51
|
Berryhill BA, Levin BR. Semantics Count in the Description of the Interactions Between Bacteria and Bacteriophage. PHAGE (NEW ROCHELLE, N.Y.) 2025; 6:3-4. [PMID: 40291344 PMCID: PMC12022465 DOI: 10.1089/phage.2024.0063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Affiliation(s)
- Brandon A. Berryhill
- Department of Biology, Emory University, Atlanta, Georgia, USA
- Program in Microbiology and Molecular Genetics, Graduate Division of Biological and Biomedical Sciences, Laney Graduate School, Emory University, Atlanta, Georgia, USA
| | - Bruce R. Levin
- Department of Biology, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
52
|
Yang CS, Shie MY, Huang SW, Wang YC, Hou MH, Chen CJ, Chen Y. Structural insights into signaling promiscuity of the CBASS anti-phage defense system from a radiation-resistant bacterium. Int J Biol Macromol 2025; 295:139534. [PMID: 39761885 DOI: 10.1016/j.ijbiomac.2025.139534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 01/13/2025]
Abstract
Radiation-resistant bacteria are of great application potential in various fields, including bioindustry and bioremediation of radioactive waste. However, how radiation-resistant bacteria combat against invading phages is seldom addressed. Here, we present a series of crystal structures of a sensor and an effector of the cyclic oligonucleotide-based anti-phage signaling system (CBASS) from a radioresistant bacterium Deinococcus wulumuqiensis. We found that the sensor CD-NTase enzyme, DwCdnB, can bind all four ribonucleotides and synthesize a variety of cyclic di-nucleotides, including the novel second messenger 3'3'-cyclic di-CMP. Crystal structures of DwCdnB in complex with ATP and dATP provide structural explanations for specific recognition of ribonucleotides via metal coordination with ribose 2'-OH. Crystal structures of DwCdnB in complex with purine and/or pyrimidine nucleotides in the presence of Mg2+ revealed similar binding modes; however, in the presence of Mn2+, the UTP/CTP rotates and flips into the donor pocket and make extensive contacts with additional five residues, suggesting essential role of Mn2+ for catalytic production of cyclic di-pyrimidines. Finally, structural analysis of the downstream effector DwCap5 further provides a structural explanation for its non-specific recognition of a broad range of cyclic di-nucleotides. In sum, this work provides key structural insights into the immune mechanisms of radioresistant bacteria.
Collapse
Affiliation(s)
- Chia-Shin Yang
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung 40227, Taiwan
| | - Ming-You Shie
- Research & Development Center for x-Dimensional Extracellular Vesicles, China Medical University Hospital, Taichung 40447, Taiwan; Department of Biomedical Engineering, China Medical University, Taichung 40447, Taiwan; Department of Bioinformatics and Medical Engineering, Asia University, Taichung 41354, Taiwan
| | - Shi-Wei Huang
- Translational Cell Therapy Center, Department of Medical Research, China Medical University Hospital, Taichung 40447, Taiwan; Institute of Biomedical Science, National Chung-Hsing University, Taichung 40227, Taiwan; Institute of New Drug Development, China Medical University, Taichung 40447, Taiwan
| | - Yu-Chuan Wang
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung 40227, Taiwan
| | - Mei-Hui Hou
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung 40227, Taiwan
| | - Chao-Jung Chen
- Graduate Institute of Integrated Medicine, China Medical University, Taichung 40447, Taiwan; Proteomics Core Laboratory, Department of Medical Research, China Medical University Hospital, Taichung 40447, Taiwan.
| | - Yeh Chen
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung 40227, Taiwan.
| |
Collapse
|
53
|
Wein T, Millman A, Lange K, Yirmiya E, Hadary R, Garb J, Melamed S, Amitai G, Dym O, Steinruecke F, Hill AB, Kranzusch PJ, Sorek R. CARD domains mediate anti-phage defence in bacterial gasdermin systems. Nature 2025; 639:727-734. [PMID: 39880956 DOI: 10.1038/s41586-024-08498-3] [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: 05/29/2023] [Accepted: 12/05/2024] [Indexed: 01/31/2025]
Abstract
Caspase recruitment domains (CARDs) and pyrin domains are important facilitators of inflammasome activity and pyroptosis1. Following pathogen recognition by nucleotide binding-domain, leucine-rich, repeat-containing (NLR) proteins, CARDs recruit and activate caspases, which, in turn, activate gasdermin pore-forming proteins to induce pyroptotic cell death2. Here we show that CARD domains are present in defence systems that protect bacteria against phage. The bacterial CARD domain is essential for protease-mediated activation of certain bacterial gasdermins, which promote cell death once phage infection is recognized. We further show that multiple anti-phage defence systems use CARD domains to activate a variety of cell death effectors, and that CARD domains mediate protein-protein interactions in these systems. We find that these systems are triggered by a conserved immune-evasion protein used by phages to overcome the bacterial defence system RexAB3, demonstrating that phage proteins inhibiting one defence system can activate another. Our results suggest that CARD domains represent an ancient component of innate immune systems conserved from bacteria to humans, and that CARD-dependent activation of gasdermins is shared in organisms across the tree of life.
Collapse
Affiliation(s)
- Tanita Wein
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Adi Millman
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Katharina Lange
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Erez Yirmiya
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Romi Hadary
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Jeremy Garb
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Sarah Melamed
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Gil Amitai
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Orly Dym
- Department of Life Science Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | | | - Aidan B Hill
- Department of Microbiology, Harvard Medical School, Boston, Ma, USA
- Deparment of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Philip J Kranzusch
- Department of Microbiology, Harvard Medical School, Boston, Ma, USA.
- Deparment of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Parker Institute for Cancer Immunotherapy at Dana-Farber Cancer Institute, Boston, MA, USA.
| | - Rotem Sorek
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
54
|
Zhang Y, Yang S, Zheng X, Tan X. Cyanobacterial type I CRISPR-Cas systems: distribution, mechanisms, and genome editing applications. Front Bioeng Biotechnol 2025; 13:1552030. [PMID: 40084131 PMCID: PMC11903412 DOI: 10.3389/fbioe.2025.1552030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Accepted: 02/10/2025] [Indexed: 03/16/2025] Open
Abstract
Cyanobacteria, renowned for their photosynthetic capabilities, serve as efficient microbial chassis capable of converting carbon dioxide into a spectrum of bio-chemicals. However, conventional genetic manipulation strategies have proven incompatible with the precise and systematic modifications required in the field of cyanobacterial synthetic biology. Here, we present an in-depth analysis of endogenous CRISPR-Cas systems within cyanobacterial genomes, with a particular focus on the Type I systems, which are the most widely distributed. We provide a comprehensive summary of the reported DNA defense mechanisms mediated by cyanobacterial Type I CRISPR-Cas systems and their current applications in genome editing. Furthermore, we offer insights into the future applications of these systems in the context of cyanobacterial genome editing, underscoring their potential to revolutionize synthetic biology approaches.
Collapse
Affiliation(s)
- Yongjiu Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei, China
| | - Shuxiao Yang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei, China
| | - Xianliang Zheng
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei, China
- AngelYeast Co., Ltd., Yichang, Hubei, China
- National Key Laboratory of Agricultural Microbiology, AngelYeast Co., Ltd., Yichang, Hubei, China
| | - Xiaoming Tan
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, Hubei, China
| |
Collapse
|
55
|
Piper KR, Souza SSR, Ikhimiukor OO, Workman AA, Martin IW, Andam CP. Lineage-specific variation in frequency and hotspots of recombination in invasive Escherichia coli. BMC Genomics 2025; 26:190. [PMID: 39994515 PMCID: PMC11853335 DOI: 10.1186/s12864-025-11367-6] [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: 09/12/2024] [Accepted: 02/14/2025] [Indexed: 02/26/2025] Open
Abstract
BACKGROUND The opportunistic bacterium Escherichia coli can invade normally sterile sites in the human body, potentially leading to life-threatening organ dysfunction and even death. However, our understanding of the evolutionary processes that shape its genetic diversity in this sterile environment remains limited. Here, we aim to quantify the frequency and characteristics of homologous recombination in E. coli from bloodstream infections. RESULTS Analysis of 557 short-read genome sequences revealed that the propensity to exchange DNA by homologous recombination varies within a distinct population (bloodstream) at narrow geographic (Dartmouth Hitchcock Medical Center, New Hampshire, USA) and temporal (years 2016 - 2022) scope. We identified the four largest monophyletic sequence clusters in the core genome phylogeny that are represented by prominent sequence types (ST): BAPS1 (mainly ST95), BAPS4 (mainly ST73), BAPS10 (mainly ST131), BAPS14 (mainly ST58). We show that the four dominant clusters vary in different characteristics of recombination: number of single nucleotide polymorphisms due to recombination, number of recombination blocks, cumulative bases in recombination blocks, ratio of probabilities that a given site was altered through recombination and mutation (r/m), and ratio of rates at which recombination and mutation occurred (ρ/θ). Each sequence cluster contains a unique set of antimicrobial resistance (AMR) and virulence genes that have experienced recombination. Common among the four sequence clusters were the recombined virulence genes with functions associated with the Curli secretion channel (csgG) and ferric enterobactin transport (entEF, fepEG). We did not identify any one recombined AMR gene that was present in all four sequence clusters. However, AMR genes mdtABC, baeSR, emrKY and tolC had experienced recombination in sequence clusters BAPS4, BAPS10, and BAPS14. These differences lie in part on the contributions of vertically inherited ancestral recombination and contemporary branch-specific recombination, with some genomes having relatively higher proportions of recombined DNA. CONCLUSIONS Our results highlight the variation in the propensity to exchange DNA via homologous recombination within a distinct population at narrow geographic and temporal ranges. Understanding the sources of the genetic variation in invasive E. coli will help inform the implementation of effective strategies to reduce the burden of disease and AMR.
Collapse
Affiliation(s)
- Kathryn R Piper
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY, USA
| | - Stephanie S R Souza
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY, USA
| | - Odion O Ikhimiukor
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY, USA
| | - Adrienne A Workman
- Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center and Dartmouth College Geisel School of Medicine, Lebanon, NH, USA
| | - Isabella W Martin
- Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center and Dartmouth College Geisel School of Medicine, Lebanon, NH, USA.
| | - Cheryl P Andam
- Department of Biological Sciences, University at Albany, State University of New York, Albany, NY, USA.
| |
Collapse
|
56
|
Zhu K, Shang K, Wang L, Yu X, Hua L, Zhang W, Qin B, Wang J, Gao X, Zhu H, Cui S. Activation of the bacterial defense-associated sirtuin system. Commun Biol 2025; 8:297. [PMID: 39994439 PMCID: PMC11850899 DOI: 10.1038/s42003-025-07743-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 02/14/2025] [Indexed: 02/26/2025] Open
Abstract
The NADase activity of the defense-associated sirtuins (DSRs) is activated by the phage tail tube protein (TTP). Herein, we report cryo-EM structures of a free-state Bacillus subtilis DSR2 tetramer and a fragment of the tetramer, a phage SPR tail tube, and two DSR2-TTP complexes. DSR2 contains an N-terminal SIR2 domain, a middle domain (MID) and a C-terminal domain (CTD). The DSR2 CTD harbors the α-solenoid tandem-repeats like the HEAT-repeat proteins. DSR2 assembles into a tetramer with four SIR2 clustered at the center, and two intertwined MID-CTD chains flank the SIR2 core. SPR TTPs self-assemble into a tube-like complex. Upon DSR2 binding, the D1 domain of SPR TTP is captured between the HEAT-repeats domains of DSR2, which conflicts with TTPs self-assembly. Binding of TTPs induces conformational changes in DSR2 tetramer, resulting in increase of the NAD+ pocket volume in SIR2, thus activates the NADase activity and leads to cellular NAD+ depletion.
Collapse
Affiliation(s)
- Kaixiang Zhu
- NHC Key Laboratory of Systems Biology of Pathogens, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of Pathogen Infection Prevention and Control (Ministry of Education), National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Kun Shang
- Yanan medical college of Yanan university, Yanan, China
- Beijing National Laboratory for Condensed Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, China
| | - Linyue Wang
- NHC Key Laboratory of Systems Biology of Pathogens, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of Pathogen Infection Prevention and Control (Ministry of Education), National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xia Yu
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory for Drug-resistant Tuberculosis Research Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, China
| | - Lei Hua
- Yanan medical college of Yanan university, Yanan, China
| | - Weihe Zhang
- National Clinical Laboratory on Tuberculosis, Beijing Key Laboratory for Drug-resistant Tuberculosis Research Beijing Chest Hospital, Capital Medical University, Beijing Tuberculosis and Thoracic Tumor Institute, Beijing, China
| | - Bo Qin
- NHC Key Laboratory of Systems Biology of Pathogens, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Key Laboratory of Pathogen Infection Prevention and Control (Ministry of Education), National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jia Wang
- Beijing Advanced Innovation Center for Structural Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing, China
| | - Xiaopan Gao
- NHC Key Laboratory of Systems Biology of Pathogens, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
- Key Laboratory of Pathogen Infection Prevention and Control (Ministry of Education), National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
- State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Hongtao Zhu
- Beijing National Laboratory for Condensed Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Songshan Lake Materials Laboratory, Dongguan, China.
| | - Sheng Cui
- NHC Key Laboratory of Systems Biology of Pathogens, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
- Key Laboratory of Pathogen Infection Prevention and Control (Ministry of Education), National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
- State Key Laboratory of Respiratory Health and Multimorbidity, National Institute of Pathogen Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
57
|
Debiasi-Anders G, Qiao C, Salim A, Li N, Mir-Sanchis I. Phage parasites targeting phage homologous recombinases provide antiviral immunity. Nat Commun 2025; 16:1889. [PMID: 39987160 PMCID: PMC11846896 DOI: 10.1038/s41467-025-57156-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 02/11/2025] [Indexed: 02/24/2025] Open
Abstract
Bacteria often carry multiple genes encoding anti-phage defense systems, clustered in defense islands and phage satellites. Various unrelated anti-phage defense systems target phage-encoded homologous recombinases (HRs) through unclear mechanisms. Here, we show that the phage satellite SaPI2, which does not encode orthodox anti-phage defense systems, provides antiviral immunity mediated by Stl2, the SaPI2-encoded transcriptional repressor. Stl2 targets and inhibits phage-encoded HRs, including Sak and Sak4, two HRs from the Rad52-like and Rad51-like superfamilies. Remarkably, apo Stl2 forms a collar of dimers oligomerizing as closed rings and as filaments, mimicking the quaternary structure of its targets. Stl2 decorates both Sak rings and Sak4 filaments. The oligomerization of Stl2 as a collar of dimers is necessary for its inhibitory activity both in vitro and in vivo. Our results shed light on the mechanisms underlying antiviral immunity against phages carrying divergent HRs.
