019 Tromsø, Norway
| |
Collapse
|
29
|
Hsieh RC, Liu R, Burgin DJ, Otto M. Understanding mechanisms of virulence in MRSA: implications for antivirulence treatment strategies. Expert Rev Anti Infect Ther 2023; 21:911-928. [PMID: 37501364 DOI: 10.1080/14787210.2023.2242585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 07/26/2023] [Indexed: 07/29/2023]
Abstract
INTRODUCTION Methicillin-resistant Staphylococcus aureus (MRSA) is a widespread pathogen, often causing recurrent and deadly infections in the hospital and community. Many S. aureus virulence factors have been suggested as potential targets for antivirulence therapy to decrease the threat of diminishing antibiotic availability. Antivirulence methods hold promise due to their adjunctive and prophylactic potential and decreased risk for selective pressure. AREAS COVERED This review describes the dominant virulence mechanisms exerted by MRSA and antivirulence therapeutics that are currently undergoing testing in clinical or preclinical stages. We also discuss the advantages and downsides of several investigational antivirulence approaches, including the targeting of bacterial transporters, host-directed therapy, and quorum-sensing inhibitors. For this review, a systematic search of literature on PubMed, Google Scholar, and Web of Science for relevant search terms was performed in April and May 2023. EXPERT OPINION Vaccine and antibody strategies have failed in clinical trials and could benefit from more basic science-informed approaches. Antivirulence-targeting approaches need to be set up better to meet the requirements of drug development, rather than only providing limited results to provide 'proof-of-principle' translational value of pathogenesis research. Nevertheless, there is great potential of such strategies and potential particular promise for novel probiotic approaches.
Collapse
Affiliation(s)
- Roger C Hsieh
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases (NIAID), U.S. National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Ryan Liu
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases (NIAID), U.S. National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Dylan J Burgin
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases (NIAID), U.S. National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases (NIAID), U.S. National Institutes of Health (NIH), Bethesda, Maryland, USA
| |
Collapse
|
30
|
Witzany C, Rolff J, Regoes RR, Igler C. The pharmacokinetic-pharmacodynamic modelling framework as a tool to predict drug resistance evolution. MICROBIOLOGY (READING, ENGLAND) 2023; 169:001368. [PMID: 37522891 PMCID: PMC10433423 DOI: 10.1099/mic.0.001368] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 07/12/2023] [Indexed: 08/01/2023]
Abstract
Pharmacokinetic-pharmacodynamic (PKPD) models, which describe how drug concentrations change over time and how that affects pathogen growth, have proven highly valuable in designing optimal drug treatments aimed at bacterial eradication. However, the fast rise of antimicrobial resistance calls for increased focus on an additional treatment optimization criterion: avoidance of resistance evolution. We demonstrate here how coupling PKPD and population genetics models can be used to determine treatment regimens that minimize the potential for antimicrobial resistance evolution. Importantly, the resulting modelling framework enables the assessment of resistance evolution in response to dynamic selection pressures, including changes in antimicrobial concentration and the emergence of adaptive phenotypes. Using antibiotics and antimicrobial peptides as an example, we discuss the empirical evidence and intuition behind individual model parameters. We further suggest several extensions of this framework that allow a more comprehensive and realistic prediction of bacterial escape from antimicrobials through various phenotypic and genetic mechanisms.
Collapse
Affiliation(s)
| | - Jens Rolff
- Evolutionary Biology, Institute for Biology, Freie Universität Berlin, Berlin, Germany
| | - Roland R. Regoes
- Institute of Integrative Biology, ETH Zurich, Zurich, Switzerland
| | - Claudia Igler
- Institute of Integrative Biology, ETH Zurich, Zurich, Switzerland
- School of Biological Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
31
|
Barman P, Joshi S, Sharma S, Preet S, Sharma S, Saini A. Strategic Approaches to Improvise Peptide Drugs as Next Generation Therapeutics. Int J Pept Res Ther 2023; 29:61. [PMID: 37251528 PMCID: PMC10206374 DOI: 10.1007/s10989-023-10524-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2023] [Indexed: 05/31/2023]
Abstract
In recent years, the occurrence of a wide variety of drug-resistant diseases has led to an increase in interest in alternate therapies. Peptide-based drugs as an alternate therapy hold researchers' attention in various therapeutic fields such as neurology, dermatology, oncology, metabolic diseases, etc. Previously, they had been overlooked by pharmaceutical companies due to certain limitations such as proteolytic degradation, poor membrane permeability, low oral bioavailability, shorter half-life, and poor target specificity. Over the last two decades, these limitations have been countered by introducing various modification strategies such as backbone and side-chain modifications, amino acid substitution, etc. which improve their functionality. This has led to a substantial interest of researchers and pharmaceutical companies, moving the next generation of these therapeutics from fundamental research to the market. Various chemical and computational approaches are aiding the production of more stable and long-lasting peptides guiding the formulation of novel and advanced therapeutic agents. However, there is not a single article that talks about various peptide design approaches i.e., in-silico and in-vitro along with their applications and strategies to improve their efficacy. In this review, we try to bring different aspects of peptide-based therapeutics under one article with a clear focus to cover the missing links in the literature. This review draws emphasis on various in-silico approaches and modification-based peptide design strategies. It also highlights the recent progress made in peptide delivery methods important for their enhanced clinical efficacy. The article would provide a bird's-eye view to researchers aiming to develop peptides with therapeutic applications. Graphical Abstract
Collapse
Affiliation(s)
- Panchali Barman
- Institute of Forensic Science and Criminology (UIEAST), Panjab University, Sector 14, Chandigarh, 160014 India
| | - Shubhi Joshi
- Energy Research Centre, Panjab University, Sector 14, Chandigarh, 160014 India
| | - Sheetal Sharma
- Department of Biophysics, Panjab University, Sector 25, Chandigarh, U.T 160014 India
| | - Simran Preet
- Department of Biophysics, Panjab University, Sector 25, Chandigarh, U.T 160014 India
| | - Shweta Sharma
- Institute of Forensic Science and Criminology (UIEAST), Panjab University, Sector 14, Chandigarh, 160014 India
| | - Avneet Saini
- Department of Biophysics, Panjab University, Sector 25, Chandigarh, U.T 160014 India
| |
Collapse
|
32
|
Kielholz T, Rohde F, Jung N, Windbergs M. Bacteriophage-loaded functional nanofibers for treatment of P. aeruginosa and S. aureus wound infections. Sci Rep 2023; 13:8330. [PMID: 37221194 DOI: 10.1038/s41598-023-35364-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/17/2023] [Indexed: 05/25/2023] Open
Abstract
The increasing incidence of infected skin wounds poses a major challenge in clinical practice, especially when conventional antibiotic therapy fails. In this context, bacteriophages emerged as promising alternatives for the treatment of antibiotic-resistant bacteria. However, clinical implementation remains hampered by the lack of efficient delivery approaches to infected wound tissue. In this study, bacteriophage-loaded electrospun fiber mats were successfully developed as next-generation wound dressings for the treatment of infected wounds. We employed a coaxial electrospinning approach, creating fibers with a protective polymer shell, enveloping bacteriophages in the core while maintaining their antimicrobial activity. The novel fibers exhibited a reproducible fiber diameter range and morphology, while the mechanical fiber properties were ideal for application onto wounds. Further, immediate release kinetics for the phages were confirmed as well as the biocompatibility of the fibers with human skin cells. Antimicrobial activity was demonstrated against Staphylococcus aureus and Pseudomonas aeruginosa and the core/shell formulation maintained the bacteriophage activity for 4 weeks when stored at - 20 °C. Based on these promising characteristics, our approach holds great potential as a platform technology for the encapsulation of bioactive bacteriophages to enable the translation of phage therapy into clinical application.
