1
|
Scott TA, Baker KS, Trotter C, Jenkins C, Mostowy S, Hawkey J, Schmidt H, Holt KE, Thomson NR, Baker S. Shigella sonnei: epidemiology, evolution, pathogenesis, resistance and host interactions. Nat Rev Microbiol 2025; 23:303-317. [PMID: 39604656 DOI: 10.1038/s41579-024-01126-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2024] [Indexed: 11/29/2024]
Abstract
Shigella sonnei is a major cause of diarrhoea globally and is increasing in prevalence relative to other Shigella because of multiple demographic and environmental influences. This single-serotype species has traditionally received less attention in comparison to Shigella flexneri and Shigella dysenteriae, which were more common in low-income countries and more tractable in the laboratory. In recent years, we have learned that Shigella are highly complex and highly susceptible to environmental change, as exemplified by epidemiological trends and increasing relevance of S. sonnei. Ultimately, methods, tools and data generated from decades of detailed research into S. flexneri have been used to gain new insights into the epidemiology, microbiology and pathogenesis of S. sonnei. In parallel, widespread adoption of genomic surveillance has yielded insights into antimicrobial resistance, evolution and organism transmission. In this Review, we provide an overview of current knowledge of S. sonnei, highlighting recent insights into this globally disseminated antimicrobial-resistant pathogen and assessing how novel data may impact future vaccine development and implementation.
Collapse
Affiliation(s)
- Timothy A Scott
- Cambridge Institute for Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Department of Medicine, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK.
| | - Kate S Baker
- Department of Clinical Microbiology, Immunology and Infection, University of Liverpool, Liverpool, UK
- Department of Genetics, University of Cambridge, Cambridge, UK
| | - Caroline Trotter
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
- Department of Pathology, University of Cambridge, Cambridge, UK
| | | | - Serge Mostowy
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
| | - Jane Hawkey
- Department of Infectious Diseases, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Hayden Schmidt
- Neutralizing Antibody Center, International AIDS Vaccine Initiative, San Diego, CA, USA
| | - Kathryn E Holt
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
- Department of Infectious Diseases, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Nicholas R Thomson
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London, UK
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Stephen Baker
- Cambridge Institute for Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Department of Medicine, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK.
- International AIDS Vaccine Initiative, London, UK.
| |
Collapse
|
2
|
Cui X, Zhang S, He L, Duan H, Xie Y, Pei X, Yan Y, Du C. In Vitro Biotransformation of Ziziphi Spinosae Semen Saponins by Gut Microbiota from Healthy and Insomniac Groups. Int J Mol Sci 2025; 26:4011. [PMID: 40362251 PMCID: PMC12072027 DOI: 10.3390/ijms26094011] [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: 02/26/2025] [Revised: 03/29/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025] Open
Abstract
Ziziphi Spinosae Semen saponins (ZSSS) show sedative-hypnotic activity but have very low bioavailability, potentially due to their conversion into bioactive metabolites by gut microbiota. In this study, the biotransformation of ZSSS by gut microbiota from healthy humans and patients with insomnia in vitro was analyzed. A total of 21 prototype compounds and 49 metabolites were identified using UHPLC-Q-Orbitrap-MS. Deglycosylation, deoxygenation, dehydration, and deacylation were detected in both healthy individuals and insomniacs. However, oxidation and hydrogenation were uniquely observed in insomniacs. ZSSS can enhance beneficial bacteria, such as Veillonella, Dialister, and Bacteroides. ZSSS can promote the synthesis of short-chain fatty acids (SCFAs), especially acetic acid, propionic acid, and butyric acid. Furthermore, it was found that the sedative-hypnotic activity of ZSSS was enhanced after biotransformation, as determined by a sodium pentobarbital-induced sleeping test (SPST), open-field behavior test (OFBT), and molecular docking experiment (MDE). These results collectively offer valuable insight into the mechanism of action of ZSSS.
Collapse
Affiliation(s)
- Xiaofang Cui
- School of Chinese Materia Medica, Shanxi University of Chinese Medicine, Taiyuan 030619, China; (X.C.); (S.Z.); (L.H.); (H.D.); (Y.X.); (X.P.)
| | - Shengmei Zhang
- School of Chinese Materia Medica, Shanxi University of Chinese Medicine, Taiyuan 030619, China; (X.C.); (S.Z.); (L.H.); (H.D.); (Y.X.); (X.P.)
| | - Ling He
- School of Chinese Materia Medica, Shanxi University of Chinese Medicine, Taiyuan 030619, China; (X.C.); (S.Z.); (L.H.); (H.D.); (Y.X.); (X.P.)
| | - Huizhu Duan
- School of Chinese Materia Medica, Shanxi University of Chinese Medicine, Taiyuan 030619, China; (X.C.); (S.Z.); (L.H.); (H.D.); (Y.X.); (X.P.)
| | - Yujun Xie
- School of Chinese Materia Medica, Shanxi University of Chinese Medicine, Taiyuan 030619, China; (X.C.); (S.Z.); (L.H.); (H.D.); (Y.X.); (X.P.)
| | - Xiangping Pei
- School of Chinese Materia Medica, Shanxi University of Chinese Medicine, Taiyuan 030619, China; (X.C.); (S.Z.); (L.H.); (H.D.); (Y.X.); (X.P.)
| | - Yan Yan
- Modern Research Center for Traditional Chinese Medicine, Shanxi University, Taiyuan 030006, China
| | - Chenhui Du
- School of Chinese Materia Medica, Shanxi University of Chinese Medicine, Taiyuan 030619, China; (X.C.); (S.Z.); (L.H.); (H.D.); (Y.X.); (X.P.)
| |
Collapse
|
3
|
Gomes MC, Brokatzky D, Mostowy S. Shigella-trained pro-inflammatory macrophages protect zebrafish from secondary infection. Cell Rep 2025; 44:115601. [PMID: 40266847 DOI: 10.1016/j.celrep.2025.115601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 02/17/2025] [Accepted: 03/31/2025] [Indexed: 04/25/2025] Open
Abstract
Shigella is an important human pathogen that has no licensed vaccine. Despite decades of seminal work suggesting that its pathogenicity relies on inflammatory cell death of macrophages, the in vivo role of macrophages in controlling Shigella infection remains poorly understood. Here, we use a zebrafish model of innate immune training to investigate the antibacterial role of macrophages following a non-lethal Shigella infection. We found that macrophages are crucial for zebrafish larvae survival during secondary Shigella infection. Consistent with signatures of trained immunity, we demonstrate that bacteria are cleared during training and that protection is independent of the secondary infection site. We show that following Shigella training, macrophages have altered mono- and tri-methylation on lysine 4 in histone 3 (H3K4me1/me3) deposition and shift toward a pro-inflammatory state, characterized by increased tumor necrosis factor alpha (TNF-α) expression and antibacterial reactive oxygen species (ROS) production. We conclude that macrophages are epigenetically reprogrammed by Shigella infection to enhance pro-inflammatory and protective responses.