Collapse
Affiliation(s)
- Gianluca Debiasi-Anders
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå, Sweden
| | - Cuncun Qiao
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå, Sweden
| | - Amrita Salim
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå, Sweden
| | - Na Li
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå, Sweden
| | - Ignacio Mir-Sanchis
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå, Sweden.
- Wallenberg Centre for Molecular Medicine, Umeå, Sweden.
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri i Reixac 10-12, Barcelona, Spain.
| |
Collapse
|
58
|
Zang Z, Duncan OK, Sabonis D, Shi Y, Miraj G, Fedorova I, Le S, Deng J, Zhu Y, Cai Y, Zhang C, Arya G, Duerkop BA, Liang H, Bondy-Denomy J, Ve T, Tamulaitiene G, Gerdt JP. Chemical inhibition of a bacterial immune system 1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.20.638879. [PMID: 40027640 PMCID: PMC11870472 DOI: 10.1101/2025.02.20.638879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
The rise of antibiotic resistance motivates a revived interest in phage therapy. However, bacteria possess dozens of anti-bacteriophage immune systems that confer resistance to therapeutic phages. Chemical inhibitors of these anti-phage immune systems could be employed as adjuvants to overcome resistance in phage-based therapies. Here, we report that anti-phage systems can be selectively inhibited by small molecules, thereby sensitizing phage-resistant bacteria to phages. We discovered a class of chemical inhibitors that inhibit the type II Thoeris anti-phage immune system. These inhibitors block the biosynthesis of a histidine-ADPR intracellular 'alarm' signal by ThsB and prevent ThsA from arresting phage replication. These inhibitors promiscuously inhibit type II Thoeris systems from diverse bacteria-including antibiotic-resistant pathogens. Chemical inhibition of the Thoeris defense improved the efficacy of a model phage therapy against a phage-resistant strain of P. aeruginosa in a mouse infection, suggesting a therapeutic potential. Furthermore, these inhibitors may be employed as chemical tools to dissect the importance of the Thoeris system for phage defense in natural microbial communities.
Collapse
Affiliation(s)
- Zhiyu Zang
- Department of Chemistry, Indiana University, Bloomington, IN 47405, USA
| | - Olivia K. Duncan
- Department of Chemistry, Indiana University, Bloomington, IN 47405, USA
| | - Dziugas Sabonis
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Yun Shi
- Institute for Biomedicine and Glycomics, Griffith University, Gold Coast, QLD, Australia
| | - Gause Miraj
- Institute for Biomedicine and Glycomics, Griffith University, Gold Coast, QLD, Australia
| | - Iana Fedorova
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Shuai Le
- Department of Microbiology, College of Basic Medical Sciences, Key Laboratory of Microbial Engineering Committee in Chongqing, Army Medical University, Chongqing, 400038, China
| | - Jun Deng
- Department of Microbiology, College of Basic Medical Sciences, Key Laboratory of Microbial Engineering Committee in Chongqing, Army Medical University, Chongqing, 400038, China
| | - Yuhao Zhu
- Department of Microbiology, College of Basic Medical Sciences, Key Laboratory of Microbial Engineering Committee in Chongqing, Army Medical University, Chongqing, 400038, China
| | - Yanyao Cai
- Department of Chemistry, Indiana University, Bloomington, IN 47405, USA
| | - Chengqian Zhang
- Department of Chemistry, Indiana University, Bloomington, IN 47405, USA
| | - Garima Arya
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, School of Medicine, Aurora, CO 80045, USA
| | - Breck A. Duerkop
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, School of Medicine, Aurora, CO 80045, USA
| | - Haihua Liang
- College of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Joseph Bondy-Denomy
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Thomas Ve
- Institute for Biomedicine and Glycomics, Griffith University, Gold Coast, QLD, Australia
| | - Giedre Tamulaitiene
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Joseph P. Gerdt
- Department of Chemistry, Indiana University, Bloomington, IN 47405, USA
| |
Collapse
|
59
|
Li Y, Li Z, Yan P, Hua C, Kong J, Wu W, Cui Y, Duan Y, Li S, Li G, Ji S, Chen Y, Zhao Y, Yang P, Hu C, Lu M, Chen M, Xiao Y. Antiviral signaling of a type III CRISPR-associated deaminase. Science 2025; 387:eadr0393. [PMID: 39666823 DOI: 10.1126/science.adr0393] [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: 06/12/2024] [Accepted: 12/01/2024] [Indexed: 12/14/2024]
Abstract
Prokaryotes have evolved diverse defense strategies against viral infection, including foreign nucleic acid degradation by CRISPR-Cas systems and DNA and RNA synthesis inhibition through nucleotide pool depletion. Here, we report an antiviral mechanism of type III CRISPR-Cas-regulated adenosine triphosphate (ATP) depletion in which ATP is converted into inosine triphosphate (ITP) by CRISPR-Cas-associated adenosine deaminase (CAAD) upon activation by either cA4 or cA6, followed by hydrolysis into inosine monophosphate (IMP) by Nudix hydrolase, ultimately resulting in cell growth arrest. The cryo-electron microscopy structures of CAAD in its apo and activated forms, together with biochemical evidence, revealed how cA4 or cA6 binds to the CRISPR-associated Rossmann fold (CARF) domain and abrogates CAAD autoinhibition, inducing substantial conformational changes that reshape the structure of CAAD and induce its deaminase activity. Our results reveal the mechanism of a CRISPR-Cas-regulated ATP depletion antiviral strategy.
Collapse
Affiliation(s)
- Yutao Li
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Zhaoxing Li
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- Chongqing Innovation Institute of China Pharmaceutical University, Chongqing, China
| | - Purui Yan
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Chenyang Hua
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- Department of Biochemistry, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Jianping Kong
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Wanqian Wu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- Department of Biochemistry, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yurong Cui
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- Department of Biochemistry, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yan Duan
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Shunxiang Li
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, China
| | - Guanglei Li
- College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing, China
| | - Shunli Ji
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yijun Chen
- Department of Biochemistry, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Yucheng Zhao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Peng Yang
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- Chongqing Innovation Institute of China Pharmaceutical University, Chongqing, China
| | - Chunyi Hu
- Precision Medicine Translational Research Programme (TRP), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Meiling Lu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- Chongqing Innovation Institute of China Pharmaceutical University, Chongqing, China
- Department of Biochemistry, School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Meirong Chen
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- Chongqing Innovation Institute of China Pharmaceutical University, Chongqing, China
| | - Yibei Xiao
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
- Chongqing Innovation Institute of China Pharmaceutical University, Chongqing, China
| |
Collapse
|
60
|
Liu Y, Botelho J, Iranzo J. Timescale and genetic linkage explain the variable impact of defense systems on horizontal gene transfer. Genome Res 2025; 35:268-278. [PMID: 39794121 PMCID: PMC11874982 DOI: 10.1101/gr.279300.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 01/06/2025] [Indexed: 01/13/2025]
Abstract
Prokaryotes have evolved a wide repertoire of defense systems to prevent invasion by mobile genetic elements (MGEs). However, because MGEs are vehicles for the exchange of beneficial accessory genes, defense systems could consequently impede rapid adaptation in microbial populations. Here, we study how defense systems impact horizontal gene transfer (HGT) in the short term and long term. By combining comparative genomics and phylogeny-aware statistical methods, we quantify the association between the presence of seven widespread defense systems and the abundance of MGEs in the genomes of 196 bacterial and one archaeal species. We also calculate the differences in the rates of gene gain and loss between lineages that possess and lack each defense system. Our results show that the impact of defense systems on HGT is highly taxon and system dependent and, in most cases, not statistically significant. Timescale analysis reveals that defense systems must persist in a lineage for a relatively long time to exert an appreciable negative impact on HGT. In contrast, for shorter evolutionary timescales, frequent coacquisition of MGEs and defense systems results in a net positive association of the latter with HGT. Given the high turnover rates experienced by defense systems, we propose that the inhibitory effect of most defense systems on HGT is masked by their strong linkage with MGEs. These findings help explain the contradictory conclusions of previous research by pointing at mobility and within-host retention times as key factors that determine the impact of defense systems on genome plasticity.
Collapse
Affiliation(s)
- Yang Liu
- Centro de Biotecnología y Genómica de Plantas, Universidad Politécnica de Madrid (UPM) - Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), 28223, Madrid, Spain
| | - João Botelho
- Centro de Biotecnología y Genómica de Plantas, Universidad Politécnica de Madrid (UPM) - Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), 28223, Madrid, Spain
| | - Jaime Iranzo
- Centro de Astrobiología (CAB), CSIC-INTA, 28850, Madrid, Spain;
- Institute for Biocomputation and Physics of Complex Systems (BIFI), University of Zaragoza, 50018, Zaragoza, Spain
| |
Collapse
|
61
|
Guliy OI, Evstigneeva SS. Bacteria- and Phage-Derived Proteins in Phage Infection. FRONT BIOSCI-LANDMRK 2025; 30:24478. [PMID: 40018916 DOI: 10.31083/fbl24478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/04/2024] [Accepted: 08/07/2024] [Indexed: 03/01/2025]
Abstract
Phages have exerted severe evolutionary pressure on prokaryotes over billions of years, resulting in major rearrangements. Without every enzyme involved in the phage-bacterium interaction being examined; bacteriophages cannot be used in practical applications. Numerous studies conducted in the past few years have uncovered a huge variety of bacterial antiphage defense systems; nevertheless, the mechanisms of most of these systems are not fully understood. Understanding the interactions between bacteriophage and bacterial proteins is important for efficient host cell infection. Phage proteins involved in these bacteriophage-host interactions often arise immediately after infection. Here, we review the main groups of phage enzymes involved in the first stage of viral infection and responsible for the degradation of the bacterial membrane. These include polysaccharide depolymerases (endosialidases, endorhamnosidases, alginate lyases, and hyaluronate lyases), and peptidoglycan hydrolases (ectolysins and endolysins). Host target proteins are inhibited, activated, or functionally redirected by the phage protein. These interactions determine the phage infection of bacteria. Proteins of interest are holins, endolysins, and spanins, which are responsible for the release of progeny during the phage lytic cycle. This review describes the main bacterial and phage enzymes involved in phage infection and analyzes the therapeutic potential of bacteriophage-derived proteins.
Collapse
Affiliation(s)
- Olga I Guliy
- Institute of Biochemistry and Physiology of Plants and Microorganisms, Saratov Scientific Centre of the Russian Academy of Sciences (IBPPM RAS), 410049 Saratov, Russia
| | - Stella S Evstigneeva
- Institute of Biochemistry and Physiology of Plants and Microorganisms, Saratov Scientific Centre of the Russian Academy of Sciences (IBPPM RAS), 410049 Saratov, Russia
| |
Collapse
|
62
|
A bacterial antiviral system detects and directly fights against infection. Nature 2025:10.1038/d41586-025-00416-5. [PMID: 39939741 DOI: 10.1038/d41586-025-00416-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025]
|
63
|
July E, Gillis A. Antiviral defence arsenal across members of the Bacillus cereus group. Sci Rep 2025; 15:4958. [PMID: 39929895 PMCID: PMC11811056 DOI: 10.1038/s41598-025-86748-8] [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: 10/22/2024] [Accepted: 01/13/2025] [Indexed: 02/13/2025] Open
Abstract
Bacteria co-evolve with bacteriophages to overcome each other's defence arsenal. Bacillus cereus group gathers bacteria of medical and agricultural importance, including foodborne pathogens. So far, few studies have portrayed a complete defence arsenal of microorganisms, and the role of antiviral systems in the Bacillus cereus group has been overlooked. Here, we investigate the repertoire of defence systems in 6354 B. cereus group's genomic assemblies, using bioinformatics tools DefenseFinder and PADLOC. Our analyses provide an overview of the diversity and abundance of defence systems in this group, with 83,738 systems distributed by 2 to 33 within each assembly. Comparing PADLOC and DefenseFinder predictions showed that the most prevalent strategy is Restriction-Modification, but many abortive infection systems also intervene in the group's defence, such as Septu, Gabija and Lamassu. Most defences were encoded on both plasmids and the chromosome, though some tend to have a preferential genomic location. We also studied the defence systems associations within the genomic assemblies. Overall, our results establish a baseline picturing the rich and complex antiviral arsenal encoded by B. cereus group's species and provide clues for studying co-existing strategies displayed by these bacteria to subvert phages and other MGEs invasions.
Collapse
Affiliation(s)
- Elise July
- Laboratory of Food and Environmental Microbiology, Earth and Life Institute, Université Catholique de Louvain, Croix du Sud 2, Box L7.05.12, 1348, Louvain-la-Neuve, Belgium
| | - Annika Gillis
- Laboratory of Food and Environmental Microbiology, Earth and Life Institute, Université Catholique de Louvain, Croix du Sud 2, Box L7.05.12, 1348, Louvain-la-Neuve, Belgium.
| |
Collapse
|
64
|
Loeff L, Walter A, Rosalen GT, Jinek M. DNA end sensing and cleavage by the Shedu anti-phage defense system. Cell 2025; 188:721-733.e17. [PMID: 39742808 DOI: 10.1016/j.cell.2024.11.030] [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: 07/06/2023] [Revised: 07/31/2024] [Accepted: 11/19/2024] [Indexed: 01/04/2025]
Abstract
The detection of molecular patterns associated with invading pathogens is a hallmark of innate immune systems. Prokaryotes deploy sophisticated host defense mechanisms in innate anti-phage immunity. Shedu is a single-component defense system comprising a putative nuclease SduA. Here, we report cryoelectron microscopy (cryo-EM) structures of apo- and double-stranded DNA (dsDNA)-bound tetrameric SduA assemblies, revealing that the N-terminal domains of SduA form a clamp that recognizes free DNA ends. End binding positions the DNA over the PD-(D/E)XK nuclease domain, resulting in dsDNA nicking at a fixed distance from the 5' end. The end-directed DNA nicking activity of Shedu prevents propagation of linear DNA in vivo. Finally, we show that phages escape Shedu immunity by suppressing their recombination-dependent DNA replication pathway. Taken together, these results define the antiviral mechanism of Shedu systems, underlining the paradigm that recognition of pathogen-specific nucleic acid structures is a conserved feature of innate immunity across all domains of life.