Collapse
Affiliation(s)
- Tobias Kielholz
- Institute of Pharmaceutical Technology and Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Max-Von-Laue-Str. 9, 60438, Frankfurt am Main, Germany
| | - Felix Rohde
- Institute of Pharmaceutical Technology and Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Max-Von-Laue-Str. 9, 60438, Frankfurt am Main, Germany
| | - Nathalie Jung
- Institute of Pharmaceutical Technology and Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Max-Von-Laue-Str. 9, 60438, Frankfurt am Main, Germany
| | - Maike Windbergs
- Institute of Pharmaceutical Technology and Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Max-Von-Laue-Str. 9, 60438, Frankfurt am Main, Germany.
| |
Collapse
|
33
|
Copling A, Akantibila M, Kumaresan R, Fleischer G, Cortes D, Tripathi RS, Carabetta VJ, Vega SL. Recent Advances in Antimicrobial Peptide Hydrogels. Int J Mol Sci 2023; 24:7563. [PMID: 37108725 PMCID: PMC10139150 DOI: 10.3390/ijms24087563] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 04/17/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
Advances in the number and type of available biomaterials have improved medical devices such as catheters, stents, pacemakers, prosthetic joints, and orthopedic devices. The introduction of a foreign material into the body comes with a risk of microbial colonization and subsequent infection. Infections of surgically implanted devices often lead to device failure, which leads to increased patient morbidity and mortality. The overuse and improper use of antimicrobials has led to an alarming rise and spread of drug-resistant infections. To overcome the problem of drug-resistant infections, novel antimicrobial biomaterials are increasingly being researched and developed. Hydrogels are a class of 3D biomaterials consisting of a hydrated polymer network with tunable functionality. As hydrogels are customizable, many different antimicrobial agents, such as inorganic molecules, metals, and antibiotics have been incorporated or tethered to them. Due to the increased prevalence of antibiotic resistance, antimicrobial peptides (AMPs) are being increasingly explored as alternative agents. AMP-tethered hydrogels are being increasingly examined for antimicrobial properties and practical applications, such as wound-healing. Here, we provide a recent update, from the last 5 years of innovations and discoveries made in the development of photopolymerizable, self-assembling, and AMP-releasing hydrogels.
Collapse
Affiliation(s)
- Aryanna Copling
- Department of Molecular & Cellular Biosciences, Rowan University, Glassboro, NJ 08028, USA;
| | - Maxwell Akantibila
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA; (M.A.); (G.F.); (D.C.); (R.S.T.)
| | - Raaha Kumaresan
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA;
| | - Gilbert Fleischer
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA; (M.A.); (G.F.); (D.C.); (R.S.T.)
| | - Dennise Cortes
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA; (M.A.); (G.F.); (D.C.); (R.S.T.)
| | - Rahul S. Tripathi
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA; (M.A.); (G.F.); (D.C.); (R.S.T.)
| | - Valerie J. Carabetta
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ 08103, USA; (M.A.); (G.F.); (D.C.); (R.S.T.)
| | - Sebastián L. Vega
- Department of Biomedical Engineering, Rowan University, Glassboro, NJ 08028, USA;
- Department of Orthopedic Surgery, Cooper Medical School of Rowan University, Camden, NJ 08103, USA
| |
Collapse
|
34
|
Teimouri H, Medvedeva A, Kolomeisky AB. Bacteria-Specific Feature Selection for Enhanced Antimicrobial Peptide Activity Predictions Using Machine-Learning Methods. J Chem Inf Model 2023; 63:1723-1733. [PMID: 36912047 DOI: 10.1021/acs.jcim.2c01551] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2023]
Abstract
There are several classes of short peptide molecules, known as antimicrobial peptides (AMPs), which are produced during the immune responses of living organisms against various infections. In recent years, substantial progress has been achieved in applying machine-learning methods to predict the activities of AMPs against bacteria. In most investigated cases, however, the outcome is not bacterium-specific since the specific features of bacteria, such as chemical composition and structure of membranes, are not considered. To overcome this problem, we developed a new computational approach that allowed us to train several supervised machine-learning models using a specific set of data associated with peptides targeting E. coli bacteria. LASSO regression and Support Vector Machine techniques have been utilized to select, among more than 1500 physicochemical descriptors, the most important features that can be used to classify a peptide as antimicrobial or ineffective against E. coli. We then performed the classification of active versus inactive AMPs using the Support Vector classifiers, Logistic Regression, and Random Forest methods. This computational study allows us to make recommendations of how to design more efficient antibacterial drug therapies.
Collapse
Affiliation(s)
- Hamid Teimouri
- Department of Chemistry, Rice University, Houston, Texas 77005, United States.,Center for Theoretical Biological Physics, Rice University, Houston, Texas 77005, United States
| | - Angela Medvedeva
- Department of Chemistry, Rice University, Houston, Texas 77005, United States.,Center for Theoretical Biological Physics, Rice University, Houston, Texas 77005, United States
| | - Anatoly B Kolomeisky
- Department of Chemistry, Rice University, Houston, Texas 77005, United States.,Center for Theoretical Biological Physics, Rice University, Houston, Texas 77005, United States.,Department of Chemical and Biomolecular Engineering, Rice University, Houston, Texas 77005, United States.,Department of Physics and Astronomy, Rice University, Houston, Texas 77005, United States
| |
Collapse
|
35
|
Wiman E, Zattarin E, Aili D, Bengtsson T, Selegård R, Khalaf H. Development of novel broad-spectrum antimicrobial lipopeptides derived from plantaricin NC8 β. Sci Rep 2023; 13:4104. [PMID: 36914718 PMCID: PMC10011573 DOI: 10.1038/s41598-023-31185-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 03/07/2023] [Indexed: 03/16/2023] Open
Abstract
Bacterial resistance towards antibiotics is a major global health issue. Very few novel antimicrobial agents and therapies have been made available for clinical use during the past decades, despite an increasing need. Antimicrobial peptides have been intensely studied, many of which have shown great promise in vitro. We have previously demonstrated that the bacteriocin Plantaricin NC8 αβ (PLNC8 αβ) from Lactobacillus plantarum effectively inhibits Staphylococcus spp., and shows little to no cytotoxicity towards human keratinocytes. However, due to its limitations in inhibiting gram-negative species, the aim of the present study was to identify novel antimicrobial peptidomimetic compounds with an enhanced spectrum of activity, derived from the β peptide of PLNC8 αβ. We have rationally designed and synthesized a small library of lipopeptides with significantly improved antimicrobial activity towards both gram-positive and gram-negative bacteria, including the ESKAPE pathogens. The lipopeptides consist of 16 amino acids with a terminal fatty acid chain and assemble into micelles that effectively inhibit and kill bacteria by permeabilizing their cell membranes. They demonstrate low hemolytic activity and liposome model systems further confirm selectivity for bacterial lipid membranes. The combination of lipopeptides with different antibiotics enhanced the effects in a synergistic or additive manner. Our data suggest that the novel lipopeptides are promising as future antimicrobial agents, however additional experiments using relevant animal models are necessary to further validate their in vivo efficacy.