Collapse
Affiliation(s)
- Margarida C Gomes
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, UK.
| | - Dominik Brokatzky
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, UK
| | - Serge Mostowy
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London, UK.
| |
Collapse
|
4
|
Shrivastava M, Roy D, Chaba R. Long-chain fatty acids as nutrients for Gram-negative bacteria: stress, proliferation, and virulence. Curr Opin Microbiol 2025; 85:102609. [PMID: 40252293 DOI: 10.1016/j.mib.2025.102609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/27/2025] [Accepted: 04/03/2025] [Indexed: 04/21/2025]
Abstract
Bacteria use host-derived long-chain fatty acids (LCFAs) as nutrients, signals, and membrane building blocks. Although the impact of LCFAs on the pathogenesis of Gram-negative bacteria via membrane remodeling or signaling is well-documented, their importance as a nutrient source for bacterial proliferation and virulence is an emerging research area with definitive studies reported only for Salmonella Typhimurium, Vibrio cholerae, and Pseudomonas aeruginosa. Moreover, recent studies in Escherichia coli have shown that LCFA degradation confers redox stress. Here, we review the known role of LCFAs as nutrients during infection in Gram-negative human pathogens and the association of LCFA degradation with redox stress and stress response mechanisms. We suggest that for understanding how, as nutrients, LCFAs influence host-bacterial interactions, it is necessary to resolve whether LCFA utilization also causes redox stress in pathogens, with defense mechanisms preconditioning them for challenging host environments, or if pathogens have pre-existing mechanisms that prevent LCFA-induced stress.
Collapse
Affiliation(s)
- Megha Shrivastava
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, SAS Nagar, Punjab 140306, India
| | - Deeptodeep Roy
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, SAS Nagar, Punjab 140306, India
| | - Rachna Chaba
- Department of Biological Sciences, Indian Institute of Science Education and Research (IISER) Mohali, SAS Nagar, Punjab 140306, India.
| |
Collapse
|
5
|
Kaldalu N, Bērziņš N, Berglund Fick S, Sharma A, Andersson NC, Aedla J, Hinnu M, Puhar A, Hauryliuk V, Tenson T. Antibacterial compounds against non-growing and intracellular bacteria. NPJ ANTIMICROBIALS AND RESISTANCE 2025; 3:25. [PMID: 40216902 PMCID: PMC11992225 DOI: 10.1038/s44259-025-00097-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 03/26/2025] [Indexed: 04/14/2025]
Abstract
Slow- and non-growing bacterial populations, along with intracellular pathogens, often evade standard antibacterial treatments and are linked to persistent and recurrent infections. This necessitates the development of therapies specifically targeting nonproliferating bacteria. To identify compounds active against non-growing uropathogenic Escherichia coli (UPEC) we performed a drug-repurposing screen of 6454 approved drugs and drug candidates. Using dilution-regrowth assays, we identified 39 compounds that either kill non-growing UPEC or delay its regrowth post-treatment. The hits include fluoroquinolones, macrolides, rifamycins, biguanide disinfectants, a pleuromutilin, and anti-cancer agents. Twenty-nine of the hits have not previously been recognized as active against non-growing bacteria. The hits were further tested against non-growing Pseudomonas aeruginosa and Staphylococcus aureus. Ten compounds - solithromycin, rifabutin, mitomycin C, and seven fluoroquinolones-have strong bactericidal activity against non-growing P. aeruginosa, killing >4 log10 of bacteria at 2.5 µM. Solithromycin, valnemulin, evofosfamide, and satraplatin are unique in their ability to selectively target non-growing bacteria, exhibiting poor efficacy against growing bacteria. Finally, 31 hit compounds inhibit the growth of intracellular Shigella flexneri in a human enterocyte infection model, indicating their ability to permeate the cytoplasm of host cells. The identified compounds hold potential for treating persistent infections, warranting further comparative studies with current standard-of-care antibiotics.
Collapse
Affiliation(s)
- Niilo Kaldalu
- Institute of Technology, University of Tartu, Tartu, Estonia.
| | | | | | - Atin Sharma
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå, Sweden
| | | | - Jüri Aedla
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Mariliis Hinnu
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Andrea Puhar
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå, Sweden
- Wellcome-Wolfson Institute for Experimental Medicine (WWIEM), School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Vasili Hauryliuk
- Institute of Technology, University of Tartu, Tartu, Estonia.
- Department of Experimental Medical Science, Lund University, Lund, Sweden.
- Science for Life Laboratory, Lund, Sweden.
- Virus Centre, Lund University, Lund, Sweden.
- NanoLund, Lund University, Lund, Sweden.
| | - Tanel Tenson
- Institute of Technology, University of Tartu, Tartu, Estonia.
| |
Collapse
|
6
|
Zheng H, Liu X, Liang X, Guo S, Qin B, Liu EH, Duan JA. Mechanisms and structure-activity relationships of natural polysaccharides as potential anti-osteoporosis agents: A review. Int J Biol Macromol 2025; 298:139852. [PMID: 39814301 DOI: 10.1016/j.ijbiomac.2025.139852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/09/2025] [Accepted: 01/12/2025] [Indexed: 01/18/2025]
Abstract
In recent years, polysaccharides derived from natural sources have garnered significant attention due to their safety and potential anti-osteoporotic effects. This review provides a comprehensive overview of the sources, distribution, structures, and mechanisms of anti-osteoporosis polysaccharides, as well as an investigation into their structure-activity relationships. Over thirty distinct, homogenous polysaccharides with anti-osteoporosis properties have been extracted from natural sources, primarily categorized as glucans, fructans, galactomannans, glucomannans, and various other heteropolysaccharides. Natural polysaccharides can effectively enhance osteoblast differentiation and mineralization while suppressing osteoclast activation, with the mechanism regulated by the BMP/SMAD/RUNX2, Wnt/Catenin, OPG/RANKL/RANK, and TLR2/NF-κB/NFATc1 signaling pathways. Furthermore, polysaccharides contribute to the prevention of osteoporosis by mitigating oxidative stress, decreasing inflammation, and modulating the gut microbiota. This review also summarizes the relationship between the monosaccharide composition, molecular weight, and glycosidic bond type of polysaccharides and their anti-osteoporotic activity. A comprehensive summary and analysis of the existing deficiencies and challenges in the research of anti-osteoporotic polysaccharides is also concluded. This review may serve as a significant reference for the discovery and utilization of naturally derived anti-osteoporotic polysaccharides in the pharmaceutical and health industries.
Collapse
Affiliation(s)
- Huili Zheng
- Nanjing University of Chinese Medicine/National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing 210029, China; Jiangsu Province Key Laboratory of High Technology Research, Nanjing 210029, China; Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing 210023, China
| | - Xinhui Liu
- Nanjing University of Chinese Medicine/National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing 210029, China; Jiangsu Province Key Laboratory of High Technology Research, Nanjing 210029, China; Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing 210023, China
| | - Xiaofei Liang
- Shaanxi Key Laboratory of Phytochemistry, College of Chemistry and Chemical Engineering, Baoji University of Arts and Sciences, Baoji 721013, China
| | - Sheng Guo
- Nanjing University of Chinese Medicine/National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing 210029, China; Jiangsu Province Key Laboratory of High Technology Research, Nanjing 210029, China; Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing 210023, China
| | - Bing Qin
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - E-Hu Liu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Jin-Ao Duan
- Nanjing University of Chinese Medicine/National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing 210029, China; Jiangsu Province Key Laboratory of High Technology Research, Nanjing 210029, China; Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing 210023, China.
| |
Collapse
|
7
|
Xu D, Guo M, Xu X, Luo G, Liu Y, Bush SJ, Wang C, Xu T, Zeng W, Liao C, Wang Q, Zhao W, Zhao W, Liu Y, Li S, Zhao S, Jiu Y, Sauvonnet N, Lu W, Sansonetti PJ, Ye K. Shigella infection is facilitated by interaction of human enteric α-defensin 5 with colonic epithelial receptor P2Y11. Nat Microbiol 2025; 10:509-526. [PMID: 39901059 DOI: 10.1038/s41564-024-01901-9] [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: 02/28/2024] [Accepted: 12/02/2024] [Indexed: 02/05/2025]
Abstract
Human enteric α-defensin 5 (HD5) is an immune system peptide that acts as an important antimicrobial factor but is also known to promote pathogen infections by enhancing adhesion of the pathogens. The mechanistic basis of these conflicting functions is unknown. Here we show that HD5 induces abundant filopodial extensions in epithelial cells that capture Shigella, a major human enteroinvasive pathogen that is able to exploit these filopodia for invasion, revealing a mechanism for HD5-augmented bacterial invasion. Using multi-omics screening and in vitro, organoid, dynamic gut-on-chip and in vivo models, we identify the HD5 receptor as P2Y11, a purinergic receptor distributed apically on the luminal surface of the human colonic epithelium. Inhibitor screening identified cAMP-PKA signalling as the main pathway mediating the cytoskeleton-regulating activity of HD5. In illuminating this mechanism of Shigella invasion, our findings raise the possibility of alternative intervention strategies against HD5-augmented infections.