Collapse
Affiliation(s)
- Luuk Loeff
- Department of Biochemistry, University of Zurich, Zurich, Switzerland.
| | - Alexander Walter
- Department of Biochemistry, University of Zurich, Zurich, Switzerland
| | | | - Martin Jinek
- Department of Biochemistry, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
65
|
Gu Y, Li H, Deep A, Enustun E, Zhang D, Corbett KD. Bacterial Shedu immune nucleases share a common enzymatic core regulated by diverse sensor domains. Mol Cell 2025; 85:523-536.e6. [PMID: 39742666 PMCID: PMC11805627 DOI: 10.1016/j.molcel.2024.12.004] [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] [Revised: 07/25/2024] [Accepted: 12/05/2024] [Indexed: 01/04/2025]
Abstract
Prokaryotes possess diverse anti-bacteriophage immune systems, including the single-protein Shedu nuclease. Here, we reveal the structural basis for activation of Bacillus cereus Shedu. Two cryoelectron microscopy structures of Shedu show that it switches between inactive and active states through conformational changes affecting active-site architecture, which are controlled by the protein's N-terminal domain (NTD). We find that B. cereus Shedu cleaves near DNA ends with a 3' single-stranded overhang, likely enabling it to specifically degrade the DNA injected by certain bacteriophages. Bioinformatic analysis of Shedu homologs reveals a conserved nuclease domain with remarkably diverse N-terminal regulatory domains: we identify 79 distinct NTD types falling into eight broad classes, including those with predicted nucleic acid binding, enzymatic, and other activities. Together, these data reveal Shedu as a broad family of immune nucleases with a common nuclease core regulated by diverse NTDs that likely respond to a range of signals.
Collapse
Affiliation(s)
- Yajie Gu
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Huan Li
- Department of Biology, Saint Louis University, Saint Louis, MO, USA
| | - Amar Deep
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Eray Enustun
- Department of Molecular Biology, University of California, San Diego, La Jolla, CA, USA
| | - Dapeng Zhang
- Department of Biology, Saint Louis University, Saint Louis, MO, USA.
| | - Kevin D Corbett
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, USA; Department of Molecular Biology, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
66
|
Li G, Cheng Y, Yu J, Zhu Y, Ma H, Zhou Y, Pu Z, Zhu G, Yuan Y, Zhang Z, Zhou X, Tian K, Qiao J, Hu X, Chen XX, Ji Q, Huang X, Ma B, Yao Y. Compact RNA editors with natural miniature Cas13j nucleases. Nat Chem Biol 2025; 21:280-290. [PMID: 39300230 DOI: 10.1038/s41589-024-01729-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 08/15/2024] [Indexed: 09/22/2024]
Abstract
Clustered regularly interspaced short palindromic repeats-Cas13 effectors are used for RNA editing but the adeno-associated virus (AAV) packaging limitations because of their big sizes hinder their therapeutic application. Here we report the identification of the Cas13j family, with LepCas13j (529 aa) and ChiCas13j (424 aa) being the smallest and most highly efficient variants for RNA interference. The miniaturized Cas13j proteins enable the development of compact RNA base editors. Chi-RESCUE-S, by fusing dChiCas13j with hADAR2dd, demonstrates high efficiency and specificity in A-to-G and C-to-U conversions. Importantly, this system is compatible with single-AAV packaging without the need for protein sequence truncation. It successfully corrected pathogenic mutations, such as APOC3D65N and SCN9AR896Q, to the wild-type forms. In addition, we developed an optimized system, Chi-RESCUE-S-mini3, which pioneered efficient in vivo C-to-U RNA editing of PCSK9 in mice through single-AAV delivery, resulting in reduced total cholesterol levels. These results highlight the potential of Cas13j to treat human diseases.
Collapse
Affiliation(s)
- Guo Li
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, China.
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China.
- Xianghu Laboratory, Hangzhou, China.
| | - Yaxian Cheng
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, China
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
| | - Jingwen Yu
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Agricultural Resources and Environment, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, China
| | - Yunfei Zhu
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
| | - Hongru Ma
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, China
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
| | - Yuqiao Zhou
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, China
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
| | - Zhongji Pu
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, China
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
- Xianghu Laboratory, Hangzhou, China
| | - Guanglin Zhu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
| | | | - Ziyue Zhang
- Zhejiang Institute of Tianjin University, Shaoxing, China
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, China
| | - Xinzhi Zhou
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, China
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
| | - Kairen Tian
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
- Zhejiang Institute of Tianjin University, Shaoxing, China
| | - Jianjun Qiao
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
- Zhejiang Institute of Tianjin University, Shaoxing, China
| | - Xiaoxiang Hu
- State Key Laboratory of Animal Biotech Breeding, China Agricultural University, Beijing, China
| | - Xue-Xin Chen
- Institute of Insect Sciences, College of Agriculture and Biotechnology, Zhejiang University, Hangzhou, China.
| | - Quanjiang Ji
- School of Physical Science and Technology, ShanghaiTech University, Shanghai, China
| | | | - Bin Ma
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, China.
- Zhejiang Provincial Key Laboratory of Agricultural Resources and Environment, College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, China.
| | - Yuan Yao
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, China.
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China.
| |
Collapse
|
67
|
Huang D, Liao J, Balcazar JL, Ye M, Wu R, Wang D, Alvarez PJJ, Yu P. Adaptive modification of antiviral defense systems in microbial community under Cr-induced stress. MICROBIOME 2025; 13:34. [PMID: 39891205 PMCID: PMC11786475 DOI: 10.1186/s40168-025-02030-z] [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: 07/02/2024] [Accepted: 01/05/2025] [Indexed: 02/03/2025]
Abstract
BACKGROUND The prokaryotic antiviral defense systems are crucial for mediating prokaryote-virus interactions that influence microbiome functioning and evolutionary dynamics. Despite the prevalence and significance of prokaryotic antiviral defense systems, their responses to abiotic stress and ecological consequences remain poorly understood in soil ecosystems. We established microcosm systems with varying concentrations of hexavalent chromium (Cr(VI)) to investigate the adaptive modifications of prokaryotic antiviral defense systems under abiotic stress. RESULTS Utilizing hybrid metagenomic assembly with long-read and short-read sequencing, we discovered that antiviral defense systems were more diverse and prevalent in heavily polluted soils, which was corroborated by meta-analyses of public datasets from various heavy metal-contaminated sites. As the Cr(VI) concentration increased, prokaryotes with defense systems favoring prokaryote-virus mutualism gradually supplanted those with defense systems incurring high adaptive costs. Additionally, as Cr(VI) concentrations increased, enriched antiviral defense systems exhibited synchronization with microbial heavy metal resistance genes. Furthermore, the proportion of antiviral defense systems carried by mobile genetic elements (MGEs), including plasmids and viruses, increased by approximately 43% and 39%, respectively, with rising Cr concentrations. This trend is conducive to strengthening the dissemination and sharing of defense resources within microbial communities. CONCLUSIONS Overall, our study reveals the adaptive modification of prokaryotic antiviral defense systems in soil ecosystems under abiotic stress, as well as their positive contributions to establishing prokaryote-virus mutualism and the evolution of microbial heavy metal resistance. These findings advance our understanding of microbial adaptation in stressful environments and may inspire novel approaches for microbiome manipulation and bioremediation. Video Abstract.
Collapse
Affiliation(s)
- Dan Huang
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jingqiu Liao
- Department of Civil and Environmental Engineering, Virginia Tech, Blacksburg, VA, 24060, USA
| | | | - Mao Ye
- Key Laboratory of Soil Environment and Pollution Remediation, Institute of Soil Science, Chinese Academy of Sciences, Nanjing, 210008, China.
| | - Ruonan Wu
- Earth and Biological Sciences Directorate, Pacific Northwest National Lab, Richland, WA, 99352, USA
| | - Dongsheng Wang
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Pedro J J Alvarez
- Department of Civil and Environmental Engineering, Rice University, Houston, TX, 77005, USA
| | - Pingfeng Yu
- College of Environmental and Resource Sciences, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
68
|
Sabzali S, Pazhouhnia S, Shahzamani K, Sedeh PA. Role of phage therapy in acute gastroenteritis. JOURNAL OF RESEARCH IN MEDICAL SCIENCES : THE OFFICIAL JOURNAL OF ISFAHAN UNIVERSITY OF MEDICAL SCIENCES 2025; 30:2. [PMID: 40200968 PMCID: PMC11974603 DOI: 10.4103/jrms.jrms_464_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/30/2024] [Accepted: 11/25/2024] [Indexed: 04/10/2025]
Abstract
The gut ecosystem, comprising the gut microbiota and its interactions, plays a crucial role in human health and disease. This complex ecosystem involves a diverse array of microorganisms such as viruses, fungi, and bacteria, collectively known as the gut microbiota. These microorganisms contribute to various functions, including nutrient metabolism and immune modulation, thereby impacting human health. Dysbiosis, or an imbalance in the gut microbiota, has been associated with the pathogenesis of several diseases, ranging from intestinal disorders such as inflammatory bowel disease to extra-intestinal conditions such as metabolic and neurological disorders. The implications of dysbiosis in the gut ecosystem are far-reaching, affecting not only gastrointestinal health but also contributing to the development and progression of conditions such as autoimmune gastritis and gastric cancer. Furthermore, the burden of antimicrobial use and subsequent side effects, including antibiotic resistance, poses additional challenges in managing gastrointestinal diseases. In light of these complexities, investigating the role of bacteriophages as regulators of the gut ecosystem and their potential clinical applications presents a promising opportunity to tackle antibiotic resistance and fight infectious diseases.
Collapse
Affiliation(s)
- Somaieh Sabzali
- Department of Biology, Faculty of Basic Sciences, Lorestan University, Khorramabad, Iran
| | - Setareh Pazhouhnia
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Kiana Shahzamani
- Hepatitis Research Center, Faculty of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Peyman Adibi Sedeh
- Gastroenterology and Hepatology Research Center, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
69
|
Rubí-Rangel LM, León-Félix J, Villicaña C. Exploring Viral Interactions in Clavibacter Species: In Silico Analysis of Prophage Prevalence and Antiviral Defenses. Life (Basel) 2025; 15:187. [PMID: 40003596 PMCID: PMC11856565 DOI: 10.3390/life15020187] [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: 11/26/2024] [Revised: 12/30/2024] [Accepted: 12/30/2024] [Indexed: 02/27/2025] Open
Abstract
Clavibacter is a phytopathogenic genus that causes severe diseases in economically important crops, yet the role of prophages in its evolution, pathogenicity, and adaptation remains poorly understood. In this study, we used PHASTER, Prophage Hunter, and VirSorter2 to identify prophage-like sequences in publicly available Clavibacter genomes. Prophage predictions were checked by hand to make them more accurate. We identified 353 prophages, predominantly in chromosomes, with some detected phage-plasmids. Most prophages exhibited traits of advanced domestication, such as an unimodal genome length distribution, reduced numbers of integrases, and minimal transposable elements, suggesting long-term interactions with their bacterial hosts. Comparative genomic analyses uncovered high genetic diversity, with distinct prophage clusters showing species-specific and interspecies conservation patterns. Functional annotation revealed prophage-encoded genes were involved in sugar metabolism, heavy metal resistance, virulence factors, and antibiotic resistance, highlighting their contribution to host fitness and environmental adaptation. Defense system analyses revealed that, despite lacking CRISPR-Cas, Clavibacter genomes harbor diverse antiviral systems, including PD-Lambda-1, AbiE, and MMB_gp29_gp30, some encoded within prophages. These findings underscore the pervasive presence of prophages in Clavibacter and their role in shaping bacterial adaptability and evolution.
Collapse
Affiliation(s)
- Lucía Margarita Rubí-Rangel
- Centro de Investigación en Alimentación y Desarrollo A. C., Carretera a Eldorado Km 5.5, Campo El Diez, Culiacán 80110, Sinaloa, Mexico; (L.M.R.-R.); (J.L.-F.)
| | - Josefina León-Félix
- Centro de Investigación en Alimentación y Desarrollo A. C., Carretera a Eldorado Km 5.5, Campo El Diez, Culiacán 80110, Sinaloa, Mexico; (L.M.R.-R.); (J.L.-F.)
| | - Claudia Villicaña
- CONAHCYT-Centro de Investigación en Alimentación y Desarrollo A. C., Carretera a Eldorado Km 5.5, Campo El Diez, Culiacán 80110, Sinaloa, Mexico
| |
Collapse
|
70
|
Dai Z, Chen Y, Guan Z, Chen X, Tan K, Yang K, Yan X, Liu Y, Gong Z, Han W, Zou T. Structural and mechanistic insights into the activation of a short prokaryotic argonaute system from archaeon Sulfolobus islandicus. Nucleic Acids Res 2025; 53:gkaf059. [PMID: 39898546 PMCID: PMC11788926 DOI: 10.1093/nar/gkaf059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 01/13/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025] Open
Abstract
Prokaryotic Argonaute proteins (pAgos) defend the host against invading nucleic acids, including plasmids and viruses. Short pAgo systems confer immunity by inducing cell death upon detecting invading nucleic acids. However, the activation mechanism of the SiAgo system, comprising a short pAgo from the archaeon Sulfolobus islandicus and its associated proteins SiAga1 and SiAga2, remains largely unknown. Here, we determined the cryo-electron microscopy structures of the SiAgo-Aga1 apo complex and the RNA-DNA-bound SiAgo-Aga1 complex at resolutions of 2.7 and 3.0 Å, respectively. Our results revealed that a positively charged pocket is generated from the interaction between SiAgo and SiAga1, exhibiting an architecture similar to APAZ-pAgo of short pAgo systems and accommodating the nucleic acids. Further investigation elucidated the conserved mechanism of nucleic acid recognition by SiAgo-Aga1. Both the SiAgo-Aga1 interaction and nucleic acid recognition by the complex are essential for antiviral defense. Biochemical and structural analyses demonstrated that SiAgo-Aga1 undergoes extensive conformational changes upon binding to the RNA-DNA duplex, thereby licensing its interaction with the effector SiAga2 to trigger the immune response. Overall, our findings highlight the evolutionary conservation of Agos across phylogenetic clades and provide structural insights into the activation mechanism of the SiAgo system.