Collapse
Affiliation(s)
- Emanuel Wiman
- School of Medical Sciences, Faculty of Medicine and Health, Department of Microbiology, Immunology and Reproductive Science, Örebro University, Örebro, Sweden
| | - Elisa Zattarin
- Laboratory of Molecular Materials, Division of Biophysics and Bioengineering, Department of Physics, Chemistry and Biology, Linköping University, 581 83, Linköping, Sweden
| | - Daniel Aili
- Laboratory of Molecular Materials, Division of Biophysics and Bioengineering, Department of Physics, Chemistry and Biology, Linköping University, 581 83, Linköping, Sweden
| | - Torbjörn Bengtsson
- School of Medical Sciences, Faculty of Medicine and Health, Department of Microbiology, Immunology and Reproductive Science, Örebro University, Örebro, Sweden
| | - Robert Selegård
- Laboratory of Molecular Materials, Division of Biophysics and Bioengineering, Department of Physics, Chemistry and Biology, Linköping University, 581 83, Linköping, Sweden.
| | - Hazem Khalaf
- School of Medical Sciences, Faculty of Medicine and Health, Department of Microbiology, Immunology and Reproductive Science, Örebro University, Örebro, Sweden.
| |
Collapse
|
36
|
Yang S, Duncan G. Synthetic Mucus Biomaterials for Antimicrobial Peptide Delivery. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.07.531025. [PMID: 36945438 PMCID: PMC10028879 DOI: 10.1101/2023.03.07.531025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
Despite the promise of antimicrobial peptides (AMPs) as treatments for antibiotic-resistant infections, their therapeutic efficacy is limited due to the rapid degradation and low bioavailability of AMPs. To address this, we have developed and characterized a synthetic mucus (SM) biomaterial capable of delivering AMPs and enhancing their therapeutic effect. LL37 loaded SM hydrogels demonstrated controlled release of LL37 over 8 hours as a result of charge-mediated interactions between mucins and LL37 AMPs. Compared to treatment with LL37 alone where antimicrobial activity was reduced after 3 hours, LL37-SM hydrogels inhibited Pseudomonas aeruginosa PAO1 growth over 12 hours. LL37-SM hydrogel treatment reduced PAO1 viability over 6 hours whereas a rebound in bacterial growth was observed when treated with LL37 only. These data demonstrate LL37-SM hydrogels enhance antimicrobial activity by preserving LL37 AMP activity and bioavailability. Overall, this work establishes SM biomaterials as a platform for enhanced AMP delivery for antimicrobial applications.
Collapse
Affiliation(s)
- Sydney Yang
- University of Maryland, Fischell Department of Bioengineering, College Park, MD
| | - Gregg Duncan
- University of Maryland, Fischell Department of Bioengineering, College Park, MD
| |
Collapse
|
37
|
Sun Y, Chan J, Bose K, Tam C. Simultaneous control of infection and inflammation with keratin-derived antibacterial peptides targeting TLRs and co-receptors. Sci Transl Med 2023; 15:eade2909. [PMID: 36888696 PMCID: PMC10173409 DOI: 10.1126/scitranslmed.ade2909] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 02/10/2023] [Indexed: 03/10/2023]
Abstract
Controlling infection-driven inflammation is a major clinical dilemma because of limited therapeutic options and possible adverse effects on microbial clearance. Compounding this difficulty is the continued emergence of drug-resistant bacteria, where experimental strategies aiming to augment inflammatory responses for enhanced microbial killing are not applicable treatment options for infections of vulnerable organs. As with corneal infections, severe or prolonged inflammation jeopardizes corneal transparency, leading to devastating vision loss. We hypothesized that keratin 6a-derived antimicrobial peptides (KAMPs) may be a two-pronged remedy capable of tackling bacterial infection and inflammation at once. We used murine peritoneal neutrophils and macrophages, together with an in vivo model of sterile corneal inflammation, to find that nontoxic and prohealing KAMPs with natural 10- and 18-amino acid sequences suppressed lipoteichoic acid (LTA)- and lipopolysaccharide (LPS)-induced NFκB and IRF3 activation, proinflammatory cytokine production, and phagocyte recruitment independently of their bactericidal function. Mechanistically, KAMPs not only competed with bacterial ligands for cell surface Toll-like receptor (TLR) and co-receptors (MD2, CD14, and TLR2) but also reduced cell surface availability of TLR2 and TLR4 through promotion of receptor endocytosis. Topical KAMP treatment effectively alleviated experimental bacterial keratitis, as evidenced by substantial reductions of corneal opacification, inflammatory cell infiltration, and bacterial burden. These findings reveal the TLR-targeting activities of KAMPs and demonstrate their therapeutic potential as a multifunctional drug for managing infectious inflammatory disease.
Collapse
Affiliation(s)
- Yan Sun
- Department of Ophthalmic Research, Cole Eye Institute and Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jonathan Chan
- Department of Ophthalmic Research, Cole Eye Institute and Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
| | - Karthikeyan Bose
- Department of Ophthalmic Research, Cole Eye Institute and Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Connie Tam
- Department of Ophthalmic Research, Cole Eye Institute and Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
| |
Collapse
|
38
|
Adélaïde M, Salnikov E, Ramos-Martín F, Aisenbrey C, Sarazin C, Bechinger B, D’Amelio N. The Mechanism of Action of SAAP-148 Antimicrobial Peptide as Studied with NMR and Molecular Dynamics Simulations. Pharmaceutics 2023; 15:pharmaceutics15030761. [PMID: 36986623 PMCID: PMC10051583 DOI: 10.3390/pharmaceutics15030761] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 02/18/2023] [Accepted: 02/21/2023] [Indexed: 03/02/2023] Open
Abstract
Background: SAAP-148 is an antimicrobial peptide derived from LL-37. It exhibits excellent activity against drug-resistant bacteria and biofilms while resisting degradation in physiological conditions. Despite its optimal pharmacological properties, its mechanism of action at the molecular level has not been explored. Methods: The structural properties of SAAP-148 and its interaction with phospholipid membranes mimicking mammalian and bacterial cells were studied using liquid and solid-state NMR spectroscopy as well as molecular dynamics simulations. Results: SAAP-148 is partially structured in solution and stabilizes its helical conformation when interacting with DPC micelles. The orientation of the helix within the micelles was defined by paramagnetic relaxation enhancements and found similar to that obtained using solid-state NMR, where the tilt and pitch angles were determined based on 15N chemical shift in oriented models of bacterial membranes (POPE/POPG). Molecular dynamic simulations revealed that SAAP-148 approaches the bacterial membrane by forming salt bridges between lysine and arginine residues and lipid phosphate groups while interacting minimally with mammalian models containing POPC and cholesterol. Conclusions: SAAP-148 stabilizes its helical fold onto bacterial-like membranes, placing its helix axis almost perpendicular to the surface normal, thus probably acting by a carpet-like mechanism on the bacterial membrane rather than forming well-defined pores.
