1
|
Jiang Z, Feng J, Wang F, Wang J, Wang N, Zhang M, Hsieh CY, Hou T, Cui W, Ma L. AI-Guided Design of Antimicrobial Peptide Hydrogels for Precise Treatment of Drug-resistant Bacterial Infections. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2500043. [PMID: 40159831 DOI: 10.1002/adma.202500043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 03/04/2025] [Indexed: 04/02/2025]
Abstract
Traditional biomaterial development lacks systematicity and predictability, posing significant challenges in addressing the intricate engineering issues related to infections with drug-resistant bacteria. The unprecedented ability of artificial intelligence (AI) to manage complex systems offers a novel paradigm for materials development. However, no AI model currently guides the development of antibacterial biomaterials based on an in-depth understanding of the interplay between biomaterials and bacteria. In this study, an AI-guided design platform (AMP-hydrogel-Designer) is developed to generate antibacterial biomaterials. This platform utilizes generative design and multi-objective constrained optimization to generate a novel thiol-containing high-efficiency antimicrobial peptide (AMP), that is functionally coupled with hydrogel to form a complex network structure. Additionally, Cu-modified barium titanate (Cu-BTO) is incorporated to facilitate further complex cross-linking via Cu2+/SH coordination to produce an AI-AMP-hydrogel. In vitro, the AI-AMP-hydrogel exhibits > 99.99% bactericidal efficacy against Methicillin-resistant Staphylococcus aureus (MRSA) and Escherichia coli (E. coli). Furthermore, Cu-BTO converts mechanical stimulation into electrical signals, thereby promoting the expression of growth factors and angiogenesis. In a rat model with dynamic wounds, the AI-AMP hydrogel significantly reduces the MRSA load and markedly accelerates wound healing. Therefore, the AI-guided biomaterial development strategy offers an innovative solution to precisely treat drug-resistant bacterial infections.
Collapse
Affiliation(s)
- Zhihui Jiang
- Department of Pharmacy, General Hospital of Southern Theatre Command, Guangzhou, Guangdong, 510010, P.R. China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, P.R. China
- Graduate School, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, P.R. China
| | - Jianwen Feng
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, P.R. China
| | - Fan Wang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P.R. China
| | - Jike Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, P.R. China
| | - Ningtao Wang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P.R. China
| | - Mengmiao Zhang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, P.R. China
| | - Chang-Yu Hsieh
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, P.R. China
| | - Tingjun Hou
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, P.R. China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P.R. China
| | - Limin Ma
- Department of Orthopedics, Guang Dong Engineering Technology Research Center of Functional, Repair of Bone Defects and Biomaterials, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, P. R. China
- Department of Orthopedics, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, NO.49, Dagong Road, Ganzhou, Jiangxi, 341000, P. R. China
| |
Collapse
|
2
|
Li C, Sutherland D, Salehi A, Richter A, Lin D, Aninta SI, Ebrahimikondori H, Yanai A, Coombe L, Warren RL, Kotkoff M, Hoang LMN, Helbing CC, Birol I. Mining the UniProtKB/Swiss-Prot database for antimicrobial peptides. Protein Sci 2025; 34:e70083. [PMID: 40100125 PMCID: PMC11917140 DOI: 10.1002/pro.70083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 03/20/2025]
Abstract
The ever-growing global health threat of antibiotic resistance is compelling researchers to explore alternatives to conventional antibiotics. Antimicrobial peptides (AMPs) are emerging as a promising solution to fill this need. Naturally occurring AMPs are produced by all forms of life as part of the innate immune system. High-throughput bioinformatics tools have enabled fast and large-scale discovery of AMPs from genomic, transcriptomic, and proteomic resources of selected organisms. Public protein sequence databases, comprising over 200 million records and growing, serve as comprehensive compendia of sequences from a broad range of source organisms. Yet, large-scale in silico probing of those databases for novel AMP discovery using modern deep learning techniques has rarely been reported. In the present study, we propose an AMP mining workflow to predict novel AMPs from the UniProtKB/Swiss-Prot database using the AMP prediction tool, AMPlify, as its discovery engine. Using this workflow, we identified 8008 novel putative AMPs from all eukaryotic sequences in the database. Focusing on the practical use of AMPs as suitable antimicrobial agents with applications in the poultry industry, we prioritized 40 of those AMPs based on their similarities to known chicken AMPs in predicted structures. In our tests, 13 out of the 38 successfully synthesized peptides showed antimicrobial activity against Escherichia coli and/or Staphylococcus aureus. AMPlify and the companion scripts supporting the AMP mining workflow presented herein are publicly available at https://github.com/bcgsc/AMPlify.
Collapse
Affiliation(s)
- Chenkai Li
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
- Bioinformatics Graduate ProgramUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Darcy Sutherland
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
- Public Health LaboratoryBritish Columbia Centre for Disease ControlVancouverBritish ColumbiaCanada
- Department of Pathology and Laboratory MedicineUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Ali Salehi
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
- Public Health LaboratoryBritish Columbia Centre for Disease ControlVancouverBritish ColumbiaCanada
| | - Amelia Richter
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
- Public Health LaboratoryBritish Columbia Centre for Disease ControlVancouverBritish ColumbiaCanada
| | - Diana Lin
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
| | - Sambina Islam Aninta
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
| | - Hossein Ebrahimikondori
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
- Bioinformatics Graduate ProgramUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Anat Yanai
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
- Public Health LaboratoryBritish Columbia Centre for Disease ControlVancouverBritish ColumbiaCanada
| | - Lauren Coombe
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
| | - René L. Warren
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
| | - Monica Kotkoff
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
| | - Linda M. N. Hoang
- Public Health LaboratoryBritish Columbia Centre for Disease ControlVancouverBritish ColumbiaCanada
- Department of Pathology and Laboratory MedicineUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Caren C. Helbing
- Department of Biochemistry and MicrobiologyUniversity of VictoriaVictoriaBritish ColumbiaCanada
| | - Inanc Birol
- Canada's Michael Smith Genome Sciences CentreBC Cancer AgencyVancouverBritish ColumbiaCanada
- Public Health LaboratoryBritish Columbia Centre for Disease ControlVancouverBritish ColumbiaCanada
- Department of Pathology and Laboratory MedicineUniversity of British ColumbiaVancouverBritish ColumbiaCanada
- Department of Medical GeneticsUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| |
Collapse
|
3
|
Peng H, Kotelnikov S, Egbert ME, Ofaim S, Stevens GC, Phanse S, Saccon T, Ignatov M, Dutta S, Istace Z, Moutaoufik MT, Aoki H, Kewalramani N, Sun J, Gong Y, Padhorny D, Poda G, Alekseenko A, Porter KA, Jones G, Rodionova I, Guo H, Pogoutse O, Datta S, Saier M, Crovella M, Vajda S, Moreno-Hagelsieb G, Parkinson J, Segre D, Babu M, Kozakov D, Emili A. Ligand interaction landscape of transcription factors and essential enzymes in E. coli. Cell 2025; 188:1441-1455.e15. [PMID: 39862855 DOI: 10.1016/j.cell.2025.01.003] [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: 12/05/2022] [Revised: 09/23/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025]
Abstract
Knowledge of protein-metabolite interactions can enhance mechanistic understanding and chemical probing of biochemical processes, but the discovery of endogenous ligands remains challenging. Here, we combined rapid affinity purification with precision mass spectrometry and high-resolution molecular docking to precisely map the physical associations of 296 chemically diverse small-molecule metabolite ligands with 69 distinct essential enzymes and 45 transcription factors in the gram-negative bacterium Escherichia coli. We then conducted systematic metabolic pathway integration, pan-microbial evolutionary projections, and independent in-depth biophysical characterization experiments to define the functional significance of ligand interfaces. This effort revealed principles governing functional crosstalk on a network level, divergent patterns of binding pocket conservation, and scaffolds for designing selective chemical probes. This structurally resolved ligand interactome mapping pipeline can be scaled to illuminate the native small-molecule networks of complete cells and potentially entire multi-cellular communities.
Collapse
Affiliation(s)
- Hui Peng
- Department of Chemistry, University of Toronto, Toronto, ON M5S 3H6, Canada
| | - Sergei Kotelnikov
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Megan E Egbert
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - Shany Ofaim
- Program in Bioinformatics, Boston University, Boston, MA 02215, USA
| | - Grant C Stevens
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Sadhna Phanse
- Center for Network Systems Biology, Boston University, Boston, MA 02218, USA; Department of Biochemistry, University of Regina, Regina, SK S4S 0A2, Canada
| | - Tatiana Saccon
- Department of Biochemistry, University of Regina, Regina, SK S4S 0A2, Canada
| | - Mikhail Ignatov
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Shubham Dutta
- Department of Biochemistry, University of Regina, Regina, SK S4S 0A2, Canada
| | - Zoe Istace
- Department of Biochemistry, University of Regina, Regina, SK S4S 0A2, Canada
| | - Mohamed Taha Moutaoufik
- Department of Biochemistry, University of Regina, Regina, SK S4S 0A2, Canada; Faculty of Medical Sciences, University Mohammed VI Polytechnic, Benguerir, Morocco
| | - Hiroyuki Aoki
- Department of Biochemistry, University of Regina, Regina, SK S4S 0A2, Canada
| | - Neal Kewalramani
- Program in Bioinformatics, Boston University, Boston, MA 02215, USA; Center for Network Systems Biology, Boston University, Boston, MA 02218, USA
| | - Jianxian Sun
- Department of Chemistry, University of Toronto, Toronto, ON M5S 3H6, Canada
| | - Yufeng Gong
- Department of Chemistry, University of Toronto, Toronto, ON M5S 3H6, Canada
| | - Dzmitry Padhorny
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Gennady Poda
- Drug Discovery Program, Ontario Institute for Cancer Research, Toronto, ON M5G 0A3, Canada; Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Andrey Alekseenko
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Kathryn A Porter
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA
| | - George Jones
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Irina Rodionova
- Department of Molecular Biology, University of California, San Diego, La Jolla, San Diego, CA 920930, USA
| | - Hongbo Guo
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Oxana Pogoutse
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Suprama Datta
- Center for Network Systems Biology, Boston University, Boston, MA 02218, USA
| | - Milton Saier
- Department of Molecular Biology, University of California, San Diego, La Jolla, San Diego, CA 920930, USA
| | - Mark Crovella
- Program in Bioinformatics, Boston University, Boston, MA 02215, USA; Department of Computer Science, Boston University, Boston, MA 02215, USA
| | - Sandor Vajda
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; Department of Chemistry, Boston University, Boston, MA 02215, USA
| | | | - John Parkinson
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Daniel Segre
- Department of Biomedical Engineering, Boston University, Boston, MA 02215, USA; Program in Bioinformatics, Boston University, Boston, MA 02215, USA; Department of Biology, Boston University, Boston, MA 02215, USA
| | - Mohan Babu
- Department of Biochemistry, University of Regina, Regina, SK S4S 0A2, Canada.
| | - Dima Kozakov
- Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY 11794, USA.
| | - Andrew Emili
- Program in Bioinformatics, Boston University, Boston, MA 02215, USA; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada; Center for Network Systems Biology, Boston University, Boston, MA 02218, USA; Department of Chemistry, Boston University, Boston, MA 02215, USA; Department of Chemical Physiology and Biochemistry, Division of Oncological Sciences, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA.