Collapse
Affiliation(s)
- Dan Xu
- Key Laboratory of Biomedical Information Engineering (MOE), School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Mengyao Guo
- Key Laboratory of Biomedical Information Engineering (MOE), School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Xin Xu
- Key Laboratory of Biomedical Information Engineering (MOE), School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Gan Luo
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Science, Fudan University, Shanghai, China
| | - Yaxin Liu
- Key Laboratory of Biomedical Information Engineering (MOE), School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Stephen J Bush
- School of Automation Science and Engineering, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Chengyao Wang
- The First Affiliated Hospital, Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Tun Xu
- School of Automation Science and Engineering, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Wenxin Zeng
- Key Laboratory of Biomedical Information Engineering (MOE), School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Chongbing Liao
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Science, Fudan University, Shanghai, China
| | - Qingxia Wang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Science, Fudan University, Shanghai, China
| | - Wei Zhao
- The First Affiliated Hospital, Xi'an Jiaotong University School of Medicine, Xi'an, China
| | - Wenying Zhao
- Key Laboratory of Biomedical Information Engineering (MOE), School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Yuezhuangnan Liu
- Key Laboratory of Biomedical Information Engineering (MOE), School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Shanshan Li
- Key Laboratory of Biomedical Information Engineering (MOE), School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Shuangshuang Zhao
- CAS Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Yaming Jiu
- CAS Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China
| | - Nathalie Sauvonnet
- Tissue Homeostasis group, Biomaterials and Microfluidics Core Facility, Institut Pasteur, Université Paris Cité, Paris, France
| | - Wuyuan Lu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Science, Fudan University, Shanghai, China.
| | - Philippe J Sansonetti
- CAS Key Laboratory of Molecular Virology and Immunology, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai, China.
- Institut Pasteur, Paris, France.
| | - Kai Ye
- Key Laboratory of Biomedical Information Engineering (MOE), School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China.
- School of Automation Science and Engineering, Faculty of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China.
- The First Affiliated Hospital, Xi'an Jiaotong University School of Medicine, Xi'an, China.
| |
Collapse
|
8
|
Haldar R, Halder P, Koley H, Miyoshi SI, Das S. A newly developed oral infection mouse model of shigellosis for immunogenicity and protective efficacy studies of a candidate vaccine. Infect Immun 2025; 93:e0034624. [PMID: 39692481 PMCID: PMC11784180 DOI: 10.1128/iai.00346-24] [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: 08/06/2024] [Accepted: 11/06/2024] [Indexed: 12/19/2024] Open
Abstract
Shigella infection poses a significant public health challenge in the developing world. However, lack of a widely available mouse model that replicates human shigellosis creates a major bottleneck to better understanding of disease pathogenesis and development of newer drugs and vaccines. BALB/c mice pre-treated with streptomycin and iron (FeCl3) plus desferrioxamine intraperitoneally followed by oral infection with virulent Shigella flexneri 2a resulted in diarrhea, loss of body weight, bacterial colonization and progressive colitis characterized by disruption of epithelial lining, loss of crypt architecture with goblet cell depletion, increased polymorphonuclear infiltration into the mucosa, submucosal swelling (edema), and raised proinflammatory cytokines and chemokines in the large intestine. To evaluate the usefulness of the model for vaccine efficacy studies, mice were immunized intranasally with a recombinant protein vaccine containing Shigella invasion protein invasion plasmid antigen B (IpaB). Vaccinated mice conferred protection against Shigella, indicating that the model is suitable for testing of vaccine candidates. To protect both Shigella and Salmonella, a chimeric recombinant vaccine (rIpaB-T2544) was developed by fusing IpaB with Salmonella outer membrane protein T2544. Vaccinated mice developed antigen-specific serum IgG and IgA antibodies and a balanced Th1/Th2 response and were protected against oral challenge with Shigella (S. flexneri 2a, Shigella dysenteriae, and Shigella sonnei) using our present mouse model and Salmonella (Salmonella Typhi and Paratyphi) using an iron overload mouse model. We describe here the development of an oral Shigella infection model in wild-type mouse. This model was successfully used to demonstrate the immunogenicity and protective efficacy of a candidate protein subunit vaccine against Shigella.
Collapse
MESH Headings
- Animals
- Dysentery, Bacillary/prevention & control
- Dysentery, Bacillary/immunology
- Dysentery, Bacillary/microbiology
- Dysentery, Bacillary/pathology
- Disease Models, Animal
- Mice
- Shigella Vaccines/immunology
- Shigella Vaccines/administration & dosage
- Mice, Inbred BALB C
- Antibodies, Bacterial/blood
- Shigella flexneri/immunology
- Female
- Antigens, Bacterial/immunology
- Antigens, Bacterial/genetics
- Bacterial Proteins/immunology
- Bacterial Proteins/genetics
- Vaccines, Synthetic/immunology
- Vaccines, Synthetic/administration & dosage
- Immunoglobulin G/blood
Collapse
Affiliation(s)
- Risha Haldar
- Division of Clinical Medicine, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India
| | - Prolay Halder
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India
| | - Hemanta Koley
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India
| | - Shin-ichi Miyoshi
- Division of Medicine, Dentistry and Pharmaceutical Sciences, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Okayama, Japan
| | - Santasabuj Das
- Division of Clinical Medicine, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, West Bengal, India
- Division of Biological Science, ICMR-National Institute of Occupational Health, Ahmedabad, Gujarat, India
| |
Collapse
|
9
|
Guodong W, Yinhang W, Xinyue W, Hong S, Jian C, Zhanbo Q, Shuwen H. Fecal occult blood affects intestinal microbial community structure in colorectal cancer. BMC Microbiol 2025; 25:34. [PMID: 39833681 PMCID: PMC11745023 DOI: 10.1186/s12866-024-03721-7] [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: 03/17/2024] [Accepted: 12/19/2024] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Gut microbes have been used to predict CRC risk. Fecal occult blood test (FOBT) has been recommended for population screening of CRC. OBJECTIVE To analyze the effects of fecal occult blood test (FOBT) on gut microbes. METHODS Fecal samples from 107 healthy individuals (FOBT-negative) and 111 CRC patients (39 FOBT-negative and 72 FOBT-positive) were included for 16 S ribosomal RNA sequencing. Based on the results of different FOBT, the community structure and diversity of intestinal bacteria in healthy individuals and CRC patients were analyzed. Characteristic gut bacteria were screened, and various machine learning algorithms were applied to construct CRC risk prediction models. RESULTS The gut microbiota of healthy people and CRC patients with different fecal occult blood were mapped. There was no statistical difference in diversity between CRC patients with negative FOBT and positive FOBT. Bacteroides, Blautia and Escherichia-Shigella were more correlated to healthy individuals, while Streptococcus showed higher correlation with CRC patients with negative FOBT. The accuracy of CRC risk prediction model based on the support vector machines (SVM) algorithm was the highest (89.71%). Subsequently, FOBT was included as a characteristic element in the model construction, and the prediction accuracy of the model was all increased. Similarly, the CRC risk prediction model based on SVM algorithm had the highest accuracy (92%). CONCLUSION FOB affects the community composition of gut microbes. When predicting CRC risk based on gut microbiome, considering the influence of FOBT is expected to improve the accuracy of CRC risk prediction.