Collapse
Affiliation(s)
- Morgane Adélaïde
- Unité de Génie Enzymatique et Cellulaire UMR 7025 CNRS, Université de Picardie Jules Verne, 80039 Amiens, France
| | - Evgeniy Salnikov
- Institut de Chimie, UMR7177, Université de Strasbourg/CNRS, 67000 Strasbourg, France
| | - Francisco Ramos-Martín
- Unité de Génie Enzymatique et Cellulaire UMR 7025 CNRS, Université de Picardie Jules Verne, 80039 Amiens, France
- Correspondence: (F.R.-M.); (N.D.); Tel.: +33-3-22-82-74-73 (F.R.-M. & N.D.)
| | - Christopher Aisenbrey
- Institut de Chimie, UMR7177, Université de Strasbourg/CNRS, 67000 Strasbourg, France
| | - Catherine Sarazin
- Unité de Génie Enzymatique et Cellulaire UMR 7025 CNRS, Université de Picardie Jules Verne, 80039 Amiens, France
| | - Burkhard Bechinger
- Institut de Chimie, UMR7177, Université de Strasbourg/CNRS, 67000 Strasbourg, France
| | - Nicola D’Amelio
- Unité de Génie Enzymatique et Cellulaire UMR 7025 CNRS, Université de Picardie Jules Verne, 80039 Amiens, France
- Correspondence: (F.R.-M.); (N.D.); Tel.: +33-3-22-82-74-73 (F.R.-M. & N.D.)
| |
Collapse
|
39
|
Hussein M, Jasim R, Gocol H, Baker M, Thombare VJ, Ziogas J, Purohit A, Rao GG, Li J, Velkov T. Comparative Proteomics of Outer Membrane Vesicles from Polymyxin-Susceptible and Extremely Drug-Resistant Klebsiella pneumoniae. mSphere 2023; 8:e0053722. [PMID: 36622250 PMCID: PMC9942579 DOI: 10.1128/msphere.00537-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 12/06/2022] [Indexed: 01/10/2023] Open
Abstract
Outer membrane vesicles (OMVs) secreted by Gram-negative bacteria serve as transporters for the delivery of cargo such as virulence and antibiotic resistance factors. OMVs play a key role in the defense against membrane-targeting antibiotics such as the polymyxin B. Herein, we conducted comparative proteomics of OMVs from paired Klebsiella pneumoniae ATCC 700721 polymyxin-susceptible (polymyxin B MIC = 0.5 mg/L) and an extremely resistant (polymyxin B MIC ≥128 mg/L), following exposure to 2 mg/L of polymyxin B. Comparative profiling of the OMV subproteome of each strain revealed proteins from multiple perturbed pathways, particularly in the polymyxin-susceptible strain, including outer membrane assembly (lipopolysaccharide, O-antigen, and peptidoglycan biosynthesis), cationic antimicrobial peptide resistance, β-lactam resistance, and quorum sensing. In the polymyxin-susceptible strain, polymyxin B treatment reduced the expression of OMV proteins in the pathways related to adhesion, virulence, and the cell envelope stress responses, whereas, in the polymyxin-resistant strain, the proteins involved in LPS biosynthesis, RNA degradation, and nucleotide excision repair were significantly overexpressed in response to polymyxin B treatment. Intriguingly, the key polymyxin resistance enzymes 4-amino-4-deoxy-l-arabinose transferase and the PhoPQ two-component protein kinase were significantly downregulated in the OMVs of the polymyxin-susceptible strain. Additionally, a significant reduction in class A β-lactamase proteins was observed following polymyxin B treatment in the OMVs of both strains, particularly the OMVs of the polymyxin-susceptible strain. These findings shed new light on the OMV subproteome of extremely polymyxin resistant K. pneumoniae, which putatively may serve as active decoys to make the outer membrane more impervious to polymyxin attack. IMPORTANCE OMVs can help bacteria to fight antibiotics not only by spreading antibiotic resistance genes but also by acting as protective armor against antibiotics. By employing proteomics, we found that OMVs have a potential role in shielding K. pneumoniae and acting as decoys to polymyxin attack, through declining the export of proteins (e.g., 4-amino-4-deoxy-l-arabinose transferase) involved in polymyxin resistance. Furthermore, polymyxin B treatment of both strains leads to shedding of the OMVs with perturbed proteins involved in outer membrane remodeling (e.g., LPS biosynthesis) as well as pathogenic potential of K. pneumoniae (e.g., quorum sensing). The problematic extended spectrum beta-lactamases SHV and TEM were significantly reduced in both strains, suggesting that polymyxin B may act as a potentiator to sensitize the bacterium to β-lactam antibiotics. This study highlights the importance of OMVs as "molecular mules" for the intercellular transmission and delivery of resistance and cellular repair factors in the bacterial response to polymyxins.
Collapse
Affiliation(s)
- Maytham Hussein
- Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, Victoria, Australia
| | - Raad Jasim
- Department of Pharmacology, College of Pharmacy, University of Babylon, Iraq
| | - Hakan Gocol
- Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Mark Baker
- Discipline of Biological Sciences, Priority Research Centre in Reproductive Biology, Faculty of Science and IT, University of Newcastle, Callaghan, New South Wales, Australia
| | - Varsha J. Thombare
- Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, Victoria, Australia
| | - James Ziogas
- Department of Biochemistry and Pharmacology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, Victoria, Australia
| | - Aayush Purohit
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Gauri G. Rao
- Division of Pharmacotherapy and Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Jian Li
- Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, Victoria, Australia
| | - Tony Velkov
- Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
40
|
Li T, Wang Z, Guo J, de la Fuente-Nunez C, Wang J, Han B, Tao H, Liu J, Wang X. Bacterial resistance to antibacterial agents: Mechanisms, control strategies, and implications for global health. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 860:160461. [PMID: 36435256 DOI: 10.1016/j.scitotenv.2022.160461] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 11/19/2022] [Accepted: 11/20/2022] [Indexed: 06/16/2023]
Abstract
The spread of bacterial drug resistance has posed a severe threat to public health globally. Here, we cover bacterial resistance to current antibacterial drugs, including traditional herbal medicines, conventional antibiotics, and antimicrobial peptides. We summarize the influence of bacterial drug resistance on global health and its economic burden while highlighting the resistance mechanisms developed by bacteria. Based on the One Health concept, we propose 4A strategies to combat bacterial resistance, including prudent Application of antibacterial agents, Administration, Assays, and Alternatives to antibiotics. Finally, we identify several opportunities and unsolved questions warranting future exploration for combating bacterial resistance, such as predicting genetic bacterial resistance through the use of more effective techniques, surveying both genetic determinants of bacterial resistance and the transmission dynamics of antibiotic resistance genes (ARGs).