Collapse
Affiliation(s)
- Zhikang Dai
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, 430070 Wuhan, Hubei, China
| | - Yu Chen
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, 430070 Wuhan, Hubei, China
| | - Zeyuan Guan
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, 430070 Wuhan, Hubei, China
| | - Xueting Chen
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, 430070 Wuhan, Hubei, China
| | - Keyi Tan
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, 430070 Wuhan, Hubei, China
| | - Kaiyue Yang
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, 430070 Wuhan, Hubei, China
| | - Xuhui Yan
- National Key Laboratory of Crop Genetic Improvement, Hubei Hongshan Laboratory, Huazhong Agricultural University, 430070 Wuhan, Hubei, China
| | - Yidong Liu
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, 430070 Wuhan, Hubei, China
| | - Zhou Gong
- Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences,430071 Wuhan, Hubei, China
| | - Wenyuan Han
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, 430070 Wuhan, Hubei, China
| | - Tingting Zou
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, 430070 Wuhan, Hubei, China
| |
Collapse
|
71
|
Zhang Z, Todeschini TC, Wu Y, Kogay R, Naji A, Rodriguez JC, Mondi R, Kaganovich D, Taylor DW, Bravo JPK, Teplova M, Amen T, Koonin EV, Patel DJ, Nobrega FL. Kiwa is a bacterial membrane-embedded defence supercomplex activated by phage-induced membrane changes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2023.02.26.530102. [PMID: 39896579 PMCID: PMC11785009 DOI: 10.1101/2023.02.26.530102] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Bacteria and archaea deploy diverse, sophisticated defence systems to counter virus infection, yet many immunity mechanisms remain poorly understood. Here, we characterise the Kiwa defence system as a membrane-associated supercomplex that senses changes in the membrane induced by phage infection and plasmid conjugation. This supercomplex, comprising KwaA tetramers bound to KwaB dimers, as its basic repeating unit, detects structural stress via KwaA, activating KwaB, which binds ejected phage DNA through its DUF4868 domain, stalling phage DNA replication forks and thus disrupting replication and late transcription. We show that phage-encoded DNA mimic protein Gam, which inhibits RecBCD, also targets Kiwa through KwaB recognition. However, Gam binding to one defence system precludes its inhibition of the other. These findings reveal a distinct mechanism of bacterial immune coordination, where sensing of membrane disruptions and inhibitor partitioning enhance protection against phages and plasmids.
Collapse
Affiliation(s)
- Zhiying Zhang
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Shared first authors
| | - Thomas C. Todeschini
- School of Biological Sciences, University of Southampton, SO17 1BJ Southampton, UK
- Shared first authors
- Current address: RNA Therapeutics Institute, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Yi Wu
- School of Biological Sciences, University of Southampton, SO17 1BJ Southampton, UK
- Shared first authors
| | - Roman Kogay
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland, USA
| | - Ameena Naji
- School of Biological Sciences, University of Southampton, SO17 1BJ Southampton, UK
- Current address: School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, UK
| | | | - Rupavidhya Mondi
- School of Biological Sciences, University of Southampton, SO17 1BJ Southampton, UK
- Current address: The William Harvey Research Institute, Barts and The London School of Medicine, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Daniel Kaganovich
- School of Biological Sciences, University of Southampton, SO17 1BJ Southampton, UK
| | - David W. Taylor
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, 78712, USA
- Interdisciplinary Life Sciences Graduate Programs, Austin, TX 78712, USA
- Center for Systems and Synthetic Biology, University of Texas at Austin, Austin, TX, 78712, USA
- LIVESTRONG Cancer Institutes, Dell Medical School, Austin, TX, 78712, USA
| | - Jack P. K. Bravo
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX, 78712, USA
- Current address: Institute of Science and Technology Austria (ISTA), Klosterneuburg, Austria
| | - Marianna Teplova
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Triana Amen
- School of Biological Sciences, University of Southampton, SO17 1BJ Southampton, UK
| | - Eugene V. Koonin
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland, USA
| | - Dinshaw J. Patel
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Franklin L. Nobrega
- School of Biological Sciences, University of Southampton, SO17 1BJ Southampton, UK
| |
Collapse
|
72
|
Van Goethem MW, Bezuidt OKI, Pierneef R, Vikram S, Hopkins DW, Aspray T, Hall G, Woodborne S, Hogg ID, Northen TR, Kong W, Daffonchio D, Cowan DA, Van de Peer Y, Delgado-Baquerizo M, Makhalanyane TP. Novel adaptive immune systems in pristine Antarctic soils. Sci Rep 2025; 15:2368. [PMID: 39827180 PMCID: PMC11742911 DOI: 10.1038/s41598-024-83942-y] [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: 08/29/2024] [Accepted: 12/18/2024] [Indexed: 01/22/2025] Open
Abstract
Antarctic environments are dominated by microorganisms, which are vulnerable to viral infection. Although several studies have investigated the phylogenetic repertoire of bacteria and viruses in these poly-extreme environments with freezing temperatures, high ultra violet irradiation levels, low moisture availability and hyper-oligotrophy, the evolutionary mechanisms governing microbial immunity remain poorly understood. Using genome-resolved metagenomics, we test the hypothesis that Antarctic poly-extreme high-latitude microbiomes harbour diverse adaptive immune systems. Our analysis reveals the prevalence of prophages in bacterial genomes (Bacteroidota and Verrucomicrobiota), suggesting the significance of lysogenic infection strategies in Antarctic soils. Furthermore, we demonstrate the presence of diverse CRISPR-Cas arrays, including Class 1 arrays (Types I-B, I-C, and I-E), alongside systems exhibiting novel gene architecture among their effector cas genes. Notably, a Class 2 system featuring type V variants lacks CRISPR arrays, encodes Cas1 and Cas2 adaptation module genes. Phylogenetic analysis of Cas12 effector proteins hints at divergent evolutionary histories compared to classified type V effectors and indicates that TnpB is likely the ancestor of Cas12 nucleases. Our findings suggest substantial novelty in Antarctic cas sequences, likely driven by strong selective pressures. These results underscore the role of viral infection as a key evolutionary driver shaping polar microbiomes.
Collapse
Affiliation(s)
- Marc W Van Goethem
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Pretoria, 0028, South Africa
- Biological and Environmental Sciences and Engineering Division (BESE), King Abdullah University of Science and Technology, 23955-6900, Thuwal, Saudi Arabia
| | - Oliver K I Bezuidt
- Department of Biochemistry, Genetics and Microbiology, Faculty of Natural and Agricultural Sciences, University of Pretoria, Hatfield, Pretoria, 0028, South Africa
- Department of Biochemistry, Genetics and Microbiology, Faculty of Natural and Agricultural Sciences, DSI/NRF SARChI in Marine Microbiomics, University of Pretoria, Hatfield, Pretoria, 0028, South Africa
| | - Rian Pierneef
- Department of Biochemistry, Genetics and Microbiology, Faculty of Natural and Agricultural Sciences, University of Pretoria, Hatfield, Pretoria, 0028, South Africa
- Department of Biochemistry, Genetics and Microbiology, Faculty of Natural and Agricultural Sciences, DSI/NRF SARChI in Marine Microbiomics, University of Pretoria, Hatfield, Pretoria, 0028, South Africa
| | - Surendra Vikram
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Pretoria, 0028, South Africa
| | - David W Hopkins
- Scotland's Rural College, West Mains Road, Edinburgh, EH9 3JG, UK
| | - Thomas Aspray
- School of Energy, Geoscience, Infrastructure and Society, Heriot-Watt University, Edinburgh, EH14 4AS, UK
| | - Grant Hall
- Mammal Research Institute, University of Pretoria, Private Bag X20, Hatfield, 0028, South Africa
| | | | - Ian D Hogg
- Canadian High Arctic Research Station, Polar Knowledge Canada; and School of Science, University of Waikato, Waitkato, New Zealand
| | - Trent R Northen
- Molecular EcoSystems Biology Division, Lawrence Berkeley National Laboratory, 1 Cyclotron Rd, Berkeley, CA, 94720, USA
| | - Weidong Kong
- State Key Laboratory of Tibetan Plateau Earth System and Resources Environment, Institute of Tibetan Plateau Research, Chinese Academy of Sciences, Beijing, 100101, China
| | - Daniele Daffonchio
- Biological and Environmental Sciences and Engineering Division (BESE), King Abdullah University of Science and Technology, 23955-6900, Thuwal, Saudi Arabia
| | - Don A Cowan
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Pretoria, 0028, South Africa
| | - Yves Van de Peer
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Pretoria, 0028, South Africa
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052, Ghent, Belgium
- Center for Plant Systems Biology, VIB, 9052, Ghent, Belgium
- Bioinformatics Institute Ghent, Ghent University, 9052, Ghent, Belgium
| | - Manuel Delgado-Baquerizo
- Laboratorio de Biodiversidad y Funcionamiento Ecosistémico, Instituto de Recursos Naturales y Agrobiología de Sevilla (IRNAS), CSIC, Seville, Spain
- Unidad Asociada CSIC-UPO (BioFun), Universidad Pablo de Olavide, Seville, Spain
| | - Thulani P Makhalanyane
- Department of Biochemistry, Genetics and Microbiology, Faculty of Natural and Agricultural Sciences, University of Pretoria, Hatfield, Pretoria, 0028, South Africa.
- Department of Microbiology, Faculty of Science, Stellenbosch University, Stellenbosch, 7600, South Africa.
- The School for Data Science and Computational Thinking, Stellenbosch University, Stellenbosch, 7600, South Africa.
| |
Collapse
|
73
|
Baca CF, Marraffini LA. Nucleic acid recognition during prokaryotic immunity. Mol Cell 2025; 85:309-322. [PMID: 39824170 PMCID: PMC11750177 DOI: 10.1016/j.molcel.2024.12.007] [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: 09/27/2024] [Revised: 12/04/2024] [Accepted: 12/09/2024] [Indexed: 01/20/2025]
Abstract
Parasitic elements often spread to hosts through the delivery of their nucleic acids to the recipient. This is particularly true for the primary parasites of bacteria, bacteriophages (phages) and plasmids. Although bacterial immune systems can sense a diverse set of infection signals, such as a protein unique to the invader or the disruption of natural host processes, phage and plasmid nucleic acids represent some of the most common molecules that are recognized as foreign to initiate defense. In this review, we will discuss the various elements of invader nucleic acids that can be distinguished by bacterial host immune systems as "non-self" and how this signal is relayed to activate an immune response.
Collapse
Affiliation(s)
- Christian F Baca
- Laboratory of Bacteriology, The Rockefeller University, New York, NY 10065, USA; Tri-Institutional PhD Program in Chemical Biology, Weill Cornell Medical College, Rockefeller University and Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| | - Luciano A Marraffini
- Laboratory of Bacteriology, The Rockefeller University, New York, NY 10065, USA; Howard Hughes Medical Institute, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
74
|
Bae HW, Choi SY, Ki HJ, Cho YH. Pseudomonas aeruginosa as a model bacterium in antiphage defense research. FEMS Microbiol Rev 2025; 49:fuaf014. [PMID: 40240293 PMCID: PMC12035536 DOI: 10.1093/femsre/fuaf014] [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: 12/31/2024] [Revised: 04/09/2025] [Accepted: 04/15/2025] [Indexed: 04/18/2025] Open
Abstract
Bacteriophages, or phages, depend on their bacterial hosts for proliferation, leading to a coevolutionary relationship characterized by on-going arms races, where bacteria evolve diverse antiphage defense systems. The development of in silico methods and high-throughput screening techniques has dramatically expanded our understanding of bacterial antiphage defense systems, enormously increasing the known repertoire of the distinct mechanisms across various bacterial species. These advances have revealed that bacterial antiphage defense systems exhibit a remarkable level of complexity, ranging from highly conserved to specialized mechanisms, underscoring the intricate nature of bacterial antiphage defense systems. In this review, we provide a concise snapshot of antiphage defense research highlighting two preponderantly commandeered approaches and classification of the known antiphage defense systems. A special focus is placed on the model bacterial pathogen, Pseudomonas aeruginosa in antiphage defense research. We explore the complexity and adaptability of these systems, which play crucial roles in genome evolution and adaptation of P. aeruginosa in response to an arsenal of diverse phage strains, emphasizing the importance of this organism as a key emerging model bacterium in recent antiphage defense research.
Collapse
Affiliation(s)
- Hee-Won Bae
- Program of Biopharmaceutical Science, Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Gyeonggi-do 13488, Korea
| | - Shin-Yae Choi
- Program of Biopharmaceutical Science, Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Gyeonggi-do 13488, Korea
| | - Hyeong-Jun Ki
- Program of Biopharmaceutical Science, Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Gyeonggi-do 13488, Korea
| | - You-Hee Cho
- Program of Biopharmaceutical Science, Department of Pharmacy, College of Pharmacy and Institute of Pharmaceutical Sciences, CHA University, Gyeonggi-do 13488, Korea
| |
Collapse
|
75
|
Johnson S, Weigele P, Fomenkov A, Ge A, Vincze A, Eaglesham J, Roberts R, Sun Z. Domainator, a flexible software suite for domain-based annotation and neighborhood analysis, identifies proteins involved in antiviral systems. Nucleic Acids Res 2025; 53:gkae1175. [PMID: 39657740 PMCID: PMC11754643 DOI: 10.1093/nar/gkae1175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 11/07/2024] [Accepted: 11/15/2024] [Indexed: 12/12/2024] Open
Abstract
The availability of large databases of biological sequences presents an opportunity for in-depth exploration of gene diversity and function. Bacterial defense systems are a rich source of diverse but difficult to annotate genes with biotechnological applications. In this work, we present Domainator, a flexible and modular software suite for domain-based gene neighborhood and protein search, extraction and clustering. We demonstrate the utility of Domainator through three examples related to bacterial defense systems. First, we cluster CRISPR-associated Rossman fold (CARF) containing proteins with difficult to annotate effector domains, classifying most of them as likely transcriptional regulators and a subset as likely RNases. Second, we extract and cluster P4-like phage satellite defense hotspots, identify an abundant variant of Lamassu defense systems and demonstrate its in vivo activity against several T-even phages. Third, we integrate a protein language model into Domainator and use it to identify restriction endonucleases with low similarity to known reference sequences, validating the activity of one example in vitro. Domainator is made available as an open-source package with detailed documentation and usage examples.