Collapse
Affiliation(s)
- Wu Guodong
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China
| | - Wu Yinhang
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central Hospital, No.1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, No.1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China
| | - Wu Xinyue
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central Hospital, No.1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, No.1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China
| | - Shen Hong
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China
| | - Chu Jian
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central Hospital, No.1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, No.1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China
| | - Qu Zhanbo
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central Hospital, No.1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, No.1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China
| | - Han Shuwen
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China.
- Fifth Affiliated Clinical Medical College of Zhejiang Chinese Medical University, Huzhou Central Hospital, No.1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China.
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, No.1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, People's Republic of China.
- ASIR (Institute - Association of intelligent systems and robotics), 14B rue Henri Sainte Claire, Deville, Rueil-Malmaison, 92500, France.
| |
Collapse
|
10
|
Li H, Lv Y, Teng Z, Guo R, Jiang L. Shigella Senses the Environmental Cue Leucine to Promote its Virulence Gene Expression in the Colon. J Mol Biol 2024; 436:168798. [PMID: 39303765 DOI: 10.1016/j.jmb.2024.168798] [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: 06/27/2024] [Revised: 09/14/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
Shigella is a foodborne enteropathogenic bacteria that causes severe bacillary dysentery in humans. Shigella primarily colonizes the human colon and causes disease via invasion of colon epithelial cells. However, the signal regulatory mechanisms associated with its colonization and pathogenesis in the colon remain poorly defined. Here, we report a leucine-mediated regulatory mechanism that promotes Shigella virulence gene expression and invasion of colon epithelial cells. Shigella in response to leucine, which is highly abundant in the colon, via the leucine-responsive regulator Lrp and the binding of Lrp with leucine induces the expression of a newly identified small RNA SsrV. SsrV then activates the expression of virF and downstream invasion-related virulence genes by increasing the protein level of the LysR-type transcription regulator LrhA, therefore enabling Shigella invasion of colon epithelial cells. Shigella lacking ssrV displays impaired invasion ability. Collectively, these findings suggest that Shigella employs a leucine-responsive environmental activation mechanism to establish colonization and pathogenicity.
Collapse
Affiliation(s)
- Huiying Li
- Department of Biochemistry and Molecular Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China; National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin 300457, China
| | - Yongyao Lv
- Department of Biochemistry and Molecular Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
| | - Zhiqi Teng
- Department of Biochemistry and Molecular Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250062, China
| | - Rui Guo
- Shandong Center for Food and Drug Evaluation & Inspection, Jinan 250014, China
| | - Lingyan Jiang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, Tianjin 300457, China.
| |
Collapse
|
11
|
Raab JE, Hamilton DJ, Harju TB, Huynh TN, Russo BC. Pushing boundaries: mechanisms enabling bacterial pathogens to spread between cells. Infect Immun 2024; 92:e0052423. [PMID: 38661369 PMCID: PMC11385730 DOI: 10.1128/iai.00524-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024] Open
Abstract
For multiple intracellular bacterial pathogens, the ability to spread directly into adjacent epithelial cells is an essential step for disease in humans. For pathogens such as Shigella, Listeria, Rickettsia, and Burkholderia, this intercellular movement frequently requires the pathogens to manipulate the host actin cytoskeleton and deform the plasma membrane into structures known as protrusions, which extend into neighboring cells. The protrusion is then typically resolved into a double-membrane vacuole (DMV) from which the pathogen quickly escapes into the cytosol, where additional rounds of intercellular spread occur. Significant progress over the last few years has begun to define the mechanisms by which intracellular bacterial pathogens spread. This review highlights the interactions of bacterial and host factors that drive mechanisms required for intercellular spread with a focus on how protrusion structures form and resolve.
Collapse
Affiliation(s)
- Julie E. Raab
- Department of Immunology and Microbiology, School of Medicine, University of Colorado—Anschutz Medical Campus, Denver, Colorado, USA
| | - Desmond J. Hamilton
- Department of Immunology and Microbiology, School of Medicine, University of Colorado—Anschutz Medical Campus, Denver, Colorado, USA
| | - Tucker B. Harju
- Department of Immunology and Microbiology, School of Medicine, University of Colorado—Anschutz Medical Campus, Denver, Colorado, USA
| | - Thao N. Huynh
- Department of Immunology and Microbiology, School of Medicine, University of Colorado—Anschutz Medical Campus, Denver, Colorado, USA
| | - Brian C. Russo
- Department of Immunology and Microbiology, School of Medicine, University of Colorado—Anschutz Medical Campus, Denver, Colorado, USA
| |
Collapse
|
12
|
Bernshtein B, Zhiteneva JA, Janardhanan J, Wagh C, Kelly M, Verma S, Jung W, Basher SR, Amin MA, Mahamud S, Rajib NH, Chowdhury F, Khan AI, Charles RC, Xu P, Kováč P, Chakraborty S, Kaminski RW, Alter G, Bhuiyan TR, Qadri F, Ryan ET. Limited O-specific polysaccharide (OSP)-specific functional antibody responses in young children with Shigella infection in Bangladesh. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.04.611236. [PMID: 39345393 PMCID: PMC11429955 DOI: 10.1101/2024.09.04.611236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Shigellosis is the second leading cause of diarrheal death in children younger than five years of age globally. At present, there is no broadly licensed vaccine against shigella infection. Previous vaccine candidates have failed at providing protection for young children in endemic settings. Improved understanding of correlates of protection against Shigella infection and severe shigellosis in young children living in endemic settings is needed. Here, we applied a functional antibody profiling approach to define Shigella-specific antibody responses in young children versus older individuals with culture-confirmed shigellosis in Bangladesh, a Shigella endemic area. We analyzed Shigella-specific antibody isotypes, FcR binding and antibody-mediated innate immune cell activation in longitudinal serum samples collected at clinical presentation and up to 1 year later. We found that higher initial Shigella O-specific polysaccharide (OSP)-specific and protein-specific IgG and FcγR binding levels correlated with less severe disease regardless of patient age, but that individuals under 5 years of age developed a less prominent class switched, FcR-binding, functional and durable antibody response against both OSP and protein Shigella antigens than older individuals. Focusing on the largest cohort, we found that functional S. flexneri 2a OSP-specific responses were significantly induced only in individuals over age 5 years, and that these responses promoted monocyte phagocytosis and activation. Our findings suggest that in a Shigella endemic region, young children with shigellosis harbor a functional antibody response that fails to maximally activate monocytes; such a response may be important in facilitating subsequent innate cell clearance of Shigella, especially via recruitment and activation of polymorphonuclear cells capable of directly killing Shigella.