| |
Collapse
|
4
|
Xu X, Jian Y, Huang L, Luo W, Wu B, Feng S, Zhou C, Zhang L. Characterization of avian β-defensin genes in Galliformes reveals widespread evolutionary diversification and distinct evolutionary relationships with infection risk. BMC Genomics 2025; 26:211. [PMID: 40033205 PMCID: PMC11874394 DOI: 10.1186/s12864-025-11390-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 02/20/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Avian β-defensins (AvBDs) represent a key family of antimicrobial host defense peptides in birds. Accumulating evidence suggests that the evolutionary trajectory of β-defensin genes is specific to the gene, timescale, and species involved, implying that species-specific ecological and life-history differences drive divergent selective pressures on these genes. However, their evolutionary dynamics, particularly the interactions with ecological factors and life-history traits, remain insufficiently explored. RESULTS Through a comprehensive survey of 25 species spanning all major clades of Galliformes, 354 AvBD genes were identified. Comparative sequence analysis, genomic organization, and phylogenetic studies collectively reveal significant evolutionary diversification characterized by gene duplication, pseudogenization, and gene loss across these species. Notably, chicken AvBD3 exhibits significant differences in its coding regions, while AvBD6 and AvBD7 appear to have copy number variations, with species-specific paralogs of AvBD6 being especially prominent. Moreover, positive selection was more frequently observed in recently diverged gene lineages compared to ancestral ones. Using 70 samples from eight galliform species, the study further identified the prevalence of species-specific amino acid alleles. Phylogenetic comparative analysis demonstrated that the evolution of nine AvBD genes (AvBD2, -4, -5, -8, -9, -10, -11, -12, and -14) is significantly associated with specific ecological factors and life-history characteristics. Additionally, the evolutionary rates of these genes showed distinct relationship with inferred infection risk, likely reflecting the multifunctionality of β-defensins and potential trade-offs between immune defense and other biological functions. CONCLUSIONS This cross-species identification and systematic evolutionary analysis of AvBDs in Galliformes deepen our understanding of the co-evolution of host defense peptides, offering valuable insights into their natural biology and evolution, and paving the way for future applications as alternatives to traditional antibiotics.
Collapse
Affiliation(s)
- Xiaoqin Xu
- Key Laboratory of Southwest China Wildlife Resources Conservation, Ministry of Education, China West Normal University, Nanchong, 637000, P. R. China
- Sichuan Wildlife Rehabilitation and Breeding Research Center, China West Normal University, Nanchong, 637009, P. R. China
- Institute of Ecology, China West Normal University, Nanchong, 637009, P. R. China
| | - Yi Jian
- Key Laboratory of Southwest China Wildlife Resources Conservation, Ministry of Education, China West Normal University, Nanchong, 637000, P. R. China
- Sichuan Wildlife Rehabilitation and Breeding Research Center, China West Normal University, Nanchong, 637009, P. R. China
- College of Life Science, China West Normal University, Nanchong, 637000, P. R. China
| | - Lijing Huang
- Key Laboratory of Southwest China Wildlife Resources Conservation, Ministry of Education, China West Normal University, Nanchong, 637000, P. R. China
- Sichuan Wildlife Rehabilitation and Breeding Research Center, China West Normal University, Nanchong, 637009, P. R. China
- College of Life Science, China West Normal University, Nanchong, 637000, P. R. China
| | - Wei Luo
- Key Laboratory of Southwest China Wildlife Resources Conservation, Ministry of Education, China West Normal University, Nanchong, 637000, P. R. China
- Sichuan Wildlife Rehabilitation and Breeding Research Center, China West Normal University, Nanchong, 637009, P. R. China
- College of Life Science, China West Normal University, Nanchong, 637000, P. R. China
| | - Bangyuan Wu
- Key Laboratory of Southwest China Wildlife Resources Conservation, Ministry of Education, China West Normal University, Nanchong, 637000, P. R. China
- Sichuan Wildlife Rehabilitation and Breeding Research Center, China West Normal University, Nanchong, 637009, P. R. China
- College of Life Science, China West Normal University, Nanchong, 637000, P. R. China
| | - Shaohua Feng
- Key Laboratory of Southwest China Wildlife Resources Conservation, Ministry of Education, China West Normal University, Nanchong, 637000, P. R. China
- Sichuan Wildlife Rehabilitation and Breeding Research Center, China West Normal University, Nanchong, 637009, P. R. China
- College of Life Science, China West Normal University, Nanchong, 637000, P. R. China
| | - Caiquan Zhou
- Key Laboratory of Southwest China Wildlife Resources Conservation, Ministry of Education, China West Normal University, Nanchong, 637000, P. R. China
- Sichuan Wildlife Rehabilitation and Breeding Research Center, China West Normal University, Nanchong, 637009, P. R. China
- Institute of Ecology, China West Normal University, Nanchong, 637009, P. R. China
| | - Long Zhang
- Key Laboratory of Southwest China Wildlife Resources Conservation, Ministry of Education, China West Normal University, Nanchong, 637000, P. R. China.
- Sichuan Wildlife Rehabilitation and Breeding Research Center, China West Normal University, Nanchong, 637009, P. R. China.
- Institute of Ecology, China West Normal University, Nanchong, 637009, P. R. China.
| |
Collapse
|
5
|
Maharramov E, Czikkely MS, Szili P, Farkas Z, Grézal G, Daruka L, Kurkó E, Mészáros L, Daraba A, Kovács T, Bognár B, Juhász S, Papp B, Lázár V, Pál C. Exploring the principles behind antibiotics with limited resistance. Nat Commun 2025; 16:1842. [PMID: 39984459 PMCID: PMC11845477 DOI: 10.1038/s41467-025-56934-3] [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: 09/30/2024] [Accepted: 02/05/2025] [Indexed: 02/23/2025] Open
Abstract
Antibiotics that target multiple cellular functions are anticipated to be less prone to bacterial resistance. Here we hypothesize that while dual targeting is crucial, it is not sufficient in preventing resistance. Only those antibiotics that simultaneously target membrane integrity and block another cellular pathway display reduced resistance development. To test the hypothesis, we focus on three antibiotic candidates, POL7306, Tridecaptin M152-P3 and SCH79797, all of which fulfill the above criteria. Here we show that resistance evolution against these antibiotics is limited in ESKAPE pathogens, including Escherichia coli, Klebsiella pneumoniae, Acinetobacter baumannii and Pseudomonas aeruginosa, while dual-target topoisomerase antibiotics are prone to resistance. We discover several mechanisms restricting resistance. First, de novo mutations result in only a limited elevation in resistance, including those affecting the molecular targets and efflux pumps. Second, resistance is inaccessible through gene amplification. Third, functional metagenomics reveal that mobile resistance genes are rare in human gut, soil and clinical microbiomes. Finally, we detect rapid eradication of bacterial populations upon toxic exposure to membrane targeting antibiotics. We conclude that resistance mechanisms commonly found in natural bacterial pathogens provide only limited protection to these antibiotics. Our work provides guidelines for the future development of antibiotics.
Collapse
Grants
- This work was supported by: National Academy of Scientist Education Program of the National Biomedical Foundation under the sponsorship of the Hungarian Ministry of Culture and Innovation (CzM, LM) Cooperative Doctoral Program Scholarship of the Hungarian Ministry of Culture and Innovation (CzM, BB) The National Research, Development and Innovation Office, Hungary (NKFIH) grant FK-131961 (SJ) H2020-WIDESPREA-01-2016-2017-TeamingPhase2, GA:739593-HCEMM, EU’s Horizon 2020 research and innovation program under grant agreement No. 739593 (SJ) Culture and Innovation of Hungary from the National Research, Development and Innovation Fund, financed under the TKP-2021-EGA-05 funding scheme (SJ) Lendulet “Momentum” program of the Hungarian Academy of Sciences (grant agreement LP2022-12/2022) (VL) EMBO Installation Grant (grant number 5709_2024) (VL) National Laboratory for Health Security Grant RRF-2.3.1-21-2022-00006 (BP) The European Union’s Horizon 2020 Research and Innovation Programme no. 739593 (BP) National Research Development and Innovation Office grants: ‘Élvonal’ Programme KKP 129814 (BP) ERA-NET JPIAMR-ACTION (BP) National Laboratory of Biotechnology Grant 2022-2.1.1-NL-2022-00008 (CP, BP) National Research, Development and Innovation Office K146323 (CP) The European Research Council ERC-2023-ADG 101142626 FutureAntibiotics (CP)
Collapse
Affiliation(s)
- Elvin Maharramov
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre Szeged, Szeged, Hungary
- Doctoral School of Biology, University of Szeged, Szeged, Hungary
| | - Márton Simon Czikkely
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre Szeged, Szeged, Hungary
- Doctoral School of Multidisciplinary Medical Sciences, University of Szeged, Szeged, Hungary
- Department of Forensic Medicine, Albert-Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Petra Szili
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre Szeged, Szeged, Hungary
| | - Zoltán Farkas
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre Szeged, Szeged, Hungary
| | - Gábor Grézal
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre Szeged, Szeged, Hungary
- HCEMM-BRC Metabolic Systems Biology Group, Szeged, Hungary
| | - Lejla Daruka
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre Szeged, Szeged, Hungary
| | - Eszter Kurkó
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre Szeged, Szeged, Hungary
| | - Léna Mészáros
- Hungarian Centre of Excellence for Molecular Medicine, Cancer Microbiome Core Group, Budapesti út 9, Szeged, Hungary
| | - Andreea Daraba
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre Szeged, Szeged, Hungary
| | - Terézia Kovács
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre Szeged, Szeged, Hungary
- HCEMM-BRC Pharmacodynamic Drug Interaction Research Group, Szeged, Hungary
| | - Bence Bognár
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre Szeged, Szeged, Hungary
- Doctoral School of Biology, University of Szeged, Szeged, Hungary
- HCEMM-BRC Pharmacodynamic Drug Interaction Research Group, Szeged, Hungary
| | - Szilvia Juhász
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre Szeged, Szeged, Hungary
- Hungarian Centre of Excellence for Molecular Medicine, Cancer Microbiome Core Group, Budapesti út 9, Szeged, Hungary
| | - Balázs Papp
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre Szeged, Szeged, Hungary
- HCEMM-BRC Metabolic Systems Biology Group, Szeged, Hungary
| | - Viktória Lázár
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre Szeged, Szeged, Hungary
- HCEMM-BRC Pharmacodynamic Drug Interaction Research Group, Szeged, Hungary
| | - Csaba Pál
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre Szeged, Szeged, Hungary.
| |
Collapse
|
6
|
Daruka L, Czikkely MS, Szili P, Farkas Z, Balogh D, Grézal G, Maharramov E, Vu TH, Sipos L, Juhász S, Dunai A, Daraba A, Számel M, Sári T, Stirling T, Vásárhelyi BM, Ari E, Christodoulou C, Manczinger M, Enyedi MZ, Jaksa G, Kovács K, van Houte S, Pursey E, Pintér L, Haracska L, Kintses B, Papp B, Pál C. ESKAPE pathogens rapidly develop resistance against antibiotics in development in vitro. Nat Microbiol 2025; 10:313-331. [PMID: 39805953 PMCID: PMC11790497 DOI: 10.1038/s41564-024-01891-8] [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: 07/23/2023] [Accepted: 11/15/2024] [Indexed: 01/16/2025]
Abstract
Despite ongoing antibiotic development, evolution of resistance may render candidate antibiotics ineffective. Here we studied in vitro emergence of resistance to 13 antibiotics introduced after 2017 or currently in development, compared with in-use antibiotics. Laboratory evolution showed that clinically relevant resistance arises within 60 days of antibiotic exposure in Escherichia coli, Klebsiella pneumoniae, Acinetobacter baumannii and Pseudomonas aeruginosa, priority Gram-negative ESKAPE pathogens. Resistance mutations are already present in natural populations of pathogens, indicating that resistance in nature can emerge through selection of pre-existing bacterial variants. Functional metagenomics showed that mobile resistance genes to antibiotic candidates are prevalent in clinical bacterial isolates, soil and human gut microbiomes. Overall, antibiotic candidates show similar susceptibility to resistance development as antibiotics currently in use, and the corresponding resistance mechanisms overlap. However, certain combinations of antibiotics and bacterial strains were less prone to developing resistance, revealing potential narrow-spectrum antibacterial therapies that could remain effective. Finally, we develop criteria to guide efforts in developing effective antibiotic candidates.