Collapse
Affiliation(s)
- Ting Li
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, PR China; Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, PR China; State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Biotechnology, No. 20, Dongda Street, Fengtai District, Beijing 100071, PR China
| | - Zhenlong Wang
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, PR China; Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, PR China
| | - Jianhua Guo
- Australian Centre for Water and Environmental Biotechnology (ACWEB, formerly AWMC), The University of Queensland, St Lucia, Queensland 4072, Australia.
| | - Cesar de la Fuente-Nunez
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America; Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States of America; Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA, United States of America.
| | - Jinquan Wang
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, PR China; Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, PR China
| | - Bing Han
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, PR China; Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, PR China
| | - Hui Tao
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, PR China; Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, PR China
| | - Jie Liu
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, PR China; Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, PR China
| | - Xiumin Wang
- Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, PR China; Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing 100081, PR China.
| |
Collapse
|
41
|
Rybczyńska-Tkaczyk K, Grenda A, Jakubczyk A, Kiersnowska K, Bik-Małodzińska M. Natural Compounds with Antimicrobial Properties in Cosmetics. Pathogens 2023; 12:320. [PMID: 36839592 PMCID: PMC9959536 DOI: 10.3390/pathogens12020320] [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: 12/15/2022] [Revised: 02/08/2023] [Accepted: 02/10/2023] [Indexed: 02/17/2023] Open
Abstract
Currently, the cosmetic industry is a very intensively growing part of the economy. Consumer demands are adapted to the current lifestyle, which is based on technological innovations and awareness of the impact of various factors on human health and fitness. There is growing interest in cosmetics based on environmentally friendly natural compounds exerting health-promoting effects. Chemicals with antimicrobial properties used as ingredients in cosmetics ensure their durability and safety. Polyphenolic compounds, peptides, essential oils, and plant extracts characterized by these properties are natural ingredients that can replace synthetic components of cosmetics. The advantage of these compounds is that they exhibit antioxidant, anti-inflammatory, and soothing properties, enhancing the product value in addition to their antimicrobial properties. This review article describes the antimicrobial properties of natural compounds that can protect cosmetics and can replace previously used preservative agents. Various studies indicate that the use of these compounds increases consumer interest in these products and has a positive impact on the environment.
Collapse
Affiliation(s)
- Kamila Rybczyńska-Tkaczyk
- Department of Environmental Microbiology, University of Life Sciences in Lublin, St. Leszczyńskiego 7, 20-069 Lublin, Poland
| | - Anna Grenda
- Department of Pneumonology, Oncology and Allergology, Medical University in Lublin, ul. Jaczewskiego 8, 20-090 Lublin, Poland
| | - Anna Jakubczyk
- Department of Biochemistry and Food Chemistry, University of Life Sciences in Lublin, Skromna 8, 20-704 Lublin, Poland
| | - Kaja Kiersnowska
- Department of Biochemistry and Food Chemistry, University of Life Sciences in Lublin, Skromna 8, 20-704 Lublin, Poland
| | - Marta Bik-Małodzińska
- Institute of Soil Science, Engineering and Environmental Management, University of Life Sciences in Lublin, ul. Leszczyńskiego 7, 20-069 Lublin, Poland
| |
Collapse
|
42
|
Caspari OD, Garrido C, Law CO, Choquet Y, Wollman FA, Lafontaine I. Converting antimicrobial into targeting peptides reveals key features governing protein import into mitochondria and chloroplasts. PLANT COMMUNICATIONS 2023:100555. [PMID: 36733255 PMCID: PMC10363480 DOI: 10.1016/j.xplc.2023.100555] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/18/2023] [Accepted: 01/26/2023] [Indexed: 06/18/2023]
Abstract
We asked what peptide features govern targeting to the mitochondria versus the chloroplast, using antimicrobial peptides as a starting point. This approach was inspired by the endosymbiotic hypothesis that organelle-targeting peptides derive from antimicrobial amphipathic peptides delivered by the host cell, to which organelle progenitors became resistant. To explore the molecular changes required to convert antimicrobial into targeting peptides, we expressed a set of 13 antimicrobial peptides in Chlamydomonas reinhardtii. Peptides were systematically modified to test distinctive features of mitochondrion- and chloroplast-targeting peptides, and we assessed their targeting potential by following the intracellular localization and maturation of a Venus fluorescent reporter used as a cargo protein. Mitochondrial targeting can be achieved by some unmodified antimicrobial peptide sequences. Targeting to both organelles is improved by replacing lysines with arginines. Chloroplast targeting is enabled by the presence of flanking unstructured sequences, additional constraints consistent with chloroplast endosymbiosis having occurred in a cell that already contained mitochondria. If indeed targeting peptides evolved from antimicrobial peptides, then required modifications imply a temporal evolutionary scenario with an early exchange of cationic residues and a late acquisition of chloroplast-specific motifs.
Collapse
Affiliation(s)
- Oliver D Caspari
- UMR7141 (CNRS/Sorbonne Université), Institut de Biologie Physico-Chimique, 13 Rue Pierre et Marie Curie, 75005 Paris, France.
| | - Clotilde Garrido
- UMR7141 (CNRS/Sorbonne Université), Institut de Biologie Physico-Chimique, 13 Rue Pierre et Marie Curie, 75005 Paris, France
| | - Chris O Law
- Centre for Microscopy and Cellular Imaging, Biology Department Loyola Campus of Concordia University, 7141 Sherbrooke W., Montréal, QC H4B 1R6, Canada
| | - Yves Choquet
- UMR7141 (CNRS/Sorbonne Université), Institut de Biologie Physico-Chimique, 13 Rue Pierre et Marie Curie, 75005 Paris, France
| | - Francis-André Wollman
- UMR7141 (CNRS/Sorbonne Université), Institut de Biologie Physico-Chimique, 13 Rue Pierre et Marie Curie, 75005 Paris, France
| | - Ingrid Lafontaine
- UMR7141 (CNRS/Sorbonne Université), Institut de Biologie Physico-Chimique, 13 Rue Pierre et Marie Curie, 75005 Paris, France.
| |
Collapse
|
43
|
Aloke C, Achilonu I. Coping with the ESKAPE pathogens: Evolving strategies, challenges and future prospects. Microb Pathog 2023; 175:105963. [PMID: 36584930 DOI: 10.1016/j.micpath.2022.105963] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/22/2022] [Accepted: 12/26/2022] [Indexed: 12/29/2022]
Abstract
Globally, the ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species) are the major cause of nosocomial infections. These pathogens are multidrug resistant, and their negative impacts have brought serious health challenges and economic burden on many countries worldwide. Thus, this narrative review exploits different emerging alternative therapeutic strategies including combination antibiotics, antimicrobial peptides ((AMPs), bacteriophage and photodynamic therapies used in the treatment of the ESKAPE pathogens, their merits, limitations, and future prospects. Our findings indicate that ESKAPE pathogens exhibit resistance to drug using different mechanisms including drug inactivation by irreversible enzyme cleavage, drug-binding site alteration, diminution in permeability of drug or drug efflux increment to reduce accumulation of drug as well as biofilms production. However, the scientific community has shown significant interest in using these novel strategies with numerous benefits although they have some limitations including but not limited to instability and toxicity of the therapeutic agents, or the host developing immune response against the therapeutic agents. Thus, comprehension of resistance mechanisms of these pathogens is necessary to further develop or modify these approaches in order to overcome these health challenges including the barriers of bacterial resistance.