Collapse
|
13
|
Saavedra-Sanchez L, Dickinson MS, Apte S, Zhang Y, de Jong M, Skavicus S, Heaton NS, Alto NM, Coers J. The Shigella flexneri effector IpaH1.4 facilitates RNF213 degradation and protects cytosolic bacteria against interferon-induced ubiquitylation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.05.611450. [PMID: 39282383 PMCID: PMC11398459 DOI: 10.1101/2024.09.05.611450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/20/2024]
Abstract
A central signal that marshals host defense against many infections is the lymphocyte-derived cytokine interferon-gamma (IFNγ). The IFNγ receptor is expressed on most human cells and its activation leads to the expression of antimicrobial proteins that execute diverse cell-autonomous immune programs. One such immune program consists of the sequential detection, ubiquitylation, and destruction of intracellular pathogens. Recently, the IFNγ-inducible ubiquitin E3 ligase RNF213 was identified as a pivotal mediator of such a defense axis. RNF213 provides host protection against viral, bacterial, and protozoan pathogens. To establish infections, potentially susceptible intracellular pathogens must have evolved mechanisms that subdue RNF213-controlled cell-autonomous immunity. In support of this hypothesis, we demonstrate here that a causative agent of bacillary dysentery, Shigella flexneri, uses the type III secretion system (T3SS) effector IpaH1.4 to induce the degradation of RNF213. S. flexneri mutants lacking IpaH1.4 expression are bound and ubiquitylated by RNF213 in the cytosol of IFNγ-primed host cells. Linear (M1-) and lysine-linked ubiquitin is conjugated to bacteria by RNF213 independent of the linear ubiquitin chain assembly complex (LUBAC). We find that ubiquitylation of S. flexneri is insufficient to kill intracellular bacteria, suggesting that S. flexneri employs additional virulence factors to escape from host defenses that operate downstream from RNF213-driven ubiquitylation. In brief, this study identified the bacterial IpaH1.4 protein as a direct inhibitor of mammalian RNF213 and highlights evasion of RNF213-driven immunity as a characteristic of the human-tropic pathogen Shigella.
Collapse
Affiliation(s)
- Luz Saavedra-Sanchez
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Mary S. Dickinson
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Shruti Apte
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Yifeng Zhang
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Maarten de Jong
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Samantha Skavicus
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Nicholas S. Heaton
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Neal M. Alto
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jörn Coers
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, NC 27710, USA
- Department of Integrative Immunobiology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
14
|
Miles SL, Holt KE, Mostowy S. Recent advances in modelling Shigella infection. Trends Microbiol 2024; 32:917-924. [PMID: 38423917 DOI: 10.1016/j.tim.2024.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 02/06/2024] [Accepted: 02/06/2024] [Indexed: 03/02/2024]
Abstract
Shigella is an important human-adapted pathogen which contributes to a large global burden of diarrhoeal disease. Together with the increasing threat of antimicrobial resistance and lack of an effective vaccine, there is great urgency to identify novel therapeutics and preventatives to combat Shigella infection. In this review, we discuss the development of innovative technologies and animal models to study mechanisms underlying Shigella infection of humans. We examine recent literature introducing (i) the organ-on-chip model, and its substantial contribution towards understanding the biomechanics of Shigella infection, (ii) the zebrafish infection model, which has delivered transformative insights into the epidemiological success of clinical isolates and the innate immune response to Shigella, (iii) a pioneering oral mouse model of shigellosis, which has helped to discover new inflammasome biology and protective mechanisms against shigellosis, and (iv) the controlled human infection model, which has been effective in translating basic research into human health impact and assessing suitability of novel vaccine candidates. We consider the recent contributions of each model and discuss where the future of modelling Shigella infection lies.
Collapse
Affiliation(s)
- Sydney L Miles
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Kathryn E Holt
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK; Department of Infectious Diseases, Central Clinical School, Monash University, Melbourne, Victoria 3004, Australia
| | - Serge Mostowy
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK.
| |
Collapse
|
15
|
Renteria-Flores FI, García-Chagollán M, Jave-Suárez LF. Bactofection, Bacterial-Mediated Vaccination, and Cancer Therapy: Current Applications and Future Perspectives. Vaccines (Basel) 2024; 12:968. [PMID: 39340000 PMCID: PMC11435753 DOI: 10.3390/vaccines12090968] [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: 07/20/2024] [Revised: 08/19/2024] [Accepted: 08/24/2024] [Indexed: 09/30/2024] Open
Abstract
From the first report in 1891 by Dr. Coley of the effective treatment of tumors in 1000 patients with Streptococcus and the first successful use of bacterial vectors for transferring therapeutic genes in 1980 by Dr. Schnaffer, bactofection has been shown to be a promising strategy in the fields of vaccination, gene therapy, and cancer therapy. This review describes the general theory of bactofection and its advantages, disadvantages, challenges, and expectations, compiling the most notable advances in 14 vaccination studies, 27 cancer therapy studies, and 13 clinical trials. It also describes the current scope of bactofection and promising results. The extensive knowledge of Salmonella biology, as well as the multiple adequacies of the Ty21a vaccination platform, has allowed notable developments worldwide that have mainly been reflected in therapeutic efforts against cancer. In this regard, we strongly recommend the creation of a recombinant Ty21a model that constitutively expresses the GtgE protease from S. typhimurium, allowing this vector to be used in animal trials, thus enhancing the likelihood of favorable results that could quickly transition to clinical trials. From the current perspective, it is necessary to explore a greater diversity of bacterial vectors and find the best combination of implemented attenuations, generating personalized models that guarantee the maximum effectiveness in cancer therapy and vaccination.
Collapse
Affiliation(s)
- Francisco Israel Renteria-Flores
- Institute of Research in Biomedical Sciences, University Center of Health Sciences (CUCS), University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Mariel García-Chagollán
- Institute of Research in Biomedical Sciences, University Center of Health Sciences (CUCS), University of Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Luis Felipe Jave-Suárez
- Division of Immunology, Biomedical Research Centre of the West, Mexican Social Security Institute, Guadalajara 44340, Jalisco, Mexico
| |
Collapse
|
16
|
Henriquez T, Guerra S, Nerini M, Purchase D, Marvasi M. The Tomato Variety Affects the Survival of Shigella flexneri 2a in Fruit Pericarp. Pathogens 2024; 13:379. [PMID: 38787231 PMCID: PMC11124042 DOI: 10.3390/pathogens13050379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 04/08/2024] [Accepted: 04/25/2024] [Indexed: 05/25/2024] Open
Abstract
The presence of enteric pathogens in produce can serve as a significant means of transmitting infections to consumers. Notably, tomatoes, as a type of produce, have been implicated in outbreaks caused by various human pathogens, such as Salmonella enterica and pathogenic Escherichia coli. However, the survival characteristics of Shigella spp. in tomatoes have not been thoroughly investigated. In this study, we assess the survival of S. flexneri 2a in two distinct varieties of post-harvested tomatoes. S. flexneri 2a was used to inoculate both regular-sized Vine tomatoes and cherry-type Mini Plum tomatoes. Our findings reveal no significant difference in Shigella survival in the pericarp of both varieties on day 2 post-inoculation. However, a significant disparity emerges on day 6, where all recovered Shigella colonies exclusively belong to the Mini Plum variety, with none associated with the Vine type. When Shigella was inoculated into the locular cavity (deep inoculation), no significant difference between varieties was observed. Additionally, we investigate the potential role of the SRL pathogenicity island (SRL PAI) in the survival and fitness of S. flexneri 2a in post-harvested tomatoes. Our results indicate that while the SRL PAI is not linked to the survival of the strains in tomato, it does impact their fitness. These findings underscore the variability in Shigella strains' survival capabilities depending on the tomato variety, highlighting the importance of understanding Shigella ecology beyond the human host and identifying molecular determinants influencing bacterial survival to mitigate the risk of future outbreaks. The significance of this data on Shigella persistence in fresh vegetables should not be underestimated, as even a small number of Shigella cells can pose a threat to the health of individuals.