Collapse
Grants
- The European Research Council ERC-2023-ADG: 101142626 FutureAntibiotics The National Laboratory of Biotechnology Grant: 2022-2.1.1-NL-2022-00008 National Research, Development and Innovation Office ‘Élvonal’ Programme: KKP 126506 National Research, Development and Innovation Office: K146323
- H2020-WIDESPREA-01-2016-2017-TeamingPhase2: 739593 The National Research, Development and Innovation Office, Hungary (NKFIH) grant: FK-131961 The National Research, Development and Innovation Office, Hungary (NKFIH): KIM NKFIA TKP-2021-EGA-05 The National Research, Development and Innovation Office, Hungary (NKFIH): KIM NKFIA 2022-2.1.1-NL-2022-00005
- The National Research, Development and Innovation Office, Hungary (NKFIH) grant: PD-131839
- The European Union’s Horizon 2020 research and innovation programme: 739593 The National Research, Development and Innovation Office, Hungary (NKFIH) grant: FK-142312
- The Lister Institute for Preventative Medicine
- The National Research, Development, and Innovation Office: RRF-2.3.1-21-2022-00015 The National Research, Development, and Innovation Office: TKP-31-8/PALY-2021
- The National Laboratory of Biotechnology Grant: 2022-2.1.1-NL-2022-00008 The European Union’s Horizon 2020 research and innovation programme: 739593 National Research, Development and Innovation Office grant: FK-135245 Proof of Concept grant of the Eötvös Loránd Research Network: ELKH-PoC-2022-034
Collapse
Affiliation(s)
- Lejla Daruka
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre, National Laboratory of Biotechnology, Szeged, Hungary
| | - Márton Simon Czikkely
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre, National Laboratory of Biotechnology, Szeged, Hungary
- Doctoral School of Multidisciplinary Medical Sciences, University of Szeged, Szeged, Hungary
- Department of Forensic Medicine, Albert-Szent-Györgyi Medical School, University of Szeged, Szeged, Hungary
| | - Petra Szili
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre, National Laboratory of Biotechnology, Szeged, Hungary
| | - Zoltán Farkas
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre, National Laboratory of Biotechnology, Szeged, Hungary
| | - Dávid Balogh
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre, National Laboratory of Biotechnology, Szeged, Hungary
| | - Gábor Grézal
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre, National Laboratory of Biotechnology, Szeged, Hungary
- HCEMM-BRC Metabolic Systems Biology Lab, Szeged, Hungary
| | - Elvin Maharramov
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre, National Laboratory of Biotechnology, Szeged, Hungary
- Doctoral School of Biology, University of Szeged, Szeged, Hungary
| | - Thu-Hien Vu
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre, National Laboratory of Biotechnology, Szeged, Hungary
| | - Levente Sipos
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre, National Laboratory of Biotechnology, Szeged, Hungary
| | - Szilvia Juhász
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre, National Laboratory of Biotechnology, Szeged, Hungary
- Hungarian Centre of Excellence for Molecular Medicine, Cancer Microbiome Core Group, Szeged, Hungary
| | - Anett Dunai
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre, National Laboratory of Biotechnology, Szeged, Hungary
| | - Andreea Daraba
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre, National Laboratory of Biotechnology, Szeged, Hungary
| | - Mónika Számel
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre, National Laboratory of Biotechnology, Szeged, Hungary
| | - Tóbiás Sári
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre, National Laboratory of Biotechnology, Szeged, Hungary
- Doctoral School of Biology, University of Szeged, Szeged, Hungary
| | - Tamás Stirling
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre, National Laboratory of Biotechnology, Szeged, Hungary
- Doctoral School of Biology, University of Szeged, Szeged, Hungary
| | - Bálint Márk Vásárhelyi
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre, National Laboratory of Biotechnology, Szeged, Hungary
| | - Eszter Ari
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre, National Laboratory of Biotechnology, Szeged, Hungary
- HCEMM-BRC Metabolic Systems Biology Lab, Szeged, Hungary
- Department of Genetics, ELTE Eötvös Loránd University, Budapest, Hungary
| | - Chryso Christodoulou
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre, National Laboratory of Biotechnology, Szeged, Hungary
| | - Máté Manczinger
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre, National Laboratory of Biotechnology, Szeged, Hungary
- HCEMM-BRC Systems Immunology Research Group, Szeged, Hungary
- Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary
| | - Márton Zsolt Enyedi
- Single Cell Omics Advanced Core Facility, Hungarian Centre of Excellence for Molecular Medicine, Szeged, Hungary
| | | | - Károly Kovács
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre, National Laboratory of Biotechnology, Szeged, Hungary
- HCEMM-BRC Metabolic Systems Biology Lab, Szeged, Hungary
- National Laboratory for Health Security, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Stineke van Houte
- Environment and Sustainability Institute & Centre for Ecology and Conservation, Biosciences, University of Exeter, Penryn, UK
| | - Elizabeth Pursey
- Environment and Sustainability Institute & Centre for Ecology and Conservation, Biosciences, University of Exeter, Penryn, UK
| | | | - Lajos Haracska
- Mutagenesis and Carcinogenesis Research Group, Institute of Genetics, HUN-REN Biological Research Centre, Szeged, Hungary
| | - Bálint Kintses
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre, National Laboratory of Biotechnology, Szeged, Hungary.
- HCEMM-BRC Translational Microbiology Research Group, Szeged, Hungary.
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary.
| | - Balázs Papp
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre, National Laboratory of Biotechnology, Szeged, Hungary.
- HCEMM-BRC Metabolic Systems Biology Lab, Szeged, Hungary.
- National Laboratory for Health Security, HUN-REN Biological Research Centre, Szeged, Hungary.
| | - Csaba Pál
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre, National Laboratory of Biotechnology, Szeged, Hungary.
| |
Collapse
|
7
|
Martins A, Judák F, Farkas Z, Szili P, Grézal G, Csörgő B, Czikkely MS, Maharramov E, Daruka L, Spohn R, Balogh D, Daraba A, Juhász S, Vágvölgyi M, Hunyadi A, Cao Y, Sun Z, Li X, Papp B, Pál C. Antibiotic candidates for Gram-positive bacterial infections induce multidrug resistance. Sci Transl Med 2025; 17:eadl2103. [PMID: 39772773 DOI: 10.1126/scitranslmed.adl2103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 06/17/2024] [Accepted: 11/18/2024] [Indexed: 01/11/2025]
Abstract
Several antibiotic candidates are in development against Gram-positive bacterial pathogens, but their long-term utility is unclear. To investigate this issue, we studied the laboratory evolution of resistance to antibiotics that have not yet reached the market. We found that, with the exception of compound SCH79797, antibiotic resistance generally readily evolves in Staphylococcus aureus. Cross-resistance was detected between such candidates and antibiotics currently in clinical use, including vancomycin, daptomycin, and the promising antibiotic candidate teixobactin. These patterns were driven by overlapping molecular mechanisms through mutations in regulatory systems. In particular, teixobactin-resistant bacteria displayed clinically relevant multidrug resistance and retained their virulence in an invertebrate infection model, raising concerns. More generally, we demonstrate that putative resistance mutations against candidate antibiotics are already present in natural bacterial populations. Therefore, antibiotic resistance in nature may evolve readily from the selection of preexisting genetic variants. Our work highlights the importance of predicting future evolution of resistance to antibiotic candidates at an early stage of drug development.
Collapse
Affiliation(s)
- Ana Martins
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre Szeged, Szeged HU-6726, Hungary
- Institute of Pharmacognosy, Faculty of Pharmacy, University of Szeged, Szeged HU-6720, Hungary
| | - Fanni Judák
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre Szeged, Szeged HU-6726, Hungary
- Doctoral School of Pharmaceutical Sciences, University of Szeged, Szeged HU-6720, Hungary
| | - Zoltán Farkas
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre Szeged, Szeged HU-6726, Hungary
| | - Petra Szili
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre Szeged, Szeged HU-6726, Hungary
| | - Gábor Grézal
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre Szeged, Szeged HU-6726, Hungary
- HCEMM-BRC Metabolic Systems Biology Lab, Szeged HU-6726, Hungary
| | - Bálint Csörgő
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre Szeged, Szeged HU-6726, Hungary
| | - Márton Simon Czikkely
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre Szeged, Szeged HU-6726, Hungary
- Doctoral School of Multidisciplinary Medical Sciences, University of Szeged, Szeged HU-6722, Hungary
- Department of Forensic Medicine, Albert-Szent-Györgyi Medical School, University of Szeged, Szeged HU-6722, Hungary
| | - Elvin Maharramov
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre Szeged, Szeged HU-6726, Hungary
- Doctoral School of Biology, University of Szeged, Szeged HU-6726, Hungary
| | - Lejla Daruka
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre Szeged, Szeged HU-6726, Hungary
| | - Réka Spohn
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre Szeged, Szeged HU-6726, Hungary
| | - Dávid Balogh
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre Szeged, Szeged HU-6726, Hungary
| | - Andreea Daraba
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre Szeged, Szeged HU-6726, Hungary
| | - Szilvia Juhász
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre Szeged, Szeged HU-6726, Hungary
- Cancer Microbiome Core Group, Hungarian Centre of Excellence for Molecular Medicine (HCEMM), Szeged HU-6728, Hungary
| | - Máté Vágvölgyi
- Institute of Pharmacognosy, Faculty of Pharmacy, University of Szeged, Szeged HU-6720, Hungary
| | - Attila Hunyadi
- Institute of Pharmacognosy, Faculty of Pharmacy, University of Szeged, Szeged HU-6720, Hungary
- HUN-REN-SZTE Biologically Active Natural Products Research Group, Szeged HU-6720, Hungary
| | - Yihui Cao
- Department of Chemistry, State Key Lab of Synthetic Chemistry, University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Zhenquan Sun
- Department of Chemistry, State Key Lab of Synthetic Chemistry, University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Xuechen Li
- Department of Chemistry, State Key Lab of Synthetic Chemistry, University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Balázs Papp
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre Szeged, Szeged HU-6726, Hungary
- HCEMM-BRC Metabolic Systems Biology Lab, Szeged HU-6726, Hungary
| | - Csaba Pál
- Synthetic and Systems Biology Unit, Institute of Biochemistry, HUN-REN Biological Research Centre Szeged, Szeged HU-6726, Hungary
| |
Collapse
|
8
|
Xie MQ, Wang LJ, Xiao HM, Wei SJ. Regulatory networks of mRNAs and miRNAs involved in the immune response of diamondback moth, Plutella xylostella to fungal infection. BMC Genomics 2025; 26:15. [PMID: 39762741 PMCID: PMC11706182 DOI: 10.1186/s12864-024-11192-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 12/27/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND The entomopathogenic fungus, Isaria fumosorosea, shows promise as a biological control agent in managing the diamondback moth (DBM) Plutella xylostella, a highly destructive global pest of cruciferous vegetables. To date, the miRNA-mRNA regulatory networks underlying the immune response of DBM to I. fumosorosea infection are still poorly understood. Here, we characterize the expression profiles of miRNA and mRNA, and construct the miRNA-gene regulatory network in DBM infected with I. fumosorosea. RESULTS We identified 580 differentially expressed genes (DEGs) and 55 differentially expressed miRNAs (DEMs) in I. fumosorosea-infected DBM. Among these DEGs, we found 28 immunity-related genes, which mainly include pattern recognition receptors, signal modulators, and immune effectors. Integrated analysis discovered 87 negative correlation pairs between miRNA and mRNA, involving 40 DEMs and 62 DEGs in infected DBM. Additionally, 13 miRNAs and 10 corresponding mRNAs were identified as candidate miRNA-mRNA pairs for DBM immunity against fungal infection. Gene functional enrichment analysis indicated that these miRNAs could target genes associated with various pathways, such as the immune system, infectious diseases, digestive system, endocrine system, nervous system, and signal transduction. Finally, the regulatory relationships of six miRNA-mRNA pairs were validated using quantitative reverse transcription PCR. CONCLUSIONS For the first time, we present integrated miRNA and mRNA data to elucidate the immune response of the DBM to fungal infection. Our findings enhance the understanding of the immune response of the DBM to entomopathogenic fungi infection.