Collapse
Affiliation(s)
| | | | | | - Andrew Ge
- New England Biolabs Inc., Ipswich, MA 01938, USA
| | - Anna Vincze
- New England Biolabs Inc., Ipswich, MA 01938, USA
| | | | | | - Zhiyi Sun
- New England Biolabs Inc., Ipswich, MA 01938, USA
| |
Collapse
|
76
|
Yi W, Zhu N, Peng Z, Chu X, Sun H, Song L, Guo Z, Pain A, Luo Z, Guan Q. In silico characterization of defense system hotspots in Acinetobacter spp. Commun Biol 2025; 8:39. [PMID: 39794449 PMCID: PMC11723918 DOI: 10.1038/s42003-025-07459-4] [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: 10/05/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
The bacteria-phage arm race drives the evolution of diverse bacterial defenses. This study identifies and characterizes the defense hotspots in Acinetobacter baumannii using a reference-free approach. Among 4383 high-quality genomes, we found a total of 17,430 phage defense systems and with 54.54% concentrated in 21 hotspots. These hotspots exhibit distinct preferences for different defense systems, and co-occurrence patterns suggest synergistic interactions. Additionally, the mobile genetic elements are abundant around these hotspots, likely facilitating horizontal transfer and evolution of defense systems. The number of hotspots increases in species phylogenetically closer to Acinetobacter baumannii, but the number of defense systems per hotspot varies due to particular selective pressures. These findings provide critical insights into the genetic organization of phage defense systems, contributing to a broader understanding of bacterial immunity and the evolutionary dynamics that shape Acinetobacter genomes. This knowledge lays the foundation for developing targeted interventions to combat antibiotic resistance Acinetobacter baumannii.
Collapse
Affiliation(s)
- Wenjing Yi
- Bioinformatics Laboratory, Infectious Diseases and Pathogen Biology Center, The First Hospital of Jilin University, Changchun, China
| | - Ning Zhu
- Bioinformatics Laboratory, Infectious Diseases and Pathogen Biology Center, The First Hospital of Jilin University, Changchun, China
| | - Zhihan Peng
- Department of Respiratory Medicine, Infectious Diseases and Pathogen Biology Center, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
| | - Xiao Chu
- Department of Respiratory Medicine, Infectious Diseases and Pathogen Biology Center, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
| | - Haotian Sun
- Department of Respiratory Medicine, Infectious Diseases and Pathogen Biology Center, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
| | - Lei Song
- Department of Respiratory Medicine, Infectious Diseases and Pathogen Biology Center, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, The First Hospital of Jilin University, Changchun, China
| | - Zhimin Guo
- Department of Laboratory Medicine, Infectious Diseases and Pathogen Biology Center, The First Hospital of Jilin University, Changchun, China
| | - Arnab Pain
- Pathogen Genomics Laboratory, Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, 23955-6900, Jeddah, Makkah, Saudi Arabia
| | - Zhaoqing Luo
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Qingtian Guan
- Bioinformatics Laboratory, Infectious Diseases and Pathogen Biology Center, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
77
|
Mortensen S, Kuncová S, Lormand J, Myers T, Kim SK, Lee V, Winkler W, Sondermann H. Structural and bioinformatics analyses identify deoxydinucleotide-specific nucleases and their association with genomic islands in gram-positive bacteria. Nucleic Acids Res 2025; 53:gkae1235. [PMID: 39778863 PMCID: PMC11706625 DOI: 10.1093/nar/gkae1235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 11/21/2024] [Accepted: 12/02/2024] [Indexed: 01/11/2025] Open
Abstract
Dinucleases of the DEDD superfamily, such as oligoribonuclease, Rexo2 and nanoRNase C, catalyze the essential final step of RNA degradation, the conversion of di- to mononucleotides. The active sites of these enzymes are optimized for substrates that are two nucleotides long, and do not discriminate between RNA and DNA. Here, we identified a novel DEDD subfamily, members of which function as dedicated deoxydinucleases (diDNases) that specifically hydrolyze single-stranded DNA dinucleotides in a sequence-independent manner. Crystal structures of enzyme-substrate complexes reveal that specificity for DNA stems from a combination of conserved structural elements that exclude diribonucleotides as substrates. Consistently, diDNases fail to complement the loss of enzymes that act on diribonucleotides, indicating that these two groups of enzymes support distinct cellular functions. The genes encoding diDNases are found predominantly in genomic islands of Actinomycetes and Clostridia, which, together with their association with phage-defense systems, suggest potential roles in bacterial immunity.
Collapse
Affiliation(s)
- Sofia Mortensen
- CSSB Centre for Structural Systems Biology, Deutsches Elektronen-Synchrotron DESY, Notkestr. 85, 22607 Hamburg, Germany
| | - Stanislava Kuncová
- CSSB Centre for Structural Systems Biology, Deutsches Elektronen-Synchrotron DESY, Notkestr. 85, 22607 Hamburg, Germany
| | - Justin D Lormand
- CSSB Centre for Structural Systems Biology, Deutsches Elektronen-Synchrotron DESY, Notkestr. 85, 22607 Hamburg, Germany
| | - Tanner M Myers
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742, USA
| | - Soo-Kyoung Kim
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
| | - Vincent T Lee
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
| | - Wade C Winkler
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742, USA
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA
| | - Holger Sondermann
- CSSB Centre for Structural Systems Biology, Deutsches Elektronen-Synchrotron DESY, Notkestr. 85, 22607 Hamburg, Germany
- Christian-Albrechts-University, 24118 Kiel, Germany
| |
Collapse
|
78
|
Tesson F, Huiting E, Wei L, Ren J, Johnson M, Planel R, Cury J, Feng Y, Bondy-Denomy J, Bernheim A. Exploring the diversity of anti-defense systems across prokaryotes, phages and mobile genetic elements. Nucleic Acids Res 2025; 53:gkae1171. [PMID: 39657785 PMCID: PMC11724313 DOI: 10.1093/nar/gkae1171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/30/2024] [Accepted: 12/06/2024] [Indexed: 12/12/2024] Open
Abstract
The co-evolution of prokaryotes, phages and mobile genetic elements (MGEs) has driven the diversification of defense and anti-defense systems alike. Anti-defense proteins have diverse functional domains, sequences and are typically small, creating a challenge to detect anti-defense homologs across prokaryotic and phage genomes. To date, no tools comprehensively annotate anti-defense proteins within a desired sequence. Here, we developed 'AntiDefenseFinder'-a free open-source tool and web service that detects 156 anti-defense systems of one or more proteins in any genomic sequence. Using this dataset, we identified 47 981 anti-defense systems distributed across prokaryotes and their viruses. We found that some genes co-localize in 'anti-defense islands', including Escherichia coli T4 and Lambda phages, although many appear standalone. Eighty-nine per cent anti-defense systems localize only or preferentially in MGE. However, >80% of anti-Pycsar protein 1 (Apyc1) resides in nonmobile regions of bacterial genomes. Evolutionary analysis and biochemical experiments revealed that Apyc1 likely originated in bacteria to regulate cyclic nucleotide (cNMP) signaling, but phage co-opted Apyc1 to overcome cNMP-utilizing defenses. With the AntiDefenseFinder tool, we hope to facilitate the identification of the full repertoire of anti-defense systems in MGEs, the discovery of new protein functions and a deeper understanding of host-pathogen arms race.
Collapse
Affiliation(s)
- Florian Tesson
- Institut Pasteur, CNRS UMR3525, Molecular Diversity of Microbes Lab, 25-28 rue du Docteur Roux, 75015, Paris, France
| | - Erin Huiting
- Department of Microbiology and Immunology, University of California San Francisco, Genentech Hall Room N372E UCSF Mail Code 2200 600 16th Street San Francisco, San Francisco, CA 94158, USA
| | - Linlin Wei
- State Key Laboratory of Chemical Resource Engineering, Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beisanhuan EastRoad 15, Chaoyang Distract, 100029 Beijing, China
| | - Jie Ren
- State Key Laboratory for Biology of Plant Diseases and Insect Pests, Ministry of Agriculture, Institute of Plant Protection, Chinese Academy of Agricultural Sciences, 2 Yuanmingyuan W Rd, Haidian District, 100091 Beijing, China
| | - Matthew Johnson
- Department of Microbiology and Immunology, University of California San Francisco, Genentech Hall Room N372E UCSF Mail Code 2200 600 16th Street San Francisco, San Francisco, CA 94158, USA
| | - Rémi Planel
- Bioinformatics and Biostatistics Hub, Institut Pasteur, Université Paris Cité, 25-28 rue du Docteur Roux, 75015,Paris, France
| | - Jean Cury
- Institut Pasteur, CNRS UMR3525, Molecular Diversity of Microbes Lab, 25-28 rue du Docteur Roux, 75015, Paris, France
| | - Yue Feng
- State Key Laboratory of Chemical Resource Engineering, Beijing Key Laboratory of Bioprocess, College of Life Science and Technology, Beijing University of Chemical Technology, Beisanhuan EastRoad 15, Chaoyang Distract, 100029 Beijing, China
| | - Joseph Bondy-Denomy
- Department of Microbiology and Immunology, University of California San Francisco, Genentech Hall Room N372E UCSF Mail Code 2200 600 16th Street San Francisco, San Francisco, CA 94158, USA
- Quantitative Biosciences Institute, University of California San Francisco, 1700 4th St, San Francisco, CA 94158, USA
| | - Aude Bernheim
- Institut Pasteur, CNRS UMR3525, Molecular Diversity of Microbes Lab, 25-28 rue du Docteur Roux, 75015, Paris, France
| |
Collapse
|
79
|
Patel KM, Seed KD. A Class 1 OLD family nuclease encoded by Vibrio cholerae is countered by a vibriophage-encoded direct inhibitor. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.06.631583. [PMID: 39829814 PMCID: PMC11741312 DOI: 10.1101/2025.01.06.631583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Bacteria are constantly threatened by their viral predators (phages), which has resulted in the development of defense systems for bacterial survival. One family of defense systems found widely across bacteria are OLD (for overcoming lysogeny defect) family nucleases. Despite recent discoveries regarding Class 2 and 4 OLD family nucleases and how phages overcome them, Class 1 OLD family nucleases warrant further study as there has only been one anti-phage Class 1 OLD family nuclease described to date. Here, we identify the Vibrio cholerae-encoded Class 1 OLD family nuclease Vc OLD and describe its disruption of genome replication of the lytic vibriophage ICP1. Furthermore, we examine its in vitro activity, identifying Vc OLD as a DNA nickase. Finally, we identify the first direct inhibitor of a Class 1 OLD family nuclease, the ICP1-encoded Oad1. Our research further illuminates Class 1 OLD family nucleases' role in phage defense and explores the dynamic arms race between V. cholerae and its predatory phage ICP1.
Collapse
Affiliation(s)
- Kishen M. Patel
- Infectious Diseases and Immunity Graduate Group, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
- Department of Plant and Microbial Biology, University of California, Berkeley, Berkeley, California, USA
| | - Kimberley D. Seed
- Department of Plant and Microbial Biology, University of California, Berkeley, Berkeley, California, USA
| |
Collapse
|
80
|
Zhang Q, Li J, Tuo J, Liu S, Liu Y, Liu P, Ye L, Zhang XX. Long-term metagenomic insights into the roles of antiviral defense systems in stabilizing activated sludge bacterial communities. THE ISME JOURNAL 2025; 19:wraf051. [PMID: 40096540 PMCID: PMC11980602 DOI: 10.1093/ismejo/wraf051] [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: 11/27/2024] [Revised: 02/05/2025] [Accepted: 03/13/2025] [Indexed: 03/19/2025]
Abstract
Bacteria have evolved various antiviral defense systems (DSs) to protect themselves, but how DSs respond to the variation of bacteriophages in complex bacterial communities and whether DSs function effectively in maintaining the stability of bacterial community structure and function remain unknown. Here, we conducted a long-term metagenomic investigation on the composition of bacterial and phage communities of monthly collected activated sludge (AS) samples from two full-scale wastewater treatment plants over 6 years and found that DSs were widespread in AS, with 91.1% of metagenome-assembled genomes (MAGs) having more than one complete DS. The stability of the bacterial community was maintained under the fluctuations of the phage community, and DS abundance and phage abundance were strongly positively correlated; there was a 0-3-month time lag in the responses of DSs to phage fluctuations. The rapid turnover of clustered regularly interspaced short palindromic repeat spacer repertoires further highlighted the dynamic nature of bacterial defense mechanisms. A pan-immunity phenomenon was also observed, with nearly identical MAGs showing significant differences in DS composition, which contributed to community stability at the species level. This study provides novel insights into the complexity of phage-bacteria interactions in complex bacterial communities and reveals the key roles of DSs in stabilizing bacterial community structure and function.
Collapse
Affiliation(s)
- Qifeng Zhang
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environment, Nanjing University, Nanjing 210023, China
| | - Jie Li
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environment, Nanjing University, Nanjing 210023, China
| | - Jinhua Tuo
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environment, Nanjing University, Nanjing 210023, China
| | - Shengnan Liu
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environment, Nanjing University, Nanjing 210023, China
| | - Yang Liu
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environment, Nanjing University, Nanjing 210023, China
| | - Peng Liu
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environment, Nanjing University, Nanjing 210023, China
| | - Lin Ye
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environment, Nanjing University, Nanjing 210023, China
| | - Xu-Xiang Zhang
- State Key Laboratory of Pollution Control and Resource Reuse, School of Environment, Nanjing University, Nanjing 210023, China
| |
Collapse
|
81
|
Pradhan B, Deep A, König J, Baaske MD, Corbett KD, Kim E. Loop-extrusion-mediated plasmid DNA cleavage by the bacterial SMC Wadjet complex. Mol Cell 2025; 85:107-116.e5. [PMID: 39626662 DOI: 10.1016/j.molcel.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 09/16/2024] [Accepted: 11/05/2024] [Indexed: 12/11/2024]
Abstract
Structural maintenance of chromosomes (SMC) complexes play pivotal roles in genome organization and maintenance across all domains of life. In prokaryotes, SMC-family Wadjet complexes structurally resemble the widespread MukBEF but serve a defensive role by inhibiting plasmid transformation. We previously showed that Wadjet specifically cleaves plasmid DNA; however, the molecular mechanism underlying plasmid recognition remains unclear. Here, we use in vitro single-molecule imaging to directly visualize DNA loop extrusion and plasmid cleavage by Wadjet. We find that Wadjet is a symmetric loop extruder that simultaneously reels in DNA from both sides of a loop and that this activity requires a dimeric JetABC supercomplex. On surface-anchored plasmid DNAs, Wadjet extrudes the full length of a 44-kb-pair plasmid, stalls, and cleaves DNA. Our findings reveal the role of loop extrusion in the specific recognition and elimination of plasmids by Wadjet and establish loop extrusion as an evolutionarily conserved mechanism among SMC complexes across all kingdoms of life.