Collapse
Affiliation(s)
- Tania Henriquez
- Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy;
| | - Simona Guerra
- Department of Biology, University of Florence, 50019 Florence, Italy; (S.G.); (M.N.)
| | - Marta Nerini
- Department of Biology, University of Florence, 50019 Florence, Italy; (S.G.); (M.N.)
| | - Diane Purchase
- Department of Natural Sciences, Middlesex University London, London NW4 4BT, UK;
| | - Massimiliano Marvasi
- Department of Biology, University of Florence, 50019 Florence, Italy; (S.G.); (M.N.)
| |
Collapse
|
17
|
Mellouk N, Lensen A, Lopez-Montero N, Gil M, Valenzuela C, Klinkert K, Moneron G, Swistak L, DiGregorio D, Echard A, Enninga J. Post-translational targeting of Rab35 by the effector IcsB of Shigella determines intracellular bacterial niche formation. Cell Rep 2024; 43:114034. [PMID: 38568808 DOI: 10.1016/j.celrep.2024.114034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 11/12/2023] [Accepted: 03/18/2024] [Indexed: 04/05/2024] Open
Abstract
Escape from the bacterial-containing vacuole (BCV) is a key step of Shigella host cell invasion. Rab GTPases subverted to in situ-formed macropinosomes in the vicinity of the BCV have been shown to promote its rupture. The involvement of the BCV itself has remained unclear. We demonstrate that Rab35 is non-canonically entrapped at the BCV. Stimulated emission depletion imaging localizes Rab35 directly on the BCV membranes before vacuolar rupture. The bacterial effector IcsB, a lysine Nε-fatty acylase, is a key regulator of Rab35-BCV recruitment, and we show post-translational acylation of Rab35 by IcsB in its polybasic region. While Rab35 and IcsB are dispensable for the first step of BCV breakage, they are needed for the unwrapping of damaged BCV remnants from Shigella. This provides a framework for understanding Shigella invasion implicating re-localization of a Rab GTPase via its bacteria-dependent post-translational modification to support the mechanical unpeeling of the BCV.
Collapse
Affiliation(s)
- Nora Mellouk
- Institut Pasteur, Université Paris Cité, CNRS UMR3691, Dynamics of Host-Pathogen Interactions Unit, 75015 Paris, France.
| | - Arthur Lensen
- Institut Pasteur, Université Paris Cité, CNRS UMR3691, Dynamics of Host-Pathogen Interactions Unit, 75015 Paris, France
| | - Noelia Lopez-Montero
- Institut Pasteur, Université Paris Cité, CNRS UMR3691, Dynamics of Host-Pathogen Interactions Unit, 75015 Paris, France
| | - Magdalena Gil
- Institut Pasteur, Université Paris Cité, CNRS UMR3691, Dynamics of Host-Pathogen Interactions Unit, 75015 Paris, France
| | - Camila Valenzuela
- Institut Pasteur, Université Paris Cité, CNRS UMR3691, Dynamics of Host-Pathogen Interactions Unit, 75015 Paris, France
| | - Kerstin Klinkert
- Institut Pasteur, Université de Paris Cité, CNRS UMR3691, Membrane Traffic and Cell Division Unit, 75015 Paris, France
| | - Gael Moneron
- Institut Pasteur, CNRS UMR3571, Synapse and Circuit Dynamics Unit, 75015 Paris, France
| | - Léa Swistak
- Institut Pasteur, Université Paris Cité, CNRS UMR3691, Dynamics of Host-Pathogen Interactions Unit, 75015 Paris, France
| | - David DiGregorio
- Institut Pasteur, CNRS UMR3571, Synapse and Circuit Dynamics Unit, 75015 Paris, France
| | - Arnaud Echard
- Institut Pasteur, Université de Paris Cité, CNRS UMR3691, Membrane Traffic and Cell Division Unit, 75015 Paris, France
| | - Jost Enninga
- Institut Pasteur, Université Paris Cité, CNRS UMR3691, Dynamics of Host-Pathogen Interactions Unit, 75015 Paris, France.
| |
Collapse
|
18
|
Song Y, Sun M, Mu G, Tuo Y. Exopolysaccharide secreted by Lactiplantibacillus plantarum Y12 showed inhibitory effect on the pathogenicity of Shigella flexneri in vitro and in vivo. Int J Biol Macromol 2024; 261:129478. [PMID: 38237822 DOI: 10.1016/j.ijbiomac.2024.129478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 01/04/2024] [Accepted: 01/11/2024] [Indexed: 01/28/2024]
Abstract
Shigella flexneri is a prevalent foodborne and waterborne pathogen that threatens human health. Our previous research indicated that the Lactiplantibacillus plantarum Y12 exopolysaccharide (L-EPS) potentially inhibited the pathogenicity of S. flexneri. The in vitro results of this study demonstrated that L-EPS effectively mitigated the symptoms induced by S. flexneri in HT-29 cells, including inhibited gene expression levels of IL-1β, IL-6, IL-8, TNF-α, TLR 2/4, and NOD1/2; decreased apoptosis ratio; and alleviated damage degree of intestinal barrier function (Zona occludens 1, Occludin, and Claudin-1). The in vivo results demonstrated that S. flexneri treated with L-EPS elicited mild adverse physiological manifestations, an inflammatory response, and tissue damage. The infection of S. flexneri caused significant alterations in the abundance of phylum (Firmicutes, Bacteroidota, Actinobacteriota, and Proteobacteria), family (Lachnospiraceae, Muribaculaceae, Rikenellaceae, Prevotellaceaea, Ruminococcaceae, and Lactobaillaceae), and genus (Escherichia Shigella and Lachnospirillaceae NK4A136 group) within the cecal microbiota. These changes were accompanied by perturbations in taurine and hypotaurine metabolism, tricarboxylic acid (TCA) cycle activity, arginine biosynthesis, and histidine metabolic pathways. However, intervention with L-EPS attenuated the dysbiosis of cecal microbiota and metabolic disturbances. In summary, our research suggested a potential application of L-EPS as a functional food additive for mitigating S. flexneri infection.
Collapse
Affiliation(s)
- Yinglong Song
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, PR China
| | - Mengying Sun
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, PR China
| | - Guangqing Mu
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, PR China; Dalian Probiotics Function Research Key Laboratory, Dalian Polytechnic University, Dalian 116034, PR China.
| | - Yanfeng Tuo
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, PR China.
| |
Collapse
|
19
|
Manigat F, Connell LB, Stewart BN, LePabic AR, Tessier CJG, Emlaw JR, Calvert ND, Rössl A, Shuhendler AJ, daCosta CJB, Campbell-Valois FX. pUdOs: Concise Plasmids for Bacterial and Mammalian Cells. ACS Synth Biol 2024; 13:485-497. [PMID: 38235654 PMCID: PMC10878396 DOI: 10.1021/acssynbio.3c00408] [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: 07/06/2023] [Revised: 11/22/2023] [Accepted: 11/28/2023] [Indexed: 01/19/2024]
Abstract
The plasmids from the Université d'Ottawa (pUdOs) are 28 small plasmids each comprising one of four origins of replication and one of seven selection markers, which together afford flexible use in Escherichia coli and several related gram-negative bacteria. The promoterless multicloning site is insulated from upstream spurious promoters by strong transcription terminators and contains type IIP or IIS restriction sites for conventional or Golden Gate cloning. pUdOs can be converted into efficient expression vectors through the insertion of a promoter at the user's discretion. For example, we demonstrate the utility of pUdOs as the backbone for an improved version of a Type III Secretion System reporter in Shigella. In addition, we derive a series of pUdO-based mammalian expression vectors, affording distinct levels of expression and transfection efficiency comparable to commonly used mammalian expression plasmids. Thus, pUdOs could advantageously replace traditional plasmids in a wide variety of cell types and applications.