Collapse
Affiliation(s)
- Mei-Qiong Xie
- College of Life Sciences and Resources and Environment, Yichun University, Yichun, 336000, China
| | - Long-Jiang Wang
- College of Chemistry and Bioengineering, Yichun University, Yichun, 336000, China.
| | - Hua-Mei Xiao
- College of Life Sciences and Resources and Environment, Yichun University, Yichun, 336000, China
| | - Shu-Jun Wei
- Institute of Plant Protection, Beijing Academy of Agriculture and Forestry Sciences, Beijing, 100097, China.
| |
Collapse
|
9
|
Sakenova N, Cacace E, Orakov A, Huber F, Varik V, Kritikos G, Michiels J, Bork P, Cossart P, Goemans CV, Typas A. Systematic mapping of antibiotic cross-resistance and collateral sensitivity with chemical genetics. Nat Microbiol 2025; 10:202-216. [PMID: 39623067 PMCID: PMC11726442 DOI: 10.1038/s41564-024-01857-w] [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: 01/24/2024] [Accepted: 10/13/2024] [Indexed: 01/12/2025]
Abstract
By acquiring or evolving resistance to one antibiotic, bacteria can become cross-resistant to a second antibiotic, which further limits therapeutic choices. In the opposite scenario, initial resistance leads to collateral sensitivity to a second antibiotic, which can inform cycling or combinatorial treatments. Despite their clinical relevance, our knowledge of both interactions is limited. We used published chemical genetics data of the Escherichia coli single-gene deletion library in 40 antibiotics and devised a metric that discriminates between known cross-resistance and collateral-sensitivity antibiotic interactions. Thereby we inferred 404 cases of cross-resistance and 267 of collateral-sensitivity, expanding the number of known interactions by over threefold. We further validated 64/70 inferred interactions using experimental evolution. By identifying mutants driving these interactions in chemical genetics, we demonstrated that a drug pair can exhibit both interactions depending on the resistance mechanism. Finally, we applied collateral-sensitive drug pairs in combination to reduce antibiotic-resistance development in vitro.
Collapse
Affiliation(s)
- Nazgul Sakenova
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Center for Microbiology, VIB-KU Leuven, Leuven, Belgium
- Center of Microbial and Plant Genetics, KU Leuven, Leuven, Belgium
| | - Elisabetta Cacace
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Institute of Microbiology and Swiss Institute of Bioinformatics, ETH Zürich, Zürich, Switzerland
| | - Askarbek Orakov
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Florian Huber
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Vallo Varik
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - George Kritikos
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- European Food Safety Authority, Parma, Italy
| | - Jan Michiels
- Center for Microbiology, VIB-KU Leuven, Leuven, Belgium
- Center of Microbial and Plant Genetics, KU Leuven, Leuven, Belgium
| | - Peer Bork
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Department of Bioinformatics, University of Würzburg, Würzburg, Germany
- Max Delbrück Centre for Molecular Medicine, Berlin, Germany
| | - Pascale Cossart
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Department of Cell Biology and Infection, Institut Pasteur, Paris, France
| | - Camille V Goemans
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
- Global Health Institute, School of Life Sciences, École Polytechnique Federale de Lausanne, Lausanne, Switzerland.
| | - Athanasios Typas
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
| |
Collapse
|
10
|
Walker AS, Clardy J. Primed for Discovery. Biochemistry 2024; 63:2705-2713. [PMID: 39497571 PMCID: PMC11542185 DOI: 10.1021/acs.biochem.4c00464] [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: 08/10/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 11/09/2024]
Abstract
Antibiotics are essential components of current medical practice, but their effectiveness is being eroded by the increasing emergence of antimicrobial-resistant infections. At the same time, the rate of antibiotic discovery has slowed, and our future ability to treat infections is threatened. Among Christopher T. Walsh's many contributions to science was his early recognition of this threat and the potential of biosynthesis─genes and mechanisms─to contribute solutions. Here, we revisit a 2006 review by Walsh and co-workers that highlighted a major challenge in antibiotic natural product discovery: the daunting odds for identifying new naturally occurring antibiotics. The review described strategies to mitigate the odds challenge. These strategies have been used extensively by the natural product discovery community in the years since and have resulted in some promising discoveries. Despite these advances, the rarity of novel antibiotic natural products remains a barrier to discovery. We compare the challenge of discovering natural product antibiotics to the process of identifying new prime numbers, which are also challenging to find and an essential, if underappreciated, element of modern life. We propose that inclusion of filters for functional compounds early in the discovery pipeline is key to natural product antibiotic discovery, review some recent advances that enable this, and discuss some remaining challenges that need to be addressed to make antibiotic discovery sustainable in the future.
Collapse
Affiliation(s)
- Allison S. Walker
- Department
of Chemistry, Vanderbilt University, Nashville, Tennessee 37240, United States
- Department
of Biological Sciences, Vanderbilt University, Nashville, Tennessee 37232, United States
- Department
of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
| | - Jon Clardy
- Department
of Biological Chemistry and Molecular Pharmacology, Harvard Medical School and Blavatnik Institute, Boston, Massachusetts 02115, United States
| |
Collapse
|
11
|
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
|
12
|
Wei Y, Fu J, Liu E, Gao J, Lv Y, Li Z. Injectable hydrogels doped with PDA nanoparticles for photothermal bacterial inhibition and rapid wound healing in vitro. RSC Adv 2024; 14:2778-2791. [PMID: 38234872 PMCID: PMC10792480 DOI: 10.1039/d3ra08219a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 01/02/2024] [Indexed: 01/19/2024] Open
Abstract
The difficulty of wound healing due to skin defects has been a great challenge due to the complex inflammatory microenvironment. Delayed wound healing severely affects the quality of life of patients and represents a significant economic burden for public health systems worldwide. Therefore, there is an urgent need for the development of novel wound dressings that can efficiently resist drug-resistant bacteria and have superior wound repair capabilities in clinical applications. In this study, we designed an adhesive antimicrobial hydrogel dressing (GMH) based on methacrylic-anhydride-modified gelatin and oxidized hyaluronic acid formed by Schiff base and UV-induced double cross-linking for infected wound repair. By inserting PDA nanoparticles into the hydrogel (GMH/PDA), the hydrogel has the capability of photothermal conversion and exhibits good photothermal antimicrobial properties under near-infrared (NIR) light irradiation, which helps to reduce the inflammatory response and avoid bacterial infections during the wound healing process. In addition, GMH/PDA hydrogel exhibits excellent injectability, allowing the hydrogel dressings to be adapted to complex wound surfaces, making them promising candidates for wound therapy. In conclusion, the multifunctional injectable GMH/PDA hydrogel possesses high antimicrobial efficiency, antioxidant properties and good biocompatibility, making them promising candidates for the treatment of infected skin wounds.
Collapse
Affiliation(s)
- Ying Wei
- Department of Operating Room, The Affiliated Hospital of Qingdao University 266003 Qingdao China
| | - Junhua Fu
- Department of Operating Room, The Affiliated Hospital of Qingdao University 266003 Qingdao China
| | - Enrui Liu
- Department of Emergency Surgery, The Affiliated Hospital of Qingdao University 266003 Qingdao China
| | - Junru Gao
- Department of Outpatient, The Affiliated Hospital of Qingdao University 266003 Qingdao China
| | - Yaqing Lv
- Department of Outpatient, The Affiliated Hospital of Qingdao University 266003 Qingdao China
| | - Zhenlu Li
- Department of Emergency Surgery, The Affiliated Hospital of Qingdao University 266003 Qingdao China
| |
Collapse
|
13
|
Dwivedi M, Parmar MD, Mukherjee D, Yadava A, Yadav H, Saini NP. Biochemistry, Mechanistic Intricacies, and Therapeutic Potential of Antimicrobial Peptides: An Alternative to Traditional Antibiotics. Curr Med Chem 2024; 31:6110-6139. [PMID: 37818561 DOI: 10.2174/0109298673268458230926105224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/03/2023] [Accepted: 08/24/2023] [Indexed: 10/12/2023]
Abstract
The emergence of drug-resistant strains of pathogens becomes a major obstacle to treating human diseases. Antibiotics and antivirals are in the application for a long time but now these drugs are not much effective anymore against disease-causing drugresistant microbes and gradually it is becoming a serious complication worldwide. The development of new antibiotics cannot be a stable solution to treat drug-resistant strains due to their evolving nature and escaping antibiotics. At this stage, antimicrobial peptides (AMPs) may provide us with novel therapeutic leads against drug-resistant pathogens. Structurally, antimicrobial peptides are mostly α-helical peptide molecules with amphiphilic properties that carry the positive charge (cationic) and belong to host defense peptides. These positively charged AMPs can interact with negatively charged bacterial cell membranes and may cause the alteration in electrochemical potential on bacterial cell membranes and consequently lead to the death of microbial cells. In the present study, we will elaborate on the implication of AMPs in the treatment of various diseases along with their specific structural and functional properties. This review will provide information which assists in the development of new synthetic peptide analogues to natural AMPs. These analogues will eliminate the limitations of natural AMPs like toxicity and severe hemolytic activities.