Collapse
Affiliation(s)
- Chinyere Aloke
- Protein Structure-Function and Research Unit, School of Molecular and Cell Biology, Faculty of Science, University of the Witwatersrand, Braamfontein, Johannesburg, 2050, South Africa; Department of Medical Biochemistry, Alex Ekwueme Federal University Ndufu-Alike, Ebonyi State, Nigeria.
| | - Ikechukwu Achilonu
- Protein Structure-Function and Research Unit, School of Molecular and Cell Biology, Faculty of Science, University of the Witwatersrand, Braamfontein, Johannesburg, 2050, South Africa
| |
Collapse
|
44
|
Grizanova EV, Krytsyna TI, Kalmykova GV, Sokolova E, Alikina T, Kabilov M, Coates CJ, Dubovskiy IM. Virulent and necrotrophic strategies of Bacillus thuringiensis in susceptible and resistant insects, Galleria mellonella. Microb Pathog 2023; 175:105958. [PMID: 36572197 DOI: 10.1016/j.micpath.2022.105958] [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: 10/11/2022] [Revised: 12/03/2022] [Accepted: 12/21/2022] [Indexed: 12/25/2022]
Abstract
Bacillus thuringiensis (Bt) is one of the most common entomopathogenic bacteria used as a biopesticide, and source of endotoxin genes for generating insect-resistant transgenic plants. The mechanisms underpinning an insect's susceptibility or resistance to B. thuringiensis are diverse. The bacterial lifecycle does not end with the death of a host, they continue to exploit the cadaver to reproduce and sporulate. Herein, we studied the progression of B. thuringiensis subsp. galleriae infection in two populations of wax moth larvae (Galleria mellonella) to gain further insight into the "arms race" between B. thuringiensis virulence and insect defences. Two doses of B. thuringiensis subsp. galleriae (spore and crystalline toxin mixtures) were administered orally to compare the responses of susceptible (S) and resistant (R) populations at ∼30% mortality each. To investigate B. thuringiensis-insect antibiosis, we used a combination of in vivo infection trials, bacterial microbiome analysis, and RNAi targeting the antibacterial peptide gloverin. Within 48 h post-inoculation, B. thuringiensis-resistant insects purged the midgut of bacteria, i.e., colony forming unit numbers fell below detectable levels. Second, B. thuringiensis rapidly modulated gene expression to initiate sporulation (linked to quorum sensing) when exposed to resistant insects in contrast to susceptible G. mellonella. We reinforce earlier findings that elevated levels of antimicrobial peptides, specifically gloverin, are found in the midgut of resistant insects, which is an evolutionary strategy to combat B. thuringiensis infection via its main portal of entry. A sub-population of highly virulent B. thuringiensis can survive the enhanced immune defences of resistant G. mellonella by disrupting the midgut microbiome and switching rapidly to a necrotrophic strategy, prior to sporulation in the cadaver.
Collapse
Affiliation(s)
- Ekaterina V Grizanova
- Department of Plant Protection, Novosibirsk State Agrarian University, 630039, Novosibirsk, Russia.
| | - Tatiana I Krytsyna
- Department of Plant Protection, Novosibirsk State Agrarian University, 630039, Novosibirsk, Russia.
| | - Galina V Kalmykova
- Faculty of Physical Engineering, Novosibirsk State Technical University, 630039, Novosibirsk, Russia.
| | - Elina Sokolova
- Department of Plant Protection, Novosibirsk State Agrarian University, 630039, Novosibirsk, Russia.
| | - Tatyana Alikina
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630039, Novosibirsk, Russia.
| | - Marsel Kabilov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, 630039, Novosibirsk, Russia.
| | - Christopher J Coates
- Zoology, Ryan Institute, School of Natural Sciences, University of Galway, Galway, H91 TK33, Ireland; Department of Biosciences, Faculty of Science and Engineering, Swansea University, Swansea, SA2 8PP, Wales, UK.
| | - Ivan M Dubovskiy
- Department of Plant Protection, Novosibirsk State Agrarian University, 630039, Novosibirsk, Russia.
| |
Collapse
|
45
|
The broad antibacterial activity of a small synthetic receptor for cellular phosphatidylglycerol lipids. Folia Microbiol (Praha) 2023; 68:465-476. [PMID: 36622376 DOI: 10.1007/s12223-022-01023-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 12/06/2022] [Indexed: 01/10/2023]
Abstract
A small receptor molecule composed of a porphyrin core with tetrakis-ammonium glycine pickets (liptin 3e) appears to target anionic phosphatidylglycerol (PG) lipid head groups through multifunctional binding-pocket complementarity. Although a major component of bacterial cell membranes, PG is not widely found in animal cells, making PG potentially selective for bacterial targeting. Growth of microbial isolates was monitored in liquid cultures treated with liptin 3e by dilution plate counts and turbidity. Inhibition of growth by liptin 3e was observed for the ESKAPE human pathogens (Enterobacter aerogenes, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterococcus faecium), Escherichia coli, Mycobacterium smegmatis, Streptococcus sobrinus, and methicillin-resistant S. aureus (MRSA), with certain species suppressed at <1 μg/mL (sub-μM) concentrations. Prolonged lag phases were observed, although cell viability was mainly unaffected, suggesting that liptin treatment caused bacteriostasis. Cultures treated with liptin 3e eventually recovered, resumed growth, and reached the same final densities as untreated cultures. Growth of the fungus Candida albicans was not appreciably inhibited by liptin 3e. If liptins exhibit bacteriostasis through broad extracellular binding to PG head groups, thereby disrupting cellular processes, liptins may be considered for development into preclinical drug candidates or be useful as a targeting system for molecular beacons or antibacterial drugs.
Collapse
|
46
|
Lee SM, Keum HL, Sul WJ. Bacterial Crosstalk via Antimicrobial Peptides on the Human Skin: Therapeutics from a Sustainable Perspective. J Microbiol 2023; 61:1-11. [PMID: 36719618 DOI: 10.1007/s12275-022-00002-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 10/18/2022] [Accepted: 10/24/2022] [Indexed: 02/01/2023]
Abstract
The skin's epidermis is an essential barrier as the first guard against invading pathogens, and physical protector from external injury. The skin microbiome, which consists of numerous bacteria, fungi, viruses, and archaea on the epidermis, play a key role in skin homeostasis. Antibiotics are a fast-acting and effective treatment method, however, antibiotic use is a nuisance that can disrupt skin homeostasis by eradicating beneficial bacteria along with the intended pathogens and cause antibiotic-resistant bacteria spread. Increased numbers of antimicrobial peptides (AMPs) derived from humans and bacteria have been reported, and their roles have been well defined. Recently, modulation of the skin microbiome with AMPs rather than artificially synthesized antibiotics has attracted the attention of researchers as many antibiotic-resistant strains make treatment mediation difficult in the context of ecological problems. Herein, we discuss the overall insights into the skin microbiome, including its regulation by different AMPs, as well as their composition and role in health and disease.