Collapse
Affiliation(s)
- France
O. Manigat
- Center
for Chemical and Synthetic Biology, Department of Chemistry and Biomolecular
Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
- bioGARAGE,
Faculty of Science, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Louise B. Connell
- Center
for Chemical and Synthetic Biology, Department of Chemistry and Biomolecular
Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
- bioGARAGE,
Faculty of Science, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Brittany N. Stewart
- Center
for Chemical and Synthetic Biology, Department of Chemistry and Biomolecular
Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
- bioGARAGE,
Faculty of Science, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Abdel-Rahman LePabic
- Center
for Chemical and Synthetic Biology, Department of Chemistry and Biomolecular
Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
- bioGARAGE,
Faculty of Science, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Christian J. G. Tessier
- Center
for Chemical and Synthetic Biology, Department of Chemistry and Biomolecular
Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
- bioGARAGE,
Faculty of Science, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Johnathon R. Emlaw
- Center
for Chemical and Synthetic Biology, Department of Chemistry and Biomolecular
Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
- bioGARAGE,
Faculty of Science, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Nicholas D. Calvert
- Center
for Chemical and Synthetic Biology, Department of Chemistry and Biomolecular
Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
- bioGARAGE,
Faculty of Science, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Anthony Rössl
- Center
for Chemical and Synthetic Biology, Department of Chemistry and Biomolecular
Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
- bioGARAGE,
Faculty of Science, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Adam J. Shuhendler
- Center
for Chemical and Synthetic Biology, Department of Chemistry and Biomolecular
Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
- bioGARAGE,
Faculty of Science, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
- University
of Ottawa Heart Institute, Ottawa, Ontario K1Y 4W7, Canada
| | - Corrie J. B. daCosta
- Center
for Chemical and Synthetic Biology, Department of Chemistry and Biomolecular
Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
- bioGARAGE,
Faculty of Science, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - François-Xavier Campbell-Valois
- Center
for Chemical and Synthetic Biology, Department of Chemistry and Biomolecular
Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
- bioGARAGE,
Faculty of Science, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
- Centre
for Infection, Immunity and Inflammation, Department of Biochemistry,
Microbiology and Immunology, University
of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| |
Collapse
|
20
|
Jakob S, Steinchen W, Hanßmann J, Rosum J, Langenfeld K, Osorio-Valeriano M, Steube N, Giammarinaro PI, Hochberg GKA, Glatter T, Bange G, Diepold A, Thanbichler M. The virulence regulator VirB from Shigella flexneri uses a CTP-dependent switch mechanism to activate gene expression. Nat Commun 2024; 15:318. [PMID: 38182620 PMCID: PMC10770331 DOI: 10.1038/s41467-023-44509-z] [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: 06/02/2023] [Accepted: 12/11/2023] [Indexed: 01/07/2024] Open
Abstract
The transcriptional antisilencer VirB acts as a master regulator of virulence gene expression in the human pathogen Shigella flexneri. It binds DNA sequences (virS) upstream of VirB-dependent promoters and counteracts their silencing by the nucleoid-organizing protein H-NS. However, its precise mode of action remains unclear. Notably, VirB is not a classical transcription factor but related to ParB-type DNA-partitioning proteins, which have recently been recognized as DNA-sliding clamps using CTP binding and hydrolysis to control their DNA entry gate. Here, we show that VirB binds CTP, embraces DNA in a clamp-like fashion upon its CTP-dependent loading at virS sites and slides laterally on DNA after clamp closure. Mutations that prevent CTP-binding block VirB loading in vitro and abolish the formation of VirB nucleoprotein complexes as well as virulence gene expression in vivo. Thus, VirB represents a CTP-dependent molecular switch that uses a loading-and-sliding mechanism to control transcription during bacterial pathogenesis.
Collapse
Affiliation(s)
- Sara Jakob
- Department of Biology, University of Marburg, Marburg, Germany
| | - Wieland Steinchen
- Department of Chemistry, University of Marburg, Marburg, Germany
- Center for Synthetic Microbiology (SYNMIKRO), Marburg, Germany
| | - Juri Hanßmann
- Department of Biology, University of Marburg, Marburg, Germany
- Max Planck Fellow Group Bacterial Cell Biology, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Julia Rosum
- Department of Biology, University of Marburg, Marburg, Germany
| | - Katja Langenfeld
- Department of Ecophysiology, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Manuel Osorio-Valeriano
- Department of Biology, University of Marburg, Marburg, Germany
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Niklas Steube
- Evolutionary Biochemistry Group, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Pietro I Giammarinaro
- Department of Chemistry, University of Marburg, Marburg, Germany
- Heidelberg University Biochemistry Center (BZH), Heidelberg, Germany
| | - Georg K A Hochberg
- Department of Chemistry, University of Marburg, Marburg, Germany
- Center for Synthetic Microbiology (SYNMIKRO), Marburg, Germany
- Evolutionary Biochemistry Group, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Timo Glatter
- Mass Spectrometry and Proteomics Facility, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Gert Bange
- Department of Chemistry, University of Marburg, Marburg, Germany
- Center for Synthetic Microbiology (SYNMIKRO), Marburg, Germany
- Max Planck Fellow Group Molecular Physiology of Microbes, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Andreas Diepold
- Department of Ecophysiology, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany
| | - Martin Thanbichler
- Department of Biology, University of Marburg, Marburg, Germany.
- Center for Synthetic Microbiology (SYNMIKRO), Marburg, Germany.
- Max Planck Fellow Group Bacterial Cell Biology, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany.
| |
Collapse
|
21
|
León Y, Honigsberg R, Rasko DA, Faherty CS. Gastrointestinal signals in supplemented media reveal a role in adherence for the Shigella flexneri sap autotransporter gene. Gut Microbes 2024; 16:2331985. [PMID: 38549437 PMCID: PMC10984119 DOI: 10.1080/19490976.2024.2331985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 03/14/2024] [Indexed: 04/02/2024] Open
Abstract
Shigella flexneri causes severe diarrheal disease worldwide. While many aspects of pathogenesis have been elucidated, significant knowledge gaps remain regarding the role of putative chromosomally-encoded virulence genes. The uncharacterized sap gene encoded on the chromosome has significant nucleotide sequence identity to the fluffy (flu) antigen 43 autotransporter gene in pathogenic Escherichia coli. Here, we constructed a Δsap mutant in S. flexneri strain 2457T and examined the effects of this mutation on bacterial cell aggregation, biofilm formation, and adherence to colonic epithelial cells. Analyses included the use of growth media supplemented with glucose and bile salts to replicate small intestinal signals encountered by S. flexneri. Deletion of the sap gene in 2457T affected epithelial cell adherence, resulted in quicker bacterial cell aggregation, but did not affect biofilm formation. This work highlights a functional role for the sap gene in S. flexneri pathogenesis and further demonstrates the importance of using relevant and appropriate gastrointestinal signals to characterize virulence genes of enteropathogenic bacteria.
Collapse
Affiliation(s)
- Yrvin León
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Raphael Honigsberg
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, USA
- École Normale Supérieure Paris-Saclay, Département d’Enseignement et de, Recherche de Biologie, Université Paris-Saclay, Gif-sur-Yvette, France
| | - David A. Rasko
- Institute for Genome Sciences, Center for Pathogen Research, Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Christina S. Faherty
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
22
|
Torraca V, White RJ, Sealy IM, Mazon-Moya M, Duggan G, Willis AR, Busch-Nentwich EM, Mostowy S. Transcriptional profiling of zebrafish identifies host factors controlling susceptibility to Shigella flexneri. Dis Model Mech 2024; 17:dmm050431. [PMID: 38131137 PMCID: PMC10846535 DOI: 10.1242/dmm.050431] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023] Open
Abstract
Shigella flexneri is a human-adapted pathovar of Escherichia coli that can invade the intestinal epithelium, causing inflammation and bacillary dysentery. Although an important human pathogen, the host response to S. flexneri has not been fully described. Zebrafish larvae represent a valuable model for studying human infections in vivo. Here, we use a Shigella-zebrafish infection model to generate mRNA expression profiles of host response to Shigella infection at the whole-animal level. Immune response-related processes dominate the signature of early Shigella infection (6 h post-infection). Consistent with its clearance from the host, the signature of late Shigella infection (24 h post-infection) is significantly changed, and only a small set of immune-related genes remain differentially expressed, including acod1 and gpr84. Using mutant lines generated by ENU, CRISPR mutagenesis and F0 crispants, we show that acod1- and gpr84-deficient larvae are more susceptible to Shigella infection. Together, these results highlight the power of zebrafish to model infection by bacterial pathogens and reveal the mRNA expression of the early (acutely infected) and late (clearing) host response to Shigella infection.