Collapse
Affiliation(s)
- Manish Dwivedi
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow, 226028, India
| | - Meet Dineshbhai Parmar
- Department of Biological Sciences and Biotechnology, Institute of Advanced Research, Gandhinagar, Gujarat, India
| | | | - Anuradha Yadava
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow, 226028, India
| | - Hitendra Yadav
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow, 226028, India
| | - Nandini Pankaj Saini
- Amity Institute of Biotechnology, Amity University Uttar Pradesh, Lucknow, 226028, India
| |
Collapse
|
14
|
Salinas-Restrepo C, Naranjo-Duran AM, Quintana J, Bueno J, Guzman F, Hoyos Palacio LM, Segura C. Short Antimicrobial Peptide Derived from the Venom Gland Transcriptome of Pamphobeteus verdolaga Increases Gentamicin Susceptibility of Multidrug-Resistant Klebsiella pneumoniae. Antibiotics (Basel) 2023; 13:6. [PMID: 38275316 PMCID: PMC10812672 DOI: 10.3390/antibiotics13010006] [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: 10/11/2023] [Revised: 11/07/2023] [Accepted: 11/17/2023] [Indexed: 01/27/2024] Open
Abstract
Infectious diseases account for nine percent of annual human deaths, and the widespread emergence of antimicrobial resistances threatens to significantly increase this number in the coming decades. The prospect of antimicrobial peptides (AMPs) derived from venomous animals presents an interesting alternative for developing novel active pharmaceutical ingredients (APIs). Small, cationic and amphiphilic peptides were predicted from the venom gland transcriptome of Pamphobeteus verdolaga using a custom database of the arthropod's AMPs. Ninety-four candidates were chemically synthesized and screened against ATCC® strains of Escherichia coli and Staphylococcus aureus. Among them, one AMP, named PvAMP66, showed broad-spectrum antimicrobial properties with selectivity towards Gram-negative bacteria. It also exhibited activity against Pseudomonas aeruginosa, as well as both an ATCC® and a clinically isolated multidrug-resistant (MDR) strain of K. pneumoniae. The scanning electron microscopy analysis revealed that PvAMP66 induced morphological changes of the MDR K. pneumoniae strain suggesting a potential "carpet model" mechanism of action. The isobologram analysis showed an additive interaction between PvAMP66 and gentamicin in inhibiting the growth of MDR K. pneumoniae, leading to a ten-fold reduction in gentamicin's effective concentration. A cytotoxicity against erythrocytes or peripheral blood mononuclear cells was observed at concentrations three to thirteen-fold higher than those exhibited against the evaluated bacterial strains. This evidence suggests that PvAMP66 can serve as a template for the development of AMPs with enhanced activity and deserves further pre-clinical studies as an API in combination therapy.
Collapse
Affiliation(s)
- Cristian Salinas-Restrepo
- Grupo Toxinología, Alternativas Terapéuticas y Alimentarias, Facultad de Ciencias Farmacéuticas y Alimentarias, Universidad de Antioquia, Medellín 050012, Colombia; (C.S.-R.); (A.M.N.-D.)
| | - Ana María Naranjo-Duran
- Grupo Toxinología, Alternativas Terapéuticas y Alimentarias, Facultad de Ciencias Farmacéuticas y Alimentarias, Universidad de Antioquia, Medellín 050012, Colombia; (C.S.-R.); (A.M.N.-D.)
| | - Juan Quintana
- Facultad de Medicina, Universidad Cooperativa de Colombia, Medellín 050012, Colombia;
| | - Julio Bueno
- Grupo Reproducción, Facultad de Medicina, Universidad de Antioquia, Medellín 050012, Colombia;
| | - Fanny Guzman
- Núcleo Biotecnología Curauma (NBC), Pontificia Universidad Católica de Valparaíso, Valparaíso 3100000, Chile;
| | - Lina M. Hoyos Palacio
- Escuela de Ciencias de la Salud, Grupo de Investigación Biología de Sistemas, Universidad Pontificia Bolivariana, Medellín 050031, Colombia;
| | - Cesar Segura
- Grupo Malaria, Facultad de Medicina, Universidad de Antioquia, Medellín 050012, Colombia
| |
Collapse
|
15
|
Bui Thi Phuong H, Le Uyen C, Doan Ngan H, Luong Xuan H. Impact of chemical modifications on the antimicrobial and hemolytic activity of helical amphipathic peptide Lasioglossin LL-III. Amino Acids 2023; 55:1531-1544. [PMID: 37737904 DOI: 10.1007/s00726-023-03326-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 08/29/2023] [Indexed: 09/23/2023]
Abstract
Insect venom is abundant in potential antimicrobial peptides (AMPs), which can serve as novel alternatives to conventional antibiotics. Among them, Lasioglossin III LL-III) is a promising candidate with a broad spectrum against many fungi strains and both types of bacteria, whereas almost non-toxic to red blood cells. Many chemical approaches have been recently applied to improve its pharmacological properties and provide useful information regarding structure-activity relationships. Hence, this review focused on highlighting the lesson learned from each modification and supporting the future design of potent, selective, and metabolically stable AMPs.
Collapse
Affiliation(s)
| | - Chi Le Uyen
- Faculty of Pharmacy, Phenikaa University, Hanoi, 12116, Vietnam
| | - Hoa Doan Ngan
- Faculty of Medical Technology, Phenikaa University, Hanoi, 12116, Vietnam
| | - Huy Luong Xuan
- Faculty of Pharmacy, Phenikaa University, Hanoi, 12116, Vietnam.
- Phenikaa Institute for Advanced Study (PIAS), Phenikaa University, Hanoi, 12116, Vietnam.
| |
Collapse
|
16
|
Javed A, Balhuizen MD, Pannekoek A, Bikker FJ, Heesterbeek DAC, Haagsman HP, Broere F, Weingarth M, Veldhuizen EJA. Effects of Escherichia coli LPS Structure on Antibacterial and Anti-Endotoxin Activities of Host Defense Peptides. Pharmaceuticals (Basel) 2023; 16:1485. [PMID: 37895956 PMCID: PMC10609994 DOI: 10.3390/ph16101485] [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: 09/15/2023] [Revised: 10/13/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
The binding of Host Defense Peptides (HDPs) to the endotoxin of Gram-negative bacteria has important unsolved aspects. For most HDPs, it is unclear if binding is part of the antibacterial mechanism or whether LPS actually provides a protective layer against HDP killing. In addition, HDP binding to LPS can block the subsequent TLR4-mediated activation of the immune system. This dual activity is important, considering that HDPs are thought of as an alternative to conventional antibiotics, which do not provide this dual activity. In this study, we systematically determine, for the first time, the influence of the O-antigen and Lipid A composition on both the antibacterial and anti-endotoxin activity of four HDPs (CATH-2, PR-39, PMAP-23, and PMAP36). The presence of the O-antigen did not affect the antibacterial activity of any of the tested HDPs. Similarly, modification of the lipid A phosphate (MCR-1 phenotype) also did not affect the activity of the HDPs. Furthermore, assessment of inner and outer membrane damage revealed that CATH-2 and PMAP-36 are profoundly membrane-active and disrupt the inner and outer membrane of Escherichia coli simultaneously, suggesting that crossing the outer membrane is the rate-limiting step in the bactericidal activity of these HDPs but is independent of the presence of an O-antigen. In contrast to killing, larger differences were observed for the anti-endotoxin properties of HDPs. CATH-2 and PMAP-36 were much stronger at suppressing LPS-induced activation of macrophages compared to PR-39 and PMAP-23. In addition, the presence of only one phosphate group in the lipid A moiety reduced the immunomodulating activity of these HDPs. Overall, the data strongly suggest that LPS composition has little effect on bacterial killing but that Lipid A modification can affect the immunomodulatory role of HDPs. This dual activity should be considered when HDPs are considered for application purposes in the treatment of infectious diseases.
Collapse
Affiliation(s)
- Ali Javed
- Section of Immunology, Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, The Netherlands; (A.J.); (A.P.); (F.B.)
- NMR Spectroscopy, Bijvoet Centre for Biomolecular Research, Department of Chemistry, Faculty of Science, Utrecht University, 3584 CS Utrecht, The Netherlands;
| | - Melanie D. Balhuizen
- Section of Molecular Host Defense, Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, The Netherlands; (M.D.B.); (H.P.H.)
| | - Arianne Pannekoek
- Section of Immunology, Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, The Netherlands; (A.J.); (A.P.); (F.B.)
| | - Floris J. Bikker
- Department of Oral Biochemistry, Academic Centre for Dentistry Amsterdam, University of Amsterdam and VU University Amsterdam, 1081 LA Amsterdam, The Netherlands;
| | - Dani A. C. Heesterbeek
- Department of Medical Microbiology, University Medical Centre Utrecht, 3584 CX Utrecht, The Netherlands;
| | - Henk P. Haagsman
- Section of Molecular Host Defense, Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, The Netherlands; (M.D.B.); (H.P.H.)
| | - Femke Broere
- Section of Immunology, Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, The Netherlands; (A.J.); (A.P.); (F.B.)
| | - Markus Weingarth
- NMR Spectroscopy, Bijvoet Centre for Biomolecular Research, Department of Chemistry, Faculty of Science, Utrecht University, 3584 CS Utrecht, The Netherlands;
| | - Edwin J. A. Veldhuizen
- Section of Immunology, Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, 3584 CL Utrecht, The Netherlands; (A.J.); (A.P.); (F.B.)
| |
Collapse
|
17
|
Jangir PK, Ogunlana L, Szili P, Czikkely M, Shaw LP, Stevens EJ, Yu Y, Yang Q, Wang Y, Pál C, Walsh TR, MacLean CR. The evolution of colistin resistance increases bacterial resistance to host antimicrobial peptides and virulence. eLife 2023; 12:e84395. [PMID: 37094804 PMCID: PMC10129329 DOI: 10.7554/elife.84395] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 04/05/2023] [Indexed: 04/26/2023] Open
Abstract
Antimicrobial peptides (AMPs) offer a promising solution to the antibiotic resistance crisis. However, an unresolved serious concern is that the evolution of resistance to therapeutic AMPs may generate cross-resistance to host AMPs, compromising a cornerstone of the innate immune response. We systematically tested this hypothesis using globally disseminated mobile colistin resistance (MCR) that has been selected by the use of colistin in agriculture and medicine. Here, we show that MCR provides a selective advantage to Escherichia coli in the presence of key AMPs from humans and agricultural animals by increasing AMP resistance. Moreover, MCR promotes bacterial growth in human serum and increases virulence in a Galleria mellonella infection model. Our study shows how the anthropogenic use of AMPs can drive the accidental evolution of resistance to the innate immune system of humans and animals. These findings have major implications for the design and use of therapeutic AMPs and suggest that MCR may be difficult to eradicate, even if colistin use is withdrawn.