Collapse
Affiliation(s)
- Seon Mi Lee
- Department of Systems Biotechnology, Chung-Ang University, Anseong, 17546, Republic of Korea
| | - Hye Lim Keum
- Department of Systems Biotechnology, Chung-Ang University, Anseong, 17546, Republic of Korea
| | - Woo Jun Sul
- Department of Systems Biotechnology, Chung-Ang University, Anseong, 17546, Republic of Korea.
| |
Collapse
|
47
|
Chatupheeraphat C, Peamchai J, Luk-in S, Eiamphungporn W. Synergistic effect and antibiofilm activity of the antimicrobial peptide K11 with conventional antibiotics against multidrug-resistant and extensively drug-resistant Klebsiella pneumoniae. Front Cell Infect Microbiol 2023; 13:1153868. [PMID: 37113135 PMCID: PMC10126264 DOI: 10.3389/fcimb.2023.1153868] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 03/21/2023] [Indexed: 04/29/2023] Open
Abstract
Introduction Infections caused by drug-resistant Klebsiella pneumoniae are now a serious problem for public health, associated with high morbidity and mortality due to limited treatment options. Therefore, new antibacterial agents or a combination of agents as the first line of treatment are urgently needed. K11 is a novel antimicrobial peptide (AMP) that has demonstrated in vitro antimicrobial activity against several types of bacteria. Additionally, K11 has previously shown no hemolytic activity. Herein, the antibacterial activity, the synergistic action of K11 in combination with different conventional antibiotics and the antibiofilm activity of K11 against multidrug-resistant (MDR) and extensively drug-resistant (XDR) K. pneumoniae were investigated. Meanwhile, the stability and ability to induce the bacterial resistance of K11 were also tested. Methods Fifteen clinical isolates of MDR/XDR K. pneumoniae were used in this study. The minimum inhibitory concentration (MIC) of K11 against these isolates was determined by the broth microdilution method. In vitro synergy between K11 and antibiotics was evaluated using the checkerboard methodology. The antibiofilm activity of K11 against K. pneumoniae strong biofilm producers were explored by the crystal violet staining. The stability in different environments and resistance induction of K11 were evaluated by MIC determination. Results The MIC values of K11 against MDR/XDR K. pneumoniae isolates were 8-512 μg/mL. Intriguingly, the synergistic effects were clearly observed for K11 in combination with chloramphenicol, meropenem, rifampicin, or ceftazidime, whereas no synergy was observed when K11 was combined with colistin. Besides, K11 effectively prevented biofilm formation against K. pneumoniae strong biofilm producers in a concentration-dependent manner starting at 0.25×MIC and exerted an enhancing effect when administered in combination with meropenem, chloramphenicol, or rifampicin. Additionally, K11 demonstrated high thermal and wide pH stability along with good stability in serum and physiological salts. Significantly, K. pneumoniae showed no induction of resistance even after prolonged exposure to a sub-inhibitory concentration of K11. Conclusion These findings indicate that K11 is a promising candidate with potent antibacterial and antibiofilm activities without inducing resistance and acts synergistically with conventional antibiotics against drug-resistant K. pneumoniae.
Collapse
Affiliation(s)
- Chawalit Chatupheeraphat
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Nakhon Pathom, Thailand
| | - Jiratchaya Peamchai
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Sirirat Luk-in
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
| | - Warawan Eiamphungporn
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, Thailand
- *Correspondence: Warawan Eiamphungporn,
| |
Collapse
|
48
|
Basu S, Sineva E, Nguyen L, Sikdar N, Park JW, Sinev M, Kunta M, Gupta G. Host-derived chimeric peptides clear the causative bacteria and augment host innate immunity during infection: A case study of HLB in citrus and fire blight in apple. FRONTIERS IN PLANT SCIENCE 2022; 13:929478. [PMID: 36618616 PMCID: PMC9816411 DOI: 10.3389/fpls.2022.929478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 12/07/2022] [Indexed: 06/17/2023]
Abstract
Bacterial diseases cause severe losses in the production and revenue of many fruit crops, including citrus and apple. Huanglongbing (HLB) in citrus and fire blight in apple are two deadly diseases without any cure. In this article, we introduce a novel therapy for HLB and fire blight by enhancing the innate immunity of the host plants. Specifically, we constructed in silico a library of chimeras containing two different host peptides with observed or predicted antibacterial activity. Subsequently, we performed bactericidal and toxicity tests in vitro to select a few non-toxic chimeras with high antibacterial activity. Finally, we conducted ex planta studies to show that not only do the chimeras clear the causative bacteria from citrus leaves with HLB and from apple leaves with fire blight but they also augment the host's innate immunity during infection. This platform technology can be extended to design host-derived chimeras against multiple pathogenic bacteria that cause diseases in plants and animals of agricultural importance and in humans.
Collapse
Affiliation(s)
- Supratim Basu
- New Mexico Consortium, NMC-Biolab at Santa Fe Business Incubator, Santa Fe, NM, United States
| | - Elena Sineva
- New Mexico Consortium, NMC-Biolab at Santa Fe Business Incubator, Santa Fe, NM, United States
| | - Liza Nguyen
- New Mexico Consortium, NMC-Biolab at Santa Fe Business Incubator, Santa Fe, NM, United States
| | - Narattam Sikdar
- New Mexico Consortium, NMC-Biolab at Santa Fe Business Incubator, Santa Fe, NM, United States
| | - Jong Won Park
- Texas A&M Univ.-Kingsville Citrus Center, Weslaco, TX, United States
| | - Mikhail Sinev
- New Mexico Consortium, NMC-Biolab at Santa Fe Business Incubator, Santa Fe, NM, United States
| | - Madhurababu Kunta
- Texas A&M Univ.-Kingsville Citrus Center, Weslaco, TX, United States
| | - Goutam Gupta
- New Mexico Consortium, NMC-Biolab at Santa Fe Business Incubator, Santa Fe, NM, United States
| |
Collapse
|
49
|
Zhang F, Yang P, Mao W, Zhong C, Zhang J, Chang L, Wu X, Liu H, Zhang Y, Gou S, Ni J. Short, mirror-symmetric antimicrobial peptides centered on "RRR" have broad-spectrum antibacterial activity with low drug resistance and toxicity. Acta Biomater 2022; 154:145-167. [PMID: 36241015 DOI: 10.1016/j.actbio.2022.10.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/31/2022] [Accepted: 10/03/2022] [Indexed: 12/14/2022]
Abstract
The increasingly severe bacterial resistance worldwide pushes people to discover and design potential antibacterial drugs unavoidably. In this work, a series of short, mirror-symmetric peptides were designed and successfully synthesized, centered on "RRR" and labeled with hydrophobic amino acids at both ends. Based on the structure-activity relationship analysis, LWWR (LWWRRRWWL-NH2) was screened as a desirable mirror-symmetric peptide for further study. As expected, LWWR displayed broad-spectrum antibacterial activity against the standard bacteria and antibiotic-resistant strains. Undoubtedly, the high stability of LWWR in a complex physiological environment was an essential guarantee to maximizing its antibacterial activity. Indeed, LWWR also exhibited a rapid bactericidal speed and a low tendency to develop bacterial resistance, based on the multiple actions of non-receptor-mediated membrane actions and intra-cellular mechanisms. Surprisingly, although LWWR showed similar in vivo antibacterial activity compared with Polymyxin B and Melittin, the in vivo safety of LWWR was far higher than that of them, so LWWR had better therapeutic potential. In summary, the desirable mirror-symmetric peptide LWWR was promised as a potential antibacterial agent to confront the antibiotics resistance crisis. STATEMENT OF SIGNIFICANCE: Witnessing the growing problem of antibiotic resistance, a series of short, mirror-symmetric peptides based on the symmetric center "RRR" and hydrophobic terminals were designed and synthesized in this study. Among, LWWR (LWWRRRWWL-NH2) presented broad-spectrum antibacterial activity both in vitro and in vivo due to its multiple mechanisms and good stability. Meanwhile, the low drug resistance and toxicity of LWWR also suggested its potential for clinical application. The findings of this study will provide some inspiration for the design and development of potential antibacterial agents, and contribute to the elimination of bacterial infections worldwide as soon as possible.