Collapse
Affiliation(s)
- Biswajit Pradhan
- Max Planck Institute of Biophysics, 60438 Frankfurt am Main, Germany
| | - Amar Deep
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jessica König
- Max Planck Institute of Biophysics, 60438 Frankfurt am Main, Germany
| | - Martin D Baaske
- Max Planck Institute of Biophysics, 60438 Frankfurt am Main, Germany
| | - Kevin D Corbett
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093, USA; Department of Molecular Biology, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Eugene Kim
- Max Planck Institute of Biophysics, 60438 Frankfurt am Main, Germany.
| |
Collapse
|
82
|
Hegarty B. Making waves: Intelligent phage cocktail design, a pathway to precise microbial control in water systems. WATER RESEARCH 2025; 268:122594. [PMID: 39405620 DOI: 10.1016/j.watres.2024.122594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 09/23/2024] [Accepted: 10/06/2024] [Indexed: 12/19/2024]
Abstract
Current practices in water and wastewater treatment to control unwanted microbes have led to new problems, including health effects from disinfection byproducts, growth of opportunistic pathogens resistant to residual disinfectants (e.g., chlorine), and antibiotic resistance. These challenges are spurring interest in rethinking our practices of microbial control. Simultaneously, advances in molecular biology and computation power are driving renewed interest in using phages (viruses that infect bacteria) to precisely control microbial growth (aka, phage biocontrol). In this Making Waves article, I begin by reviewing the current state of research into phage cocktail design, emphasizing our limited understanding of the features of successful phage cocktails (combinations of multiple types of phages). I describe the state of modeling phage-bacteria interactions and underscore the need for increasing research efforts to predict phage cocktail success, a key gap slowing the application of phage biocontrol. I also detail how research must also focus on techniques for engineering more effective phages to offer a more rapid alternative to phage discovery from natural environments. In this way, phage cocktails comprised of phages with complementary infection strategies may be designed. The final area for increased research effort that I highlight is the need for phage cocktail design to account for possible unintended environmental effects, a risk that is increasingly acknowledged in phage ecology studies but mostly ignored by those developing phage biocontrol technologies. By focusing more research effort towards the areas necessary for intelligent phage cocktail design, we can accelerate the development of phage-based biocontrol in water systems and improve public health.
Collapse
Affiliation(s)
- Bridget Hegarty
- Civil and Environmental Engineering, Case Western Reserve University, Cleveland, OH, 44118, USA.
| |
Collapse
|
83
|
Zborowsky S, Tahan R, Lindell D. Adaptive loss of tRNA gene expression leads to phage resistance in a marine Synechococcus cyanobacterium. Nat Microbiol 2025; 10:66-76. [PMID: 39753669 PMCID: PMC11726456 DOI: 10.1038/s41564-024-01877-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 11/01/2024] [Indexed: 01/12/2025]
Abstract
Synechococcus is a significant primary producer in the oceans, coexisting with cyanophages, which are important agents of mortality. Bacterial resistance against phage infection is a topic of significant interest, yet little is known for ecologically relevant systems. Here we use exogenous gene expression and gene disruption to investigate mechanisms underlying intracellular resistance of marine Synechococcus WH5701 to the Syn9 cyanophage. The restriction-modification and Gabija defence systems possessed by Synechococcus WH5701 did not contribute to resistance. Instead, resistance was primarily driven by insufficient levels of LeuTAA tRNA, preventing translation of key phage genes in a passive, intracellular mode of resistance. Restoring cellular tRNA expression rendered the cyanobacterium sensitive to infection. We propose an evolutionary scenario whereby changes in cell codon usage, acquisition of tRNAs by the phage and loss of cell and phage tRNA expression resulted in an effective means of resistance, highlighting the dynamic interplay between bacteria and phages in shaping their co-evolutionary trajectories.
Collapse
Affiliation(s)
- Sophia Zborowsky
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
- Living Systems Institute, University of Exeter, Exeter, UK
| | - Ran Tahan
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Debbie Lindell
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
84
|
Chang RB, Toyoda HC, Hobbs SJ, Richmond-Buccola D, Wein T, Burger N, Chouchani ET, Sorek R, Kranzusch PJ. A widespread family of viral sponge proteins reveals specific inhibition of nucleotide signals in anti-phage defense. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.30.630793. [PMID: 39803557 PMCID: PMC11722364 DOI: 10.1101/2024.12.30.630793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Cyclic oligonucleotide-based antiviral signaling systems (CBASS) are bacterial anti-phage defense operons that use nucleotide signals to control immune activation. Here we biochemically screen 57 diverse E. coli and Bacillus phages for the ability to disrupt CBASS immunity and discover anti-CBASS 4 (Acb4) from the Bacillus phage SPO1 as the founding member of a large family of >1,300 immune evasion proteins. A 2.1 Å crystal structure of Acb4 in complex with 3'3'-cGAMP reveals a tetrameric assembly that functions as a sponge to sequester CBASS signals and inhibit immune activation. We demonstrate Acb4 alone is sufficient to disrupt CBASS activation in vitro and enable immune evasion in vivo. Analyzing phages that infect diverse bacteria, we explain how Acb4 selectively targets nucleotide signals in host defense and avoids disruption of cellular homeostasis. Together, our results reveal principles of immune evasion protein evolution and explain a major mechanism phages use to inhibit host immunity.
Collapse
Affiliation(s)
- Renee B. Chang
- Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Hunter C. Toyoda
- Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Samuel J. Hobbs
- Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Desmond Richmond-Buccola
- Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Tanita Wein
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Nils Burger
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Edward T. Chouchani
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - Rotem Sorek
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Philip J. Kranzusch
- Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Lead Contact
| |
Collapse
|
85
|
Dymova AA, Kovalev MA, Silantyev AS, Borzykh AA, Osipova PJ, Poddubko SV, Mitkevich VA, Karpov DS, Kostina NV. Unusual Genomic and Biochemical Features of Paenarthrobacter lasiusi sp. nov-A Novel Bacterial Species Isolated from Lasius niger Anthill Soil. Int J Mol Sci 2024; 26:67. [PMID: 39795926 PMCID: PMC11719660 DOI: 10.3390/ijms26010067] [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: 11/26/2024] [Revised: 12/20/2024] [Accepted: 12/21/2024] [Indexed: 01/13/2025] Open
Abstract
The black garden ant (Lasius niger) is a widely distributed species across Europe, North America, and North Africa, playing a pivotal role in ecological processes within its diverse habitats. However, the microbiome associated with L. niger remains poorly investigated. In the present study, we isolated a novel species, Paenarthrobacter lasiusi, from the soil of the L. niger anthill. The genome of P. lasiusi S21 was sequenced, annotated, and searched for groups of genes of physiological, medical, and biotechnological importance. Subsequently, a series of microbiological, physiological, and biochemical experiments were conducted to characterize P. lasiusi S21 with respect to its sugar metabolism, antibiotic resistance profile, lipidome, and capacity for atmospheric nitrogen fixation, among others. A notable feature of the P. lasiusi S21 genome is the presence of two prophages, which may have horizontally transferred host genes involved in stress responses. P. lasiusi S21 synthesizes a number of lipids, including mono- and digalactosyldiacylglycerol, as well as steroid compounds that are typically found in eukaryotic organisms rather than prokaryotes. P. lasiusi S21 exhibits resistance to penicillins, lincosamides, fusidins, and oxazolidinones, despite the absence of specific genes conferring resistance to these antibiotics. Genomic data and physiological tests indicate that P. lasiusi S21 is nonpathogenic to humans. The genome of P. lasiusi S21 contains multiple operons involved in heavy metal metabolism and organic compound inactivation. Consequently, P. lasiusi represents a novel species with an intriguing evolutionary history, manifesting in distinctive genomic, metabolomic, and physiological characteristics. This species may have potential applications in the bioaugmentation of contaminated soils.
Collapse
Affiliation(s)
- Alexandra A. Dymova
- Institute of Biomedical Problems, Russian Academy of Sciences, 123007 Moscow, Russia; (A.A.D.); (A.A.B.); (P.J.O.); (S.V.P.)
- Faculty of Soil Science, M.V. Lomonosov Moscow State University, Leninskie Gory, 1, 119991 Moscow, Russia;
| | - Maxim A. Kovalev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (M.A.K.); (V.A.M.)
| | - Artemiy S. Silantyev
- The Institute of Personalized Cardiology, I.M. Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia;
| | - Anna A. Borzykh
- Institute of Biomedical Problems, Russian Academy of Sciences, 123007 Moscow, Russia; (A.A.D.); (A.A.B.); (P.J.O.); (S.V.P.)
| | - Pamila J. Osipova
- Institute of Biomedical Problems, Russian Academy of Sciences, 123007 Moscow, Russia; (A.A.D.); (A.A.B.); (P.J.O.); (S.V.P.)
| | - Svetlana V. Poddubko
- Institute of Biomedical Problems, Russian Academy of Sciences, 123007 Moscow, Russia; (A.A.D.); (A.A.B.); (P.J.O.); (S.V.P.)
| | - Vladimir A. Mitkevich
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (M.A.K.); (V.A.M.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Dmitry S. Karpov
- Institute of Biomedical Problems, Russian Academy of Sciences, 123007 Moscow, Russia; (A.A.D.); (A.A.B.); (P.J.O.); (S.V.P.)
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (M.A.K.); (V.A.M.)
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Natalia V. Kostina
- Faculty of Soil Science, M.V. Lomonosov Moscow State University, Leninskie Gory, 1, 119991 Moscow, Russia;
| |
Collapse
|
86
|
Cummins T, Songra S, Garrett SR, Blower TR, Mariano G. Multi-conflict islands are a widespread trend within Serratia spp. Cell Rep 2024; 43:115055. [PMID: 39661517 DOI: 10.1016/j.celrep.2024.115055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 09/12/2024] [Accepted: 11/20/2024] [Indexed: 12/13/2024] Open
Abstract
Bacteria carry numerous anti-phage systems in "defense islands" or hotspots. Recent studies have delineated the content and boundaries of these islands in various species, revealing instances of islands that encode additional factors, including antibiotic resistance genes, stress genes, type VI secretion system (T6SS)-dependent effectors, and virulence factors. Our study identifies three defense islands in the Serratia genus with a mixed cargo of anti-phage systems, virulence factors, and different types of anti-bacterial modules, revealing a widespread trend of co-accumulation that extends beyond T6SS-dependent effectors to colicins and contact-dependent inhibition systems. We further report the identification of four distinct anti-phage system/subtypes, including a previously unreported Toll/interleukin (IL)-1 receptor (TIR)-domain-containing system with population-wide immunity, and two loci co-opting a predicted T6SS-related protein for phage defense. This study enhances our understanding of the protein domains that can be co-opted for phage defense, resulting in a highly diversified anti-phage arsenal.
Collapse
Affiliation(s)
- Thomas Cummins
- Department of Microbial Sciences, School of Biosciences, University of Surrey, Guildford, Surrey, UK
| | - Suraj Songra
- Department of Microbial Sciences, School of Biosciences, University of Surrey, Guildford, Surrey, UK
| | - Stephen R Garrett
- Michael DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON L8S 4K1, Canada; Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Tim R Blower
- Department of Biosciences, Durham University, Stockton Road, Durham, UK
| | - Giuseppina Mariano
- Department of Microbial Sciences, School of Biosciences, University of Surrey, Guildford, Surrey, UK.
| |
Collapse
|
87
|
Conte AN, Ruchel ME, Ridgeway SM, Kibby EM, Nagy TA, Whiteley AT. Phage detection by a bacterial NLR-related protein is mediated by DnaJ. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.04.597415. [PMID: 38895412 PMCID: PMC11185742 DOI: 10.1101/2024.06.04.597415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Bacteria encode a wide range of antiphage systems and a subset of these proteins are homologous to components of the human innate immune system. Mammalian nucleotide-binding and leucine-rich repeat containing proteins (NLRs) and bacterial NLR-related proteins use a central NACHT domain to link detection of infection with initiation of an antimicrobial response. Bacterial NACHT proteins provide defense against both DNA and RNA phages. Here we determine the mechanism of RNA phage detection by the bacterial NLR-related protein bNACHT25 in E. coli. bNACHT25 was specifically activated by Emesvirus ssRNA phages and analysis of MS2 phage escaper mutants that evaded detection revealed a critical role for Coat Protein (CP). A genetic assay confirmed CP was sufficient to activate bNACHT25 but the two proteins did not directly interact. Instead, we found bNACHT25 requires the host chaperone DnaJ to detect CP. Our data suggest that bNACHT25 detects a wide range of phages by guarding a host cell process rather than binding a specific phage-derived molecule.
Collapse
Affiliation(s)
- Amy N. Conte
- Department of Biochemistry, University of Colorado Boulder, Boulder, CO, USA
| | - Madison E. Ruchel
- Department of Biochemistry, University of Colorado Boulder, Boulder, CO, USA
- Department of Biology, Front Range Community College, Longmont, CO, USA
| | | | - Emily M. Kibby
- Department of Biochemistry, University of Colorado Boulder, Boulder, CO, USA
| | - Toni A. Nagy
- Department of Biochemistry, University of Colorado Boulder, Boulder, CO, USA
| | - Aaron T. Whiteley
- Department of Biochemistry, University of Colorado Boulder, Boulder, CO, USA
| |
Collapse
|
88
|
Kibby EM, Robbins LK, Deep A, Min NK, Whalen LA, Nagy TA, Freeborn L, Corbett KD, Whiteley AT. A bacterial NLR-related protein recognizes multiple unrelated phage triggers to sense infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.17.629029. [PMID: 39763729 PMCID: PMC11702601 DOI: 10.1101/2024.12.17.629029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Immune systems must rapidly sense viral infections to initiate antiviral signaling and protect the host. Bacteria encode >100 distinct viral (phage) defense systems and each has evolved to sense crucial components or activities associated with the viral lifecycle. Here we used a high-throughput AlphaFold-multimer screen to discover that a bacterial NLR-related protein directly senses multiple phage proteins, thereby limiting immune evasion. Phages encoded as many as 5 unrelated activators that were predicted to bind the same interface of a C-terminal sensor domain. Genetic and biochemical assays confirmed activators bound to the bacterial NLR-related protein at high affinity, induced oligomerization, and initiated signaling. This work highlights how in silico strategies can identify complex protein interaction networks that regulate immune signaling across the tree of life.