Collapse
Affiliation(s)
- Pramod K Jangir
- Department of Biology, University of OxfordOxfordUnited Kingdom
| | - Lois Ogunlana
- Department of Biology, University of OxfordOxfordUnited Kingdom
| | - Petra Szili
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research NetworkSzegedHungary
- Doctoral School of Multidisciplinary Medical Sciences, University of SzegedSzegedHungary
| | - Marton Czikkely
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research NetworkSzegedHungary
| | - Liam P Shaw
- Department of Biology, University of OxfordOxfordUnited Kingdom
| | - Emily J Stevens
- Department of Biology, University of OxfordOxfordUnited Kingdom
| | - Yang Yu
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural UniversityGuangzhouChina
| | - Qiue Yang
- Fujian Provincial Key Laboratory of Soil Environmental Health and RegulaWon, College of Resources and Environment, Fujian Agriculture and Forestry UniversityFuzhouChina
| | - Yang Wang
- Beijing Advanced Innovation Centre for Food Nutrition and Human Health, College of Veterinary Medicine, China Agricultural UniversityBeijingChina
| | - Csaba Pál
- Synthetic and Systems Biology Unit, Biological Research Centre, Eötvös Loránd Research NetworkSzegedHungary
| | - Timothy R Walsh
- Department of Biology, University of OxfordOxfordUnited Kingdom
| | - Craig R MacLean
- Department of Biology, University of OxfordOxfordUnited Kingdom
| |
Collapse
|
18
|
XENOFOOD—An Autoclaved Feed Supplement Containing Autoclavable Antimicrobial Peptides—Exerts Anticoccidial GI Activity, and Causes Bursa Enlargement, but Has No Detectable Harmful Effects in Broiler Cockerels despite In Vitro Detectable Cytotoxicity on LHM Cells. Pathogens 2023; 12:pathogens12030458. [PMID: 36986380 PMCID: PMC10059668 DOI: 10.3390/pathogens12030458] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 03/09/2023] [Accepted: 03/11/2023] [Indexed: 03/17/2023] Open
Abstract
Entomopathogenic bacteria are obligate symbionts of entomopathogenic nematode (EPN) species. These bacteria biosynthesize and release non-ribosomal-templated hybrid peptides (NR-AMPs), with strong, and large-spectral antimicrobial potential, capable of inactivating pathogens belonging to different prokaryote, and eukaryote taxa. The cell-free conditioned culture media (CFCM) of Xenorhabdus budapestensis and X. szentirmaii efficiently inactivate poultry pathogens like Clostridium, Histomonas, and Eimeria. To learn whether a bio-preparation containing antimicrobial peptides of Xenorhabdus origin with accompanying (in vitro detectable) cytotoxic effects could be considered a safely applicable preventive feed supplement, we conducted a 42-day feeding experiment on freshly hatched broiler cockerels. XENOFOOD (containing autoclaved X. budapestensis, and X. szentirmaii cultures developed on chicken food) were consumed by the birds. The XENOFOOD exerted detectable gastrointestinal (GI) activity (reducing the numbers of the colony-forming Clostridium perfringens units in the lower jejunum. No animal was lost in the experiment. Neither the body weight, growth rate, feed-conversion ratio, nor organ-weight data differed between the control (C) and treated (T) groups, indicating that the XENOFOOD diet did not result in any detectable adverse effects. We suppose that the parameters indicating a moderate enlargement of bursas of Fabricius (average weight, size, and individual bursa/spleen weight-ratios) in the XENOFOOD-fed group must be an indirect indication that the bursa-controlled humoral immune system neutralized the cytotoxic ingredients of the XENOFOOD in the blood, not allowing to reach their critical cytotoxic concentration in the sensitive tissues.
Collapse
|
19
|
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 PMCID: PMC11537282 DOI: 10.1016/j.scitotenv.2022.160461] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [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
|
20
|
Wang X, Xie W, Zhang S, Shao Y, Cai J, Cai L, Wang X, Shan Z, Zhou H, Li J, Cui W, Wang L, Qiao X, Li Y, Jiang Y, Tang L. Effect of Microencapsulation Techniques on the Stress Resistance and Biological Activity of Bovine Lactoferricin-Lactoferrampin-Encoding Lactobacillus reuteri. Foods 2022; 11:3169. [PMID: 37430918 PMCID: PMC9602003 DOI: 10.3390/foods11203169] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 09/16/2022] [Accepted: 10/08/2022] [Indexed: 08/27/2023] Open
Abstract
Bovine lactoferricin-lactoferrampin-encoding Lactobacillus reuteri (LR-LFCA) has been found to benefit its host by strengthening its intestinal barrier. However, several questions remain open concerning genetically engineered strains maintaining long-term biological activity at room temperature. In addition, probiotics are vulnerable to harsh conditions in the gut, such as acidity and alkalinity, and bile salts. Microencapsulation is a technique to entrap probiotic bacteria into gastro-resistant polymers to carry them directly to the intestine. We selected nine kinds of wall material combinations to encapsulate LR-LFCA by spray drying microencapsulation. The storage stability, microstructural morphology, biological activity, and simulated digestion in vivo or in vitro of the microencapsulated LR-LFCA were further evaluated. The results showed that LR-LFCA had the highest survival rate when microcapsules were prepared using a wall material mixture (skim milk, sodium glutamate, polyvinylpyrrolidone, maltodextrin, and gelatin). Microencapsulated LR-LFCA increased the stress resistance capacity and colonization abilities. In the present study, we have identified a suitable wall material formulation for spray-dried microencapsulation of genetically engineered probiotic products, which would facilitate their storage and transport.
Collapse
Affiliation(s)
- Xueying Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Weichun Xie
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Senhao Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Yilan Shao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Jiyao Cai
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Limeng Cai
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Xiaona Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Zhifu Shan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Han Zhou
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Jiaxuan Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Wen Cui
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Li Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Xinyuan Qiao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Northeast Agricultural University, Harbin 150030, China
| | - Yijing Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Northeast Agricultural University, Harbin 150030, China
| | - Yanping Jiang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Lijie Tang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
- Heilongjiang Key Laboratory for Animal Disease Control and Pharmaceutical Development, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
21
|
Jangir PK, Yang Q, Shaw LP, Caballero JD, Ogunlana L, Wheatley R, Walsh T, MacLean RC. Pre-existing chromosomal polymorphisms in pathogenic E. coli potentiate the evolution of resistance to a last-resort antibiotic. eLife 2022; 11:e78834. [PMID: 35943060 PMCID: PMC9363117 DOI: 10.7554/elife.78834] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 07/22/2022] [Indexed: 12/17/2022] Open
Abstract
Bacterial pathogens show high levels of chromosomal genetic diversity, but the influence of this diversity on the evolution of antibiotic resistance by plasmid acquisition remains unclear. Here, we address this problem in the context of colistin, a 'last line of defence' antibiotic. Using experimental evolution, we show that a plasmid carrying the MCR-1 colistin resistance gene dramatically increases the ability of Escherichia coli to evolve high-level colistin resistance by acquiring mutations in lpxC, an essential chromosomal gene involved in lipopolysaccharide biosynthesis. Crucially, lpxC mutations increase colistin resistance in the presence of the MCR-1 gene, but decrease the resistance of wild-type cells, revealing positive sign epistasis for antibiotic resistance between the chromosomal mutations and a mobile resistance gene. Analysis of public genomic datasets shows that lpxC polymorphisms are common in pathogenic E. coli, including those carrying MCR-1, highlighting the clinical relevance of this interaction. Importantly, lpxC diversity is high in pathogenic E. coli from regions with no history of MCR-1 acquisition, suggesting that pre-existing lpxC polymorphisms potentiated the evolution of high-level colistin resistance by MCR-1 acquisition. More broadly, these findings highlight the importance of standing genetic variation and plasmid/chromosomal interactions in the evolutionary dynamics of antibiotic resistance.
Collapse
Affiliation(s)
- Pramod K Jangir
- Department of Zoology, University of OxfordOxfordUnited Kingdom
| | - Qiue Yang
- Fujian Provincial Key Laboratory of Soil Environmental Health and Regulation, College of Resources and Environment, Fujian Agriculture and Forestry UniversityFuzhouChina
| | - Liam P Shaw
- Department of Zoology, University of OxfordOxfordUnited Kingdom
| | | | - Lois Ogunlana
- Department of Zoology, University of OxfordOxfordUnited Kingdom
| | - Rachel Wheatley
- Department of Zoology, University of OxfordOxfordUnited Kingdom
| | - Timothy Walsh
- Department of Zoology, University of OxfordOxfordUnited Kingdom
| | - R Craig MacLean
- Department of Zoology, University of OxfordOxfordUnited Kingdom
| |
Collapse
|
22
|
The EnvZ/OmpR Two-Component System Regulates the Antimicrobial Activity of TAT-RasGAP 317-326 and the Collateral Sensitivity to Other Antibacterial Agents. Microbiol Spectr 2022; 10:e0200921. [PMID: 35579440 PMCID: PMC9241736 DOI: 10.1128/spectrum.02009-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The rapid emergence of antibiotic-resistant bacteria poses a serious threat to public health worldwide. Antimicrobial peptides (AMPs) are promising antibiotic alternatives; however, little is known about bacterial mechanisms of AMP resistance and the interplay between AMP resistance and the bacterial response to other antimicrobials. In this study, we identified Escherichia coli mutants resistant to the TAT-RasGAP317-326 antimicrobial peptide and found that resistant bacteria show collateral sensitivity to other AMPs and antibacterial agents. We determined that resistance to TAT-RasGAP317-326 peptide arises through mutations in the histidine kinase EnvZ, a member of the EnvZ/OmpR two-component system responsible for osmoregulation in E. coli. In particular, we found that TAT-RasGAP317-326 binding and entry is compromised in E. coli peptide-resistant mutants. We showed that peptide resistance is associated with transcriptional regulation of a number of pathways and EnvZ-mediated resistance is dependent on the OmpR response regulator but is independent of the OmpC and OmpF outer membrane porins. Our findings provide insight into the bacterial mechanisms of TAT-RasGAP317-326 resistance and demonstrate that resistance to this AMP is associated with collateral sensitivity to other antibacterial agents. IMPORTANCE Antimicrobial peptides (AMP) are promising alternatives to classical antibiotics in the fight against antibiotic resistance. Resistance toward antimicrobial peptides can occur, but little is known about the mechanisms driving this phenomenon. Moreover, there is limited knowledge on how AMP resistance relates to the bacterial response to other antimicrobial agents. Here, we address these questions in the context of the antimicrobial peptide TAT-RasGAP317-326. We show that resistant Escherichia coli strains can be selected and do not show resistance to other antimicrobial agents. Resistance is caused by a mutation in a regulatory pathway, which lowers binding and entry of the peptide in E. coli. Our results highlight a mechanism of resistance that is specific to TAT-RasGAP317-326. Further research is required to characterize these mechanisms and to evaluate the potential of antimicrobial combinations to curb the development of antimicrobial resistance.