Collapse
Affiliation(s)
- Fangyan Zhang
- Institute of Materia Medica and Research Unit of Peptide Science, 2019RU066, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 1 Xian Nong Tan Street, Beijing 100050, P. R. China; Institute of Pharmaceutics, School of Pharmacy and Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, P. R. China
| | - Ping Yang
- Institute of Materia Medica and Research Unit of Peptide Science, 2019RU066, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 1 Xian Nong Tan Street, Beijing 100050, P. R. China; Institute of Pharmaceutics, School of Pharmacy and Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, P. R. China
| | - Wenbo Mao
- Institute of Materia Medica and Research Unit of Peptide Science, 2019RU066, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 1 Xian Nong Tan Street, Beijing 100050, P. R. China; Institute of Pharmaceutics, School of Pharmacy and Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, P. R. China
| | - Chao Zhong
- Institute of Materia Medica and Research Unit of Peptide Science, 2019RU066, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 1 Xian Nong Tan Street, Beijing 100050, P. R. China; Institute of Pharmaceutics, School of Pharmacy and Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, P. R. China
| | - Jingying Zhang
- Institute of Materia Medica and Research Unit of Peptide Science, 2019RU066, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 1 Xian Nong Tan Street, Beijing 100050, P. R. China; Institute of Pharmaceutics, School of Pharmacy and Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, P. R. China
| | - Linlin Chang
- Institute of Materia Medica and Research Unit of Peptide Science, 2019RU066, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 1 Xian Nong Tan Street, Beijing 100050, P. R. China; Institute of Pharmaceutics, School of Pharmacy and Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, P. R. China
| | - Xiaoyan Wu
- Institute of Materia Medica and Research Unit of Peptide Science, 2019RU066, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 1 Xian Nong Tan Street, Beijing 100050, P. R. China; Institute of Pharmaceutics, School of Pharmacy and Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, P. R. China
| | - Hui Liu
- Institute of Materia Medica and Research Unit of Peptide Science, 2019RU066, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 1 Xian Nong Tan Street, Beijing 100050, P. R. China; Institute of Pharmaceutics, School of Pharmacy and Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, P. R. China
| | - Yun Zhang
- Institute of Materia Medica and Research Unit of Peptide Science, 2019RU066, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 1 Xian Nong Tan Street, Beijing 100050, P. R. China; Institute of Pharmaceutics, School of Pharmacy and Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, P. R. China
| | - Sanhu Gou
- Institute of Materia Medica and Research Unit of Peptide Science, 2019RU066, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 1 Xian Nong Tan Street, Beijing 100050, P. R. China; Institute of Pharmaceutics, School of Pharmacy and Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, P. R. China
| | - Jingman Ni
- Institute of Materia Medica and Research Unit of Peptide Science, 2019RU066, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 1 Xian Nong Tan Street, Beijing 100050, P. R. China; Institute of Pharmaceutics, School of Pharmacy and Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, P. R. China.
| |
Collapse
|
50
|
Tan H, Wang J, Song Y, Liu S, Lu Z, Luo H, Tang X. Antibacterial Potential Analysis of Novel α-Helix Peptides in the Chinese Wolf Spider Lycosa sinensis. Pharmaceutics 2022; 14:pharmaceutics14112540. [PMID: 36432731 PMCID: PMC9698133 DOI: 10.3390/pharmaceutics14112540] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/12/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022] Open
Abstract
The spider Lycosa sinensis represents a burrowing wolf spider (family Lycosidae) widely distributed in the cotton region of northern China, whose venom is rich in various bioactive peptides. In previous study, we used a combination strategy of peptidomic and transcriptomic analyses to systematically screen and identify potential antimicrobial peptides (AMPs) in Lycosa sinensis venom that matched the α-helix structures. In this work, the three peptides (LS-AMP-E1, LS-AMP-F1, and LS-AMP-G1) were subjected to sequence analysis of the physicochemical properties and helical wheel projection, and then six common clinical pathogenic bacteria (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species) with multiple drug-resistance were isolated and cultured for the evaluation and analysis of antimicrobial activity of these peptides. The results showed that two peptides (LS-AMP-E1 and LS-AMP-F1) had different inhibitory activity against six clinical drug-resistant bacteria; they can effectively inhibit the formation of biofilm and have no obvious hemolytic effect. Moreover, both LS-AMP-E1 and LS-AMP-F1 exhibited varying degrees of synergistic therapeutic effects with traditional antibiotics (azithromycin, erythromycin, and doxycycline), significantly reducing the working concentration of antibiotics and AMPs. In terms of antimicrobial mechanisms, LS-AMP-E1 and LS-AMP-F1 destroyed the integrity of bacterial cell membranes in a short period of time and completely inhibited bacterial growth within 10 min of action. Meanwhile, high concentrations of Mg2+ effectively reduced the antibacterial activity of LS-AMP-E1 and LS-AMP-F1. Together, it suggested that the two peptides interact directly on bacterial cell membranes. Taken together, bioinformatic and functional analyses in the present work sheds light on the structure-function relationships of LS-AMPs, and facilitates the discovery and clinical application of novel AMPs.
Collapse
Affiliation(s)
- Huaxin Tan
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Ecological Environment and Critical Human Diseases Prevention of Hunan Province Department of Education, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Junyao Wang
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Ecological Environment and Critical Human Diseases Prevention of Hunan Province Department of Education, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Yuxin Song
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Ecological Environment and Critical Human Diseases Prevention of Hunan Province Department of Education, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Sisi Liu
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Ecological Environment and Critical Human Diseases Prevention of Hunan Province Department of Education, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Ziyan Lu
- Department of Biochemistry and Molecular Biology, The Key Laboratory of Ecological Environment and Critical Human Diseases Prevention of Hunan Province Department of Education, School of Basic Medicine, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Haodang Luo
- Hunan Key Laboratory for Conservation and Utilization of Biological Resources in the Nanyue Mountainous Region, College of Life Sciences, Hengyang Normal University, Hengyang 421002, China
- Correspondence: (H.L.); (X.T.)
| | - Xing Tang
- Department of Clinical Laboratory, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang 421001, China
- Correspondence: (H.L.); (X.T.)
| |
Collapse
|