Collapse
Affiliation(s)
- Emily M. Kibby
- Department of Biochemistry, University of Colorado Boulder, Boulder, CO, USA
| | - Laurel K. Robbins
- Department of Biochemistry, University of Colorado Boulder, Boulder, CO, USA
- Interdisciplinary Quantitative Biology Program (IQ Biology), BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, USA
| | - Amar Deep
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California, USA
| | - Nathan K. Min
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California, USA
| | - Lindsay A. Whalen
- Department of Biochemistry, University of Colorado Boulder, Boulder, CO, USA
| | - Toni A. Nagy
- Department of Biochemistry, University of Colorado Boulder, Boulder, CO, USA
| | - Layla Freeborn
- Research Computing, Office of Information Technology, University of Colorado Boulder, Boulder, CO, USA
| | - Kevin D. Corbett
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, California, USA
- Department of Molecular Biology, University of California, San Diego, La Jolla, California, USA
| | - Aaron T. Whiteley
- Department of Biochemistry, University of Colorado Boulder, Boulder, CO, USA
| |
Collapse
|
89
|
Blankenchip CL, Corbett KD. Bacterial WYL domain transcriptional repressors sense single-stranded DNA to control gene expression. Nucleic Acids Res 2024; 52:13723-13732. [PMID: 39588753 DOI: 10.1093/nar/gkae1101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 10/04/2024] [Accepted: 10/26/2024] [Indexed: 11/27/2024] Open
Abstract
Bacteria encode a wide array of immune systems to protect themselves against ubiquitous bacteriophages and foreign DNA elements. While these systems' molecular mechanisms are becoming increasingly well known, their regulation remains poorly understood. Here, we show that an immune system-associated transcriptional repressor of the wHTH-WYL-WCX family, CapW, directly binds single-stranded DNA to sense DNA damage and activate expression of its associated immune system. We show that CapW mediates increased expression of a reporter gene in response to DNA damage in a host cell. CapW directly binds single-stranded DNA by-products of DNA repair through its WYL domain, causing a conformational change that releases the protein from double-stranded DNA. In an Escherichia coli CBASS system with an integrated capW gene, we find that CapW-mediated transcriptional activation is important for this system's ability to prevent induction of a λ prophage. Overall, our data reveal the molecular mechanisms of WYL-domain transcriptional repressors, and provide an example of how bacteria can balance the protective benefits of carrying anti-phage immune systems against the inherent risk of these systems' aberrant activation.
Collapse
Affiliation(s)
- Chelsea L Blankenchip
- Biomedical Sciences Graduate Program, University of California, San Diego, 9500 Gilman Dr. La Jolla, CA 92093, USA
- Department of Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Dr. La Jolla, CA 92093, USA
| | - Kevin D Corbett
- Department of Cellular and Molecular Medicine, University of California, San Diego, 9500 Gilman Dr. La Jolla, CA 92093, USA
- Department of Molecular Biology, University of California, San Diego, 9500 Gilman Dr. La Jolla, CA, 92093, USA
| |
Collapse
|
90
|
Jdeed G, Morozova VV, Tikunova NV. Genome Analysis of Anti-Phage Defense Systems and Defense Islands in Stenotrophomonas maltophilia: Preservation and Variability. Viruses 2024; 16:1903. [PMID: 39772210 PMCID: PMC11680222 DOI: 10.3390/v16121903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 11/29/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
Anti-phage defense systems are widespread in bacteria due to the latter continuous adaptation to infection by bacteriophages (phages). Stenotrophomonas maltophilia has a high degree of intrinsic antibiotic resistance, which makes phage therapy relevant for the treatment of infections caused by this species. Studying the array of anti-phage defense systems that could be found in S. maltophilia helps in better adapting the phages to the systems present in the pathogenic bacteria. Pangenome analysis of the available S. maltophilia strains with complete genomes that were downloaded from GenBank, including five local genomes, indicated a wide set of 72 defense systems and subsystems that varied between the strains. Seven of these systems were present in more than 20% of the studied genomes and the proteins encoded by the systems were variable in most of the cases. A total of 27 defense islands were revealed where defense systems were found; however, more than 60% of the instances of systems were found in four defense islands. Several elements linked to the transfer of these systems were found. No obvious associations between the pattern of distribution of the anti-phage defense systems of S. maltophilia and the phylogenetic features or the isolation site were found.
Collapse
Affiliation(s)
- Ghadeer Jdeed
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, Prosp. Lavrentieva 8, Novosibirsk 630090, Russia;
| | | | - Nina V. Tikunova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, Prosp. Lavrentieva 8, Novosibirsk 630090, Russia;
| |
Collapse
|
91
|
Li J, Yuan N, Zhai Y, Wang M, Hao M, Liu X, Zhou D, Liu W, Jin Y, Wang A. Protein disulfide isomerase A4 binds to Brucella BtpB and mediates intracellular NAD +/NADH metabolism in RAW264.7 cells. Int Immunopharmacol 2024; 142:113046. [PMID: 39226825 DOI: 10.1016/j.intimp.2024.113046] [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: 07/06/2024] [Revised: 08/22/2024] [Accepted: 08/27/2024] [Indexed: 09/05/2024]
Abstract
The Toll/interleukin-1 receptor (TIR) signaling domain is distributed widely in mammalian Toll-like receptors and adaptors, plant nucleotide-binding leucine-rich repeat receptors, and specific bacterial virulence proteins. Proteins that possess TIR domain exhibit NADase activity which is distinct from the canonical signaling function of these domains. However, the effects of bacterial TIR domain proteins on host metabolic switches and the underlying mechanism of NADase activity in these proteins remain unclear. Here, we utilized Brucella TIR domain-containing type IV secretion system effector protein, BtpB, to explore the mechanism of NADase activity in host cells. We showed that using ectopic expression BtpB not only generates depletion of NAD+ but also loss of NADH and ATP in RAW264.7 macrophage cells. Moreover, immunoprecipitation-mass spectrometry, co-immunoprecipitation, and confocal microscope assays revealed that BtpB interacted with host protein disulfide isomerase A4 (PDIA4). The Brucella mutant strain deleted the gene for BtpB, significantly decreased PDIA4 expression. Furthermore, our data revealed that PDIA4 played an important role in regulating intracellular NAD+/NADH levels in macrophages, and PDIA4 overexpression restored the decline of intracellular NAD+ and NADH levels induced by Brucella BtpB. The results provide new insights into the metabolic regulatory activity of TIR domain proteins in the critical human and animal pathogen Brucella.
Collapse
Affiliation(s)
- Junmei Li
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; Department of Veterinary Medicine, College of Coastal Agricultural Science, Guangdong Ocean University, Zhanjiang, 524088, China
| | - Ningqiu Yuan
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Yunyi Zhai
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Minghui Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Mingyue Hao
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Xiaofang Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Dong Zhou
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Wei Liu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China
| | - Yaping Jin
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China; Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling 712100, China
| | - Aihua Wang
- Key Laboratory of Animal Biotechnology of the Ministry of Agriculture, Northwest A&F University, Yangling 712100, China; Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northwest A&F University, Yangling 712100, China.
| |
Collapse
|
92
|
Ramírez C, Romero J. Unveiling Hidden Allies: In Silico Discovery of Prophages in Tenacibaculum Species. Antibiotics (Basel) 2024; 13:1184. [PMID: 39766574 PMCID: PMC11672841 DOI: 10.3390/antibiotics13121184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/20/2024] [Accepted: 11/23/2024] [Indexed: 01/11/2025] Open
Abstract
Tenacibaculosis, caused by Tenacibaculum species, is a significant disease in aquaculture, leading to high mortality and economic losses. Antibiotic treatment raises concerns about resistance, making phage therapy an interesting alternative. Analyzing phage traces in Tenacibaculum genomes is crucial for developing these bacteriophage-based strategies. METHODS We assessed the presence of prophages in 212 Tenacibaculum genomes/assemblies available in the NCBI repository, comprising several species and global locations, using the PHASTEST program. Then, we focused on those regions classified as intact, evaluating the most common phages found using VICTOR. The protein of interest discovered in the prophages was evaluated using the ProtParam, DeepTMHMM, InterPro, and Phyre2 tools. In addition, we evaluated the presence of antiphage defense systems in those genomes with intact prophages using the DefenseFinder tool. RESULTS We identified 25 phage elements in 24 out of the 212 Tenacibaculum genomes/assemblies analyzed, with 11% of the assemblies containing phage elements. These were concentrated in T. maritimum and T. mesophilum, which harbored 10 and 7 prophage regions, respectively. Of the identified elements, six were classified as intact, including four in T. maritimum, with the most common phages belonging to the Pippivirus and Siphoviridae families. Bioinformatic analysis showed that the putative endolysin is a stable protein of 432 amino acids and 49.8 kDa, with three transmembrane helices and a CHAP domain, structurally similar to the CHAP lytic domain of S. aureus bacteriophage K. CONCLUSIONS Key prophage elements in Tenacibaculum, especially in T. maritimum, show promise for phage therapy against tenacibaculosis, supporting sustainable, antibiotic-free treatments in aquaculture.
Collapse
Affiliation(s)
| | - Jaime Romero
- Laboratorio de Biotecnología de Alimentos, Instituto de Nutrición y Tecnología de los Alimentos (INTA), Universidad de Chile, El Líbano 5524, Santiago 7830489, Chile;
| |
Collapse
|
93
|
Mahata T, Kanarek K, Goren MG, Marimuthu Ragavan R, Bosis E, Qimron U, Salomon D. Gamma-Mobile-Trio systems are mobile elements rich in bacterial defensive and offensive tools. Nat Microbiol 2024; 9:3268-3283. [PMID: 39443754 DOI: 10.1038/s41564-024-01840-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 09/25/2024] [Indexed: 10/25/2024]
Abstract
The evolutionary arms race between bacteria and phages led to the emergence of bacterial immune systems whose diversity and dynamics remain poorly understood. Here we use comparative genomics to describe a widespread genetic element, defined by the presence of the Gamma-Mobile-Trio (GMT) proteins, that serves as a reservoir of offensive and defensive tools. We demonstrate, using Vibrio parahaemolyticus as a model, that GMT-containing genomic islands are active mobile elements. Furthermore, we show that GMT islands' cargoes contain various anti-phage defence systems, antibacterial type VI secretion system (T6SS) effectors and antibiotic-resistance genes. We reveal four anti-phage defence systems encoded within GMT islands and further characterize one system, GAPS1, showing it is triggered by a phage capsid protein to induce cell dormancy. Our findings underscore the need to broaden the concept of 'defence islands' to include defensive and offensive tools, as both share the same mobile elements for dissemination.
Collapse
Affiliation(s)
- Tridib Mahata
- Department of Clinical Microbiology and Immunology, School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Katarzyna Kanarek
- Department of Clinical Microbiology and Immunology, School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Moran G Goren
- Department of Clinical Microbiology and Immunology, School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Rameshkumar Marimuthu Ragavan
- Department of Clinical Microbiology and Immunology, School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Eran Bosis
- Department of Biotechnology Engineering, Braude College of Engineering, Karmiel, Israel.
| | - Udi Qimron
- Department of Clinical Microbiology and Immunology, School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel.
| | - Dor Salomon
- Department of Clinical Microbiology and Immunology, School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
94
|
Murtazalieva K, Mu A, Petrovskaya A, Finn RD. The growing repertoire of phage anti-defence systems. Trends Microbiol 2024; 32:1212-1228. [PMID: 38845267 DOI: 10.1016/j.tim.2024.05.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 05/10/2024] [Accepted: 05/13/2024] [Indexed: 12/06/2024]
Abstract
The biological interplay between phages and bacteria has driven the evolution of phage anti-defence systems (ADSs), which evade bacterial defence mechanisms. These ADSs bind and inhibit host defence proteins, add covalent modifications and deactivate defence proteins, degrade or sequester signalling molecules utilised by host defence systems, synthesise and restore essential molecules depleted by bacterial defences, or add covalent modifications to phage molecules to avoid recognition. Overall, 145 phage ADSs have been characterised to date. These ADSs counteract 27 of the 152 different bacterial defence families, and we hypothesise that many more ADSs are yet to be discovered. We discuss high-throughput approaches (computational and experimental) which are indispensable for discovering new ADSs and the limitations of these approaches. A comprehensive characterisation of phage ADSs is critical for understanding phage-host interplay and developing clinical applications, such as treatment for multidrug-resistant bacterial infections.
Collapse
Affiliation(s)
- Khalimat Murtazalieva
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, UK; University of Cambridge, Cambridge, UK
| | - Andre Mu
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, UK; Sanger Institute, Wellcome Trust Genome Campus, Hinxton, UK
| | - Aleksandra Petrovskaya
- Nencki Institute of Experimental Biology, Warsaw, Poland; University of Copenhagen, Copenhagen, Denmark
| | - Robert D Finn
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Hinxton, UK.
| |
Collapse
|
95
|
Jiang S, Chen C, Huang W, He Y, Du X, Wang Y, Ou H, Deng Z, Xu C, Jiang L, Wang L, Chen S. A widespread phage-encoded kinase enables evasion of multiple host antiphage defence systems. Nat Microbiol 2024; 9:3226-3239. [PMID: 39506096 DOI: 10.1038/s41564-024-01851-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 10/10/2024] [Indexed: 11/08/2024]
Abstract
DNA degradation (Dnd) is a widespread bacterial antiphage defence system that relies on DNA phosphorothioate (PT) modification for self/non-self discrimination and subsequent degradation of unmodified DNA. Phages employ counterstrategies to evade host immunity, but anti-Dnd immunity has not been characterized. Here we report an immune evasion protein encoded by the Salmonella phage JSS1 that contributes to subverting Dnd and other defence systems. Using quantitative proteomic and phosphoproteomic analyses, we show that the protein JSS1_004 employs N-terminal Ser/Thr/Tyr protein kinase activity to catalyse the multisite phosphorylation of host DndFGH. Notably, JSS1_004 also phosphorylates other bacterial immune systems to varying degrees, including CRISPR‒Cas, QatABCD, SIR2+HerA and DUF4297+HerA. Given that JSS1_004 and its homologues are widespread in phylogenetically diverse phages, we suggest that this strategy constitutes a family of immune evasion proteins that increases the chances of phage proliferation even when a host deploys multiple defence systems.