Collapse
|
23
|
Ferguson PM, Clarke M, Manzo G, Hind CK, Clifford M, Sutton JM, Lorenz CD, Phoenix DA, Mason AJ. Temporin B Forms Hetero-Oligomers with Temporin L, Modifies Its Membrane Activity, and Increases the Cooperativity of Its Antibacterial Pharmacodynamic Profile. Biochemistry 2022; 61:1029-1040. [PMID: 35609188 PMCID: PMC9178791 DOI: 10.1021/acs.biochem.1c00762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
The pharmacodynamic
profile of antimicrobial peptides (AMPs) and
their in vivo synergy are two factors that are thought
to restrict resistance evolution and ensure their conservation. The
frog Rana temporaria secretes a family of closely
related AMPs, temporins A–L, as an effective chemical dermal
defense. The antibacterial potency of temporin L has been shown to
increase synergistically in combination with both temporins B and
A, but this is modest. Here we show that the less potent temporin
B enhances the cooperativity of the in vitro antibacterial
activity of the more potent temporin L against EMRSA-15 and that this
may be associated with an altered interaction with the bacterial plasma
membrane, a feature critical for the antibacterial activity of most
AMPs. Addition of buforin II, a histone H2A fragment, can further
increase the cooperativity. Molecular dynamics simulations indicate
temporins B and L readily form hetero-oligomers in models of Gram-positive
bacterial plasma membranes. Patch-clamp studies show transmembrane
ion conductance is triggered with lower amounts of both peptides and
more quickly when used in combination, but conductance is of a lower
amplitude and pores are smaller. Temporin B may therefore act by forming
temporin L/B hetero-oligomers that are more effective than temporin
L homo-oligomers at bacterial killing and/or by reducing the probability
of the latter forming until a threshold concentration is reached.
Exploration of the mechanism of synergy between AMPs isolated from
the same organism may therefore yield antibiotic combinations with
advantageous pharmacodynamic properties.
Collapse
Affiliation(s)
- Philip M Ferguson
- Institute of Pharmaceutical Science, School of Cancer & Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Maria Clarke
- Institute of Pharmaceutical Science, School of Cancer & Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Giorgia Manzo
- Institute of Pharmaceutical Science, School of Cancer & Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Charlotte K Hind
- Technology Development Group, UKHSA, Salisbury SP4 0JG, United Kingdom
| | - Melanie Clifford
- Technology Development Group, UKHSA, Salisbury SP4 0JG, United Kingdom
| | - J Mark Sutton
- Institute of Pharmaceutical Science, School of Cancer & Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom.,Technology Development Group, UKHSA, Salisbury SP4 0JG, United Kingdom
| | - Christian D Lorenz
- Department of Physics, King's College London, London WC2R 2LS, United Kingdom
| | - David A Phoenix
- School of Applied Science, London South Bank University, 103 Borough Road, London SE1 0AA, United Kingdom
| | - A James Mason
- Institute of Pharmaceutical Science, School of Cancer & Pharmaceutical Science, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, United Kingdom
| |
Collapse
|
24
|
Fodor A, Gualtieri M, Zeller M, Tarasco E, Klein MG, Fodor AM, Haynes L, Lengyel K, Forst SA, Furgani GM, Karaffa L, Vellai T. Type Strains of Entomopathogenic Nematode-Symbiotic Bacterium Species, Xenorhabdus szentirmaii (EMC) and X. budapestensis (EMA), Are Exceptional Sources of Non-Ribosomal Templated, Large-Target-Spectral, Thermotolerant-Antimicrobial Peptides (by Both), and Iodinin (by EMC). Pathogens 2022; 11:pathogens11030342. [PMID: 35335666 PMCID: PMC8950435 DOI: 10.3390/pathogens11030342] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/18/2022] [Accepted: 02/23/2022] [Indexed: 01/26/2023] Open
Abstract
Antimicrobial multidrug resistance (MDR) is a global challenge, not only for public health, but also for sustainable agriculture. Antibiotics used in humans should be ruled out for use in veterinary or agricultural settings. Applying antimicrobial peptide (AMP) molecules, produced by soil-born organisms for protecting (soil-born) plants, seems a preferable alternative. The natural role of peptide-antimicrobials, produced by the prokaryotic partner of entomopathogenic-nematode/bacterium (EPN/EPB) symbiotic associations, is to sustain monoxenic conditions for the EPB in the gut of the semi-anabiotic infective dauer juvenile (IJ) EPN. They keep pathobiome conditions balanced for the EPN/EPB complex in polyxenic (soil, vanquished insect cadaver) niches. Xenorhabdus szentirmaii DSM16338(T) (EMC), and X. budapestensis DSM16342(T) (EMA), are the respective natural symbionts of EPN species Steinernema rarum and S. bicornutum. We identified and characterized both of these 15 years ago. The functional annotation of the draft genome of EMC revealed 71 genes encoding non-ribosomal peptide synthases, and polyketide synthases. The large spatial Xenorhabdus AMP (fabclavine), was discovered in EMA, and its biosynthetic pathway in EMC. The AMPs produced by EMA and EMC are promising candidates for controlling MDR prokaryotic and eukaryotic pathogens (bacteria, oomycetes, fungi, protozoa). EMC releases large quantity of iodinin (1,6-dihydroxyphenazine 5,10-dioxide) in a water-soluble form into the media, where it condenses to form spectacular water-insoluble, macroscopic crystals. This review evaluates the scientific impact of international research on EMA and EMC.
Collapse
Affiliation(s)
- András Fodor
- Department of Genetics, Eötvös University, Pázmány Péter Sétány 1/C, H-1117 Budapest, Hungary; (A.M.F.); (K.L.); or (G.M.F.); or (T.V.)
- Department of Genetics, University of Szeged, Középfasor 52, H-6726 Szeged, Hungary
- Correspondence: ; Tel.: +36-(30)-490-9294
| | - Maxime Gualtieri
- Nosopharm, 110 Allée Charles Babbage, Espace Innovation 2, 30000 Nîmes, France;
| | - Matthias Zeller
- Department of Chemistry, Purdue University, 560 Oval Drive, West Lafayette, IN 47906, USA;
| | - Eustachio Tarasco
- Department of Soil, Plant and Food Sciences, University of Bari “Aldo Moro”, Via Amendola 165/A, 70126 Bari, Italy;
- Institute for Sustainable Plant Protection of CNR, Via Amendola 122/D, 70126 Bari, Italy
| | - Michael G. Klein
- USDA-ARS & Department of Entomology, The Ohio State University, 13416 Claremont Ave, Cleveland, OH 44130, USA;
| | - Andrea M. Fodor
- Department of Genetics, Eötvös University, Pázmány Péter Sétány 1/C, H-1117 Budapest, Hungary; (A.M.F.); (K.L.); or (G.M.F.); or (T.V.)
| | - Leroy Haynes
- Department of Chemistry, The College of Wooster, Wooster, OH 44691, USA;
| | - Katalin Lengyel
- Department of Genetics, Eötvös University, Pázmány Péter Sétány 1/C, H-1117 Budapest, Hungary; (A.M.F.); (K.L.); or (G.M.F.); or (T.V.)
- National Institute of Pharmacy and Nutrition (NIPN), Zrinyi utca 3, H-1051 Budapest, Hungary
| | - Steven A. Forst
- Department of Biological Sciences, University of Wisconsin-Milwaukee, P.O. Box 413, Milwaukee, WI 53201, USA;
| | - Ghazala M. Furgani
- Department of Genetics, Eötvös University, Pázmány Péter Sétány 1/C, H-1117 Budapest, Hungary; (A.M.F.); (K.L.); or (G.M.F.); or (T.V.)
- Department of Plant Protection, Faculty of Agriculture, University of Tripoli, Tripoli P.O. Box 13793, Libya
| | - Levente Karaffa
- Department of Biochemical Engineering, Faculty of Science and Technology, University of Debrecen, Egyetem Tér 1, H-4032 Debrecen, Hungary;
- Institute of Metagenomics, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary
| | - Tibor Vellai
- Department of Genetics, Eötvös University, Pázmány Péter Sétány 1/C, H-1117 Budapest, Hungary; (A.M.F.); (K.L.); or (G.M.F.); or (T.V.)
- MTA-ELTE Genetics Research Group, Pázmány Péter Sétány 1/C, H-1117 Budapest, Hungary
| |
Collapse
|
25
|
Shi G, Kang X, Dong F, Liu Y, Zhu N, Hu Y, Xu H, Lao X, Zheng H. DRAMP 3.0: an enhanced comprehensive data repository of antimicrobial peptides. Nucleic Acids Res 2021; 50:D488-D496. [PMID: 34390348 PMCID: PMC8728287 DOI: 10.1093/nar/gkab651] [Citation(s) in RCA: 141] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/06/2021] [Accepted: 07/21/2021] [Indexed: 01/14/2023] Open
Abstract
Stapled antimicrobial peptides are an emerging class of artificial cyclic peptide molecules which have antimicrobial activity and potent structure stability. We previously published the Data Repository of Antimicrobial Peptides (DRAMP) as a manually annotated and open-access database of antimicrobial peptides (AMPs). In the update of version 3.0, special emphasis was placed on the new development of stapled AMPs, and a subclass of specific AMPs was added to store information on these special chemically modified AMPs. To help design low toxicity AMPs, we also added the cytotoxicity property of AMPs, as well as the expansion of newly discovered AMP data. At present, DRAMP has been expanded and contains 22259 entries (2360 newly added), consisting of 5891 general entries, 16110 patent entries, 77 clinical entries and 181 stapled AMPs. A total of 263 entries have predicted structures, and more than 300 general entries have links to experimentally determined structures in the Protein Data Bank. The update also covers new annotations, statistics, categories, functions and download links. DRAMP is available online at http://dramp.cpu-bioinfor.org/.
Collapse
Affiliation(s)
- Guobang Shi
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211100, P.R. China
| | - Xinyue Kang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211100, P.R. China
| | - Fanyi Dong
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211100, P.R. China
| | - Yanchao Liu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211100, P.R. China
| | - Ning Zhu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211100, P.R. China
| | - Yuxuan Hu
- School of International Pharmaceutical Business, China Pharmaceutical University, Nanjing 211100, P.R. China
| | - Hanmei Xu
- The Engineering Research Centre of Peptide Drug Discovery and Development, China Pharmaceutical University, Nanjing 211100, P.R. China
| | - Xingzhen Lao
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211100, P.R. China
| | - Heng Zheng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211100, P.R. China
| |
Collapse
|
26
|
Liu J, Li H, Li H, Fang S, Shi J, Chen Y, Zhong R, Liu S, Lin S. Rational Design of Dipicolylamine-Containing Carbazole Amphiphiles Combined with Zn 2+ as Potent Broad-Spectrum Antibacterial Agents with a Membrane-Disruptive Mechanism. J Med Chem 2021; 64:10429-10444. [PMID: 34235929 DOI: 10.1021/acs.jmedchem.1c00858] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Antibiotic resistance has become one of the most urgently important problems facing healthcare providers. A novel series of dipicolylamine-containing carbazole amphiphiles with strong Zn2+ chelating ability were synthesized, biomimicking cationic antimicrobial peptides. Effective broad-spectrum 16 combined with 12.5 μg/mL Zn2+ was identified as the most promising antimicrobial candidate. 16 combined with 12.5 μg/mL Zn2+ exhibited excellent antimicrobial activity against both Gram-positive and Gram-negative bacteria (MICs = 0.78-3.125 μg/mL), weak hemolytic activity, and low cytotoxicity. Time-kill kinetics and mechanism studies revealed 16 combined with 12.5 μg/mL Zn2+ had rapid bacterial killing properties, as evidenced by disruption of the integrity of bacterial cell membranes, effectively preventing bacterial resistance development. Importantly, 16 combined with 12.5 μg/mL Zn2+ showed excellent in vivo efficacy in a murine keratitis model caused by Staphylococcus aureus ATCC29213 or Pseudomonas aeruginosa ATCC9027. Therefore, 16 combined with 12.5 μg/mL Zn2+ could be a promising candidate for treating bacterial infections.