Collapse
Affiliation(s)
- Susu Jiang
- Department of Burn and Plastic Surgery, Shenzhen Key Laboratory of Microbiology in Genomic Modification and Editing and Application, Shenzhen Institute of Translational Medicine, Medical Innovation Technology Transformation Center of Shenzhen Second People's Hospital, Shenzhen University Medical School, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
- Department of Gastroenterology, Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Disease, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Chao Chen
- Department of Burn and Plastic Surgery, Shenzhen Key Laboratory of Microbiology in Genomic Modification and Editing and Application, Shenzhen Institute of Translational Medicine, Medical Innovation Technology Transformation Center of Shenzhen Second People's Hospital, Shenzhen University Medical School, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Wanqiu Huang
- Department of Burn and Plastic Surgery, Shenzhen Key Laboratory of Microbiology in Genomic Modification and Editing and Application, Shenzhen Institute of Translational Medicine, Medical Innovation Technology Transformation Center of Shenzhen Second People's Hospital, Shenzhen University Medical School, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
- Department of Gastroenterology, Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Disease, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Yue He
- Department of Burn and Plastic Surgery, Shenzhen Key Laboratory of Microbiology in Genomic Modification and Editing and Application, Shenzhen Institute of Translational Medicine, Medical Innovation Technology Transformation Center of Shenzhen Second People's Hospital, Shenzhen University Medical School, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
- Department of Gastroenterology, Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Disease, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Xuan Du
- Department of Burn and Plastic Surgery, Shenzhen Key Laboratory of Microbiology in Genomic Modification and Editing and Application, Shenzhen Institute of Translational Medicine, Medical Innovation Technology Transformation Center of Shenzhen Second People's Hospital, Shenzhen University Medical School, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
- Department of Gastroenterology, Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Disease, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Yi Wang
- Department of Burn and Plastic Surgery, Shenzhen Key Laboratory of Microbiology in Genomic Modification and Editing and Application, Shenzhen Institute of Translational Medicine, Medical Innovation Technology Transformation Center of Shenzhen Second People's Hospital, Shenzhen University Medical School, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
- Department of Gastroenterology, Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Disease, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Hongda Ou
- Department of Burn and Plastic Surgery, Shenzhen Key Laboratory of Microbiology in Genomic Modification and Editing and Application, Shenzhen Institute of Translational Medicine, Medical Innovation Technology Transformation Center of Shenzhen Second People's Hospital, Shenzhen University Medical School, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Zixin Deng
- Department of Gastroenterology, Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Disease, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Congrui Xu
- Department of Gastroenterology, Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Disease, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Lixu Jiang
- Department of Burn and Plastic Surgery, Shenzhen Key Laboratory of Microbiology in Genomic Modification and Editing and Application, Shenzhen Institute of Translational Medicine, Medical Innovation Technology Transformation Center of Shenzhen Second People's Hospital, Shenzhen University Medical School, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Lianrong Wang
- Department of Gastroenterology, Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Disease, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
- Department of Respiratory Diseases, Institute of Pediatrics, Shenzhen Children's Hospital, Shenzhen, China
| | - Shi Chen
- Department of Burn and Plastic Surgery, Shenzhen Key Laboratory of Microbiology in Genomic Modification and Editing and Application, Shenzhen Institute of Translational Medicine, Medical Innovation Technology Transformation Center of Shenzhen Second People's Hospital, Shenzhen University Medical School, The First Affiliated Hospital of Shenzhen University, Shenzhen, China.
- Department of Gastroenterology, Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Hubei Clinical Center and Key Laboratory of Intestinal and Colorectal Disease, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, China.
| |
Collapse
|
96
|
Huang P, Yan P, Guo L, Fei W, Li Z, Liu J, Kong J, Yao Y, Lu M, Xiao Y, Chen M. The mechanism of bacterial defense system DdmDE from Lactobacillus casei. Cell Res 2024; 34:873-876. [PMID: 39448873 PMCID: PMC11614852 DOI: 10.1038/s41422-024-01042-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 10/10/2024] [Indexed: 10/26/2024] Open
Affiliation(s)
- Pingping Huang
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Purui Yan
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Lijie Guo
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Wenying Fei
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Zhaoxing Li
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Jingxian Liu
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Jianping Kong
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yue Yao
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Meiling Lu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China
- Chongqing Innovation Institute of China Pharmaceutical University, Chongqing, China
- Department of Biochemistry, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Yibei Xiao
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China.
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China.
- Chongqing Innovation Institute of China Pharmaceutical University, Chongqing, China.
- Engineering Research Center for Anti-infective Drug Discovery, Ministry of Education (ERCADD), Nanjing, Jiangsu, China.
| | - Meirong Chen
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, China.
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, China.
- Chongqing Innovation Institute of China Pharmaceutical University, Chongqing, China.
| |
Collapse
|
97
|
Tuck OT, Adler BA, Armbruster EG, Lahiri A, Hu JJ, Zhou J, Pogliano J, Doudna JA. Genome integrity sensing by the broad-spectrum Hachiman antiphage defense complex. Cell 2024; 187:6914-6928.e20. [PMID: 39395413 DOI: 10.1016/j.cell.2024.09.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 07/02/2024] [Accepted: 09/12/2024] [Indexed: 10/14/2024]
Abstract
Hachiman is a broad-spectrum antiphage defense system of unknown function. We show here that Hachiman is a heterodimeric nuclease-helicase complex, HamAB. HamA, previously a protein of unknown function, is the effector nuclease. HamB is the sensor helicase. HamB constrains HamA activity during surveillance of intact double-stranded DNA (dsDNA). When the HamAB complex detects DNA damage, HamB helicase activity activates HamA, unleashing nuclease activity. Hachiman activation degrades all DNA in the cell, creating "phantom" cells devoid of both phage and host DNA. We demonstrate Hachiman activation in the absence of phage by treatment with DNA-damaging agents, suggesting that Hachiman responds to aberrant DNA states. Phylogenetic similarities between the Hachiman helicase and enzymes from eukaryotes and archaea suggest deep functional symmetries with other important helicases across domains of life.
Collapse
Affiliation(s)
- Owen T Tuck
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720, USA; Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Benjamin A Adler
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA 94720, USA; California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, Berkeley, CA 94720, USA
| | - Emily G Armbruster
- School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Arushi Lahiri
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Jason J Hu
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Julia Zhou
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Joe Pogliano
- School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jennifer A Doudna
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720, USA; Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA 94720, USA; California Institute for Quantitative Biosciences (QB3), University of California, Berkeley, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA 94720, USA; MBIB Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA; Gladstone Institutes, University of California, San Francisco, San Francisco, CA 94720, USA; Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
98
|
Oshiro RT, Dunham DT, Seed KD. The vibriophage-encoded inhibitor OrbA abrogates BREX-mediated defense through the ATPase BrxC. J Bacteriol 2024; 206:e0020624. [PMID: 39404463 PMCID: PMC11580459 DOI: 10.1128/jb.00206-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 09/13/2024] [Indexed: 10/23/2024] Open
Abstract
Bacteria and phages are locked in a co-evolutionary arms race where each entity evolves mechanisms to restrict the proliferation of the other. Phage-encoded defense inhibitors have proven powerful tools to interrogate how defense systems function. A relatively common defense system is BREX (bacteriophage exclusion); however, how BREX functions to restrict phage infection remains poorly understood. A BREX system encoded by the sulfamethoxazole and trimethoprim (SXT) integrative and conjugative element, VchInd5, was recently identified in Vibrio cholerae, the causative agent of the diarrheal disease cholera. The lytic phage ICP1 (International Centre for Diarrhoeal Disease Research, Bangladesh cholera phage 1) that co-circulates with V. cholerae encodes the BREX-inhibitor OrbA, but how OrbA inhibits BREX is unclear. Here, we determine that OrbA inhibits BREX using a unique mechanism from known BREX inhibitors by directly binding to the BREX component BrxC. BrxC has a functional ATPase domain that, when mutated, not only disrupts BrxC function but also alters how BrxC multimerizes. Furthermore, we find that OrbA binding disrupts BrxC-BrxC interactions. We determine that OrbA cannot bind BrxC encoded by the distantly related BREX system encoded by the aSXT VchBan9, and thus fails to inhibit this BREX system that also circulates in epidemic V. cholerae. Lastly, we find that homologs of the VchInd5 BrxC are more diverse than the homologs of the VchBan9 BrxC. These data provide new insight into the function of the BrxC ATPase and highlight how phage-encoded inhibitors can disrupt phage defense systems using different mechanisms.IMPORTANCEWith renewed interest in phage therapy to combat antibiotic-resistant pathogens, understanding the mechanisms bacteria use to defend themselves against phages and the counter-strategies phages evolve to inhibit defenses is paramount. Bacteriophage exclusion (BREX) is a common defense system with few known inhibitors. Here, we probe how the vibriophage-encoded inhibitor OrbA inhibits the BREX system of Vibrio cholerae, the causative agent of the diarrheal disease cholera. By interrogating OrbA function, we have begun to understand the importance and function of a BREX component. Our results demonstrate the importance of identifying inhibitors against defense systems, as they are powerful tools for dissecting defense activity and can inform strategies to increase the efficacy of some phage therapies.
Collapse
Affiliation(s)
- Reid T. Oshiro
- Department of Plant & Microbial Biology, University of California Berkeley, Berkeley, California, USA
| | - Drew T. Dunham
- Department of Plant & Microbial Biology, University of California Berkeley, Berkeley, California, USA
| | - Kimberley D. Seed
- Department of Plant & Microbial Biology, University of California Berkeley, Berkeley, California, USA
| |
Collapse
|
99
|
Yu Z, Guan J, Hanson C, Duong T, Zeng L. Fine-tuned spatiotemporal dynamics of DNA replication during phage lambda infection. J Virol 2024; 98:e0112824. [PMID: 39480083 PMCID: PMC11575281 DOI: 10.1128/jvi.01128-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 10/07/2024] [Indexed: 11/02/2024] Open
Abstract
After the ejection of viral DNA into the host cytoplasm, the temperate bacteriophage (phage) lambda integrates a cascade of expressions from various regulatory genes, coupled with DNA replication, to commit to a decision between lysis and lysogeny. Higher multiplicity of infection (MOI) greatly shifts the decision toward the lysogenic pathway. However, how the phage separates the MOI from replicated viral DNA during lysis-lysogeny decision-making is unclear. To quantitatively understand the role of viral DNA replication, we constructed a reporter system facilitating the visualization of individual copies of phage DNA throughout the phage life cycle, along with the lysis-lysogeny reporters. We showed that intracellular viral DNA diverges between the lytic and lysogenic pathways from the early phase of the infection cycle, mostly due to the synchronization and success of DNA injection, as well as the competition for replication resources, rather than the replication rate. Strikingly, we observed two distinct replication patterns during lysogenization and surprisingly heterogeneous integration kinetics, which advances our understanding of temperate phage life cycles. We revealed that the weak repression function of Cro is critical for an optimal replication rate and plays a crucial role in establishing stable lysogens. IMPORTANCE Temperate bacteriophages, such as lambda, incorporate environmental cues including host abundance and nutrient conditions to make optimal decisions between propagation and dormancy. A higher phage-to-host ratio or multiplicity of infection (MOI) during λ infection strongly biases toward lysogeny. However, a comprehensive understanding of this decision-making process and the impact of phage replication prior to the decision is yet to be achieved. Here, we used fluorescence microscopy to quantitatively track the spatiotemporal progression of viral DNA replication in individual cells with different cell fates. The implementation of this fluorescent reporter system and quantitative analysis workflow opens a new avenue for future studies to delve deeper into various types of virus-host interactions at a high resolution.
Collapse
Affiliation(s)
- Zihao Yu
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, USA
- Center for Phage Technology, Texas A&M University, College Station, Texas, USA
| | - Jingwen Guan
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, USA
- Center for Phage Technology, Texas A&M University, College Station, Texas, USA
| | - Catherine Hanson
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, USA
| | - Trish Duong
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, USA
| | - Lanying Zeng
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, Texas, USA
- Center for Phage Technology, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
100
|
Marton HL, Sagona AP, Kilbride P, Gibson MI. Acidic polymers reversibly deactivate phages due to pH changes. RSC APPLIED POLYMERS 2024; 2:1082-1090. [PMID: 39184364 PMCID: PMC11342163 DOI: 10.1039/d4lp00202d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 08/15/2024] [Indexed: 08/27/2024]
Abstract
Bacteriophages are promising as therapeutics and biotechnological tools, but they also present a problem for routine and commercial bacterial cultures, where contamination must be avoided. Poly(carboxylic acids) have been reported to inhibit phages' ability to infect their bacterial hosts and hence offer an exciting route to discover additives to prevent infection. Their mechanism and limitations have not been explored. Here, we report the role of pH in inactivating phages to determine if the polymers are unique or simply acidic. It is shown that lower pH (=3) triggered by either acidic polymers or similar changes in pH using HCl lead to inhibition. There is no inhibitory activity at higher pHs (in growth media). This was shown across a panel of phages and different molecular weights of commercial and controlled-radical polymerization-derived poly(acrylic acid)s. It is shown that poly(acrylic acid) leads to reversible deactivation of phage, but when the pH is adjusted using HCl alone the phage is irreversibly deactivated. Further experiments using metal binders ruled out ion depletion as the mode of action. These results show that polymeric phage inhibitors may work by unique mechanisms of action and that pH alone cannot explain the observed effects whilst also placing constraints on the practical utility of poly(acrylic acid).
Collapse
Affiliation(s)
- Huba L Marton
- Department of Chemistry, University of Warwick Coventry CV4 7AL UK +44 247 652 4112
| | - Antonia P Sagona
- School of Life Sciences, University of Warwick Coventry CV4 7AL UK
| | | | - Matthew I Gibson
- Department of Chemistry, University of Warwick Coventry CV4 7AL UK +44 247 652 4112
- Warwick Medical School, University of Warwick Coventry CV4 7AL UK
- Department of Chemistry, University of Manchester Oxford Road Manchester M13 9PL UK
- Manchester Institute of Biotechnology, University of Manchester 131 Princess Street Manchester M1 7DN UK
| |
Collapse
|