Collapse
Affiliation(s)
- Jiayong Liu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P.R. China
| | - Hongxia Li
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P.R. China
| | - Haizhou Li
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P.R. China
| | - Shanfang Fang
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P.R. China
| | - Jinguo Shi
- Department of Medicinal Chemistry, School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou 510006, P. R. China
| | - Yongzhi Chen
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P.R. China
| | - Rongcui Zhong
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P.R. China
| | - Shouping Liu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P.R. China
| | - Shuimu Lin
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, P.R. China
| |
Collapse
|
27
|
BING, a novel antimicrobial peptide isolated from Japanese medaka plasma, targets bacterial envelope stress response by suppressing cpxR expression. Sci Rep 2021; 11:12219. [PMID: 34108601 PMCID: PMC8190156 DOI: 10.1038/s41598-021-91765-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 05/25/2021] [Indexed: 12/16/2022] Open
Abstract
Antimicrobial peptides (AMPs) have emerged as a promising alternative to small molecule antibiotics. Although AMPs have previously been isolated in many organisms, efforts on the systematic identification of AMPs in fish have been lagging. Here, we collected peptides from the plasma of medaka (Oryzias latipes) fish. By using mass spectrometry, 6399 unique sequences were identified from the isolated peptides, among which 430 peptides were bioinformatically predicted to be potential AMPs. One of them, a thermostable 13-residue peptide named BING, shows a broad-spectrum toxicity against pathogenic bacteria including drug-resistant strains, at concentrations that presented relatively low toxicity to mammalian cell lines and medaka. Proteomic analysis indicated that BING treatment induced a deregulation of periplasmic peptidyl-prolyl isomerases in gram-negative bacteria. We observed that BING reduced the RNA level of cpxR, an upstream regulator of envelope stress responses. cpxR is known to play a crucial role in the development of antimicrobial resistance, including the regulation of genes involved in drug efflux. BING downregulated the expression of efflux pump components mexB, mexY and oprM in P. aeruginosa and significantly synergised the toxicity of antibiotics towards these bacteria. In addition, exposure to sublethal doses of BING delayed the development of antibiotic resistance. To our knowledge, BING is the first AMP shown to suppress cpxR expression in Gram-negative bacteria. This discovery highlights the cpxR pathway as a potential antimicrobial target.
Collapse
|
28
|
Jangir PK, Ogunlana L, MacLean RC. Evolutionary constraints on the acquisition of antimicrobial peptide resistance in bacterial pathogens. Trends Microbiol 2021; 29:1058-1061. [PMID: 33836929 DOI: 10.1016/j.tim.2021.03.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 03/11/2021] [Accepted: 03/12/2021] [Indexed: 10/21/2022]
Abstract
Antimicrobial peptides (AMPs) offer a potential solution to the antibiotic resistance crisis. Recent studies have revealed important evolutionary constraints on the evolution and horizontal spread of AMP resistance in bacteria. Here, we summarize these advances and highlight their importance for therapeutic development of AMPs.
Collapse
Affiliation(s)
- Pramod K Jangir
- Department of Zoology, University of Oxford, Oxford, OX1 3PS, UK.
| | - Lois Ogunlana
- Department of Zoology, University of Oxford, Oxford, OX1 3PS, UK
| | - R Craig MacLean
- Department of Zoology, University of Oxford, Oxford, OX1 3PS, UK
| |
Collapse
|
29
|
Affiliation(s)
- Annapaula Correia
- Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK. .,Department of Veterinary Medicine, University of Cambridge, Cambridge, UK. .,Department of Zoology, University of Oxford, Oxford, UK.
| | - Aaron Weimann
- Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK. .,Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, UK.
| |
Collapse
|
30
|
Zimmermann M, Patil KR, Typas A, Maier L. Towards a mechanistic understanding of reciprocal drug-microbiome interactions. Mol Syst Biol 2021; 17:e10116. [PMID: 33734582 PMCID: PMC7970330 DOI: 10.15252/msb.202010116] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/10/2021] [Accepted: 01/25/2021] [Indexed: 02/06/2023] Open
Abstract
Broad-spectrum antibiotics target multiple gram-positive and gram-negative bacteria, and can collaterally damage the gut microbiota. Yet, our knowledge of the extent of damage, the antibiotic activity spectra, and the resistance mechanisms of gut microbes is sparse. This limits our ability to mitigate microbiome-facilitated spread of antibiotic resistance. In addition to antibiotics, non-antibiotic drugs affect the human microbiome, as shown by metagenomics as well as in vitro studies. Microbiome-drug interactions are bidirectional, as microbes can also modulate drugs. Chemical modifications of antibiotics mostly function as antimicrobial resistance mechanisms, while metabolism of non-antibiotics can also change the drugs' pharmacodynamic, pharmacokinetic, and toxic properties. Recent studies have started to unravel the extensive capacity of gut microbes to metabolize drugs, the mechanisms, and the relevance of such events for drug treatment. These findings raise the question whether and to which degree these reciprocal drug-microbiome interactions will differ across individuals, and how to take them into account in drug discovery and precision medicine. This review describes recent developments in the field and discusses future study areas that will benefit from systems biology approaches to better understand the mechanistic role of the human gut microbiota in drug actions.
Collapse
Affiliation(s)
- Michael Zimmermann
- Structural and Computational Biology UnitEuropean Molecular Biology LaboratoryHeidelbergGermany
| | - Kiran Raosaheb Patil
- Structural and Computational Biology UnitEuropean Molecular Biology LaboratoryHeidelbergGermany
- The Medical Research Council Toxicology UnitUniversity of CambridgeCambridgeUK
| | - Athanasios Typas
- Structural and Computational Biology UnitEuropean Molecular Biology LaboratoryHeidelbergGermany
- Genome Biology UnitEuropean Molecular Biology LaboratoryHeidelbergGermany
| | - Lisa Maier
- Interfaculty Institute of Microbiology and Infection MedicineUniversity of TübingenTübingenGermany
- Cluster of Excellence ‘Controlling Microbes to Fight Infections’University of TübingenTübingenGermany
| |
Collapse
|
31
|
Hernando-Amado S, Coque TM, Baquero F, Martínez JL. Antibiotic Resistance: Moving From Individual Health Norms to Social Norms in One Health and Global Health. Front Microbiol 2020; 11:1914. [PMID: 32983000 PMCID: PMC7483582 DOI: 10.3389/fmicb.2020.01914] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 07/21/2020] [Indexed: 12/16/2022] Open
Abstract
Antibiotic resistance is a problem for human health, and consequently, its study had been traditionally focused toward its impact for the success of treating human infections in individual patients (individual health). Nevertheless, antibiotic-resistant bacteria and antibiotic resistance genes are not confined only to the infected patients. It is now generally accepted that the problem goes beyond humans, hospitals, or long-term facility settings and that it should be considered simultaneously in human-connected animals, farms, food, water, and natural ecosystems. In this regard, the health of humans, animals, and local antibiotic-resistance-polluted environments should influence the health of the whole interconnected local ecosystem (One Health). In addition, antibiotic resistance is also a global problem; any resistant microorganism (and its antibiotic resistance genes) could be distributed worldwide. Consequently, antibiotic resistance is a pandemic that requires Global Health solutions. Social norms, imposing individual and group behavior that favor global human health and in accordance with the increasingly collective awareness of the lack of human alienation from nature, will positively influence these solutions. In this regard, the problem of antibiotic resistance should be understood within the framework of socioeconomic and ecological efforts to ensure the sustainability of human development and the associated human-natural ecosystem interactions.
Collapse
Affiliation(s)
- Sara Hernando-Amado
- Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Teresa M. Coque
- Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Centro de Investigación Biomédica en Red Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Fernando Baquero
- Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS) and Centro de Investigación Biomédica en Red Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - José L. Martínez
- Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| |
Collapse
|
32
|
Ting DSJ, Beuerman RW, Dua HS, Lakshminarayanan R, Mohammed I. Strategies in Translating the Therapeutic Potentials of Host Defense Peptides. Front Immunol 2020; 11:983. [PMID: 32528474 PMCID: PMC7256188 DOI: 10.3389/fimmu.2020.00983] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 04/27/2020] [Indexed: 01/13/2023] Open
Abstract
The golden era of antibiotics, heralded by the discovery of penicillin, has long been challenged by the emergence of antimicrobial resistance (AMR). Host defense peptides (HDPs), previously known as antimicrobial peptides, are emerging as a group of promising antimicrobial candidates for combatting AMR due to their rapid and unique antimicrobial action. Decades of research have advanced our understanding of the relationship between the physicochemical properties of HDPs and their underlying antimicrobial and non-antimicrobial functions, including immunomodulatory, anti-biofilm, and wound healing properties. However, the mission of translating novel HDP-derived molecules from bench to bedside has yet to be fully accomplished, primarily attributed to their intricate structure-activity relationship, toxicity, instability in host and microbial environment, lack of correlation between in vitro and in vivo efficacies, and dwindling interest from large pharmaceutical companies. Based on our previous experience and the expanding knowledge gleaned from the literature, this review aims to summarize the novel strategies that have been employed to enhance the antimicrobial efficacy, proteolytic stability, and cell selectivity, which are all crucial factors for bench-to-bedside translation of HDP-based treatment. Strategies such as residues substitution with natural and/or unnatural amino acids, hybridization, L-to-D heterochiral isomerization, C- and N-terminal modification, cyclization, incorporation with nanoparticles, and "smart design" using artificial intelligence technology, will be discussed. We also provide an overview of HDP-based treatment that are currently in the development pipeline.
Collapse
Affiliation(s)
- Darren Shu Jeng Ting
- Larry A. Donoso Laboratory for Eye Research, Academic Ophthalmology, Division of Clinical Neuroscience, School of Medicine, University of Nottingham, Nottingham, United Kingdom.,Department of Ophthalmology, Queen's Medical Centre, Nottingham, United Kingdom.,Anti-infectives Research Group, Singapore Eye Research Institute, The Academia, Singapore, Singapore
| | - Roger W Beuerman
- Anti-infectives Research Group, Singapore Eye Research Institute, The Academia, Singapore, Singapore
| | - Harminder S Dua
- Larry A. Donoso Laboratory for Eye Research, Academic Ophthalmology, Division of Clinical Neuroscience, School of Medicine, University of Nottingham, Nottingham, United Kingdom.,Department of Ophthalmology, Queen's Medical Centre, Nottingham, United Kingdom
| | - Rajamani Lakshminarayanan
- Anti-infectives Research Group, Singapore Eye Research Institute, The Academia, Singapore, Singapore
| | - Imran Mohammed
- Larry A. Donoso Laboratory for Eye Research, Academic Ophthalmology, Division of Clinical Neuroscience, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|