Collapse
Affiliation(s)
- Vincenzo Torraca
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK
- Section of Microbiology, MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London SW7 2AZ, UK
- Department of Infectious Diseases, School of Immunology and Microbial Sciences, King's College London, London SE1 9RT, UK
- School of Life Sciences, University of Westminster, London W1W 6UW, UK
| | - Richard J. White
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge CB2 0AW, UK
- School of Biological and Behavioural Sciences, Faculty of Science and Engineering, Queen Mary University of London, London E1 4NS, UK
| | - Ian M. Sealy
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge CB2 0AW, UK
- School of Biological and Behavioural Sciences, Faculty of Science and Engineering, Queen Mary University of London, London E1 4NS, UK
| | - Maria Mazon-Moya
- Section of Microbiology, MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London SW7 2AZ, UK
| | - Gina Duggan
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK
- Section of Microbiology, MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London SW7 2AZ, UK
| | - Alexandra R. Willis
- Section of Microbiology, MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London SW7 2AZ, UK
| | - Elisabeth M. Busch-Nentwich
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge CB2 0AW, UK
- School of Biological and Behavioural Sciences, Faculty of Science and Engineering, Queen Mary University of London, London E1 4NS, UK
| | - Serge Mostowy
- Department of Infection Biology, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK
- Section of Microbiology, MRC Centre for Molecular Bacteriology and Infection, Imperial College London, London SW7 2AZ, UK
| |
Collapse
|
23
|
Li H, Liu S, Zhang K, Zhu X, Dai J, Lu Y. Gut microbiome and plasma metabolome alterations in myopic mice. Front Microbiol 2023; 14:1251243. [PMID: 38179454 PMCID: PMC10764480 DOI: 10.3389/fmicb.2023.1251243] [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: 07/05/2023] [Accepted: 11/30/2023] [Indexed: 01/06/2024] Open
Abstract
Background Myopia is one of the most common eye diseases leading to blurred distance vision. Inflammatory diseases could trigger or exacerbate myopic changes. Although gut microbiota bacteria are associated with various inflammatory diseases, little is known about its role in myopia. Materials and methods The mice were randomly divided into control and model groups, with the model group being attached-30D lens onto the eyes for 3 weeks. Then, mouse cecal contents and plasma were collected to analyze their intestinal microbiota and plasma metabolome. Results We identified that the microbial composition differed considerably between the myopic and non-myopic mice, with the relative abundance of Firmicutes phylum decreased obviously while that of Actinobacteria phylum was increased in myopia. Furthermore, Actinobacteria and Bifidobacterium were positively correlated with axial lengths (ALs) of eyeballs while negatively correlated with refractive diopters. Untargeted metabolomic analysis identified 141 differentially expressed metabolites, and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis revealed considerable enrichment mainly in amino acid metabolism pathways. Notably, pathways involved glutamate metabolism including "Glutamine and D-glutamate metabolism" and "Alanine, aspartate and glutamate metabolism" was changed dramatically, which presented as the concentrations of L-Glutamate and L-Glutamine decreased obviously in myopia. Interestingly, microbiome dysbiosis and metabolites alternations in myopia have a disrupting gut barrier feature. We further demonstrated that the gut barrier function was impaired in myopic mice manifesting in decreased expression of Occludin, ZO-1 and increased permeation of FITC-dextran. Discussion Myopic mice had obviously altered gut microbiome and metabolites profiles compared to non-myopic mice. The dysbiosis and plasma metabolomics shift in myopia had an interrupting gut barrier feature. Our study provides new insights into the possible role of the gut microbiota in myopia and reinforces the potential feasibility of microbiome-based therapies in myopia.
Collapse
Affiliation(s)
- Hao Li
- Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, China
- Eye Institute, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia (Fudan University), Shanghai, China
- Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Shuyu Liu
- Eye Institute, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia (Fudan University), Shanghai, China
- Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Keke Zhang
- Eye Institute, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia (Fudan University), Shanghai, China
- Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Xiangjia Zhu
- Eye Institute, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia (Fudan University), Shanghai, China
- Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| | - Jinhui Dai
- Department of Ophthalmology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yi Lu
- Eye Institute, Eye & ENT Hospital, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia (Fudan University), Shanghai, China
- Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai, China
| |
Collapse
|
24
|
Ascari A, Frölich S, Zang M, Tran ENH, Wilson DW, Morona R, Eijkelkamp BA. Shigella flexneri remodeling and consumption of host lipids during infection. J Bacteriol 2023; 205:e0032023. [PMID: 37991380 PMCID: PMC10729657 DOI: 10.1128/jb.00320-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 11/10/2023] [Indexed: 11/23/2023] Open
Abstract
IMPORTANCE Bacterial pathogens have vastly distinct sites that they inhabit during infection. This requires adaptation due to changes in nutrient availability and antimicrobial stress. The bacterial surface is a primary barrier, and here, we show that the bacterial pathogen Shigella flexneri increases its surface decorations when it transitions to an intracellular lifestyle. We also observed changes in bacterial and host cell fatty acid homeostasis. Specifically, intracellular S. flexneri increased the expression of their fatty acid degradation pathway, while the host cell lipid pool was significantly depleted. Importantly, bacterial proliferation could be inhibited by fatty acid supplementation of host cells, thereby providing novel insights into the possible link between human malnutrition and susceptibility to S. flexneri.
Collapse
Affiliation(s)
- Alice Ascari
- Department of Molecular and Biomedical Science, School of Biological Sciences, Research Centre for Infectious Diseases, University of Adelaide, Adelaide, Australia
- Molecular Sciences and Technology, College of Science and Engineering, Flinders University, Adelaide, Australia
| | - Sonja Frölich
- Department of Molecular and Biomedical Science, School of Biological Sciences, Research Centre for Infectious Diseases, University of Adelaide, Adelaide, Australia
- Institute for Photonics and Advanced Sensing (IPAS), University of Adelaide, Adelaide, Australia
| | - Maoge Zang
- Molecular Sciences and Technology, College of Science and Engineering, Flinders University, Adelaide, Australia
| | - Elizabeth N. H. Tran
- Department of Molecular and Biomedical Science, School of Biological Sciences, Research Centre for Infectious Diseases, University of Adelaide, Adelaide, Australia
| | - Danny W. Wilson
- Department of Molecular and Biomedical Science, School of Biological Sciences, Research Centre for Infectious Diseases, University of Adelaide, Adelaide, Australia
- Institute for Photonics and Advanced Sensing (IPAS), University of Adelaide, Adelaide, Australia
| | - Renato Morona
- Department of Molecular and Biomedical Science, School of Biological Sciences, Research Centre for Infectious Diseases, University of Adelaide, Adelaide, Australia
| | - Bart A. Eijkelkamp
- Molecular Sciences and Technology, College of Science and Engineering, Flinders University, Adelaide, Australia
| |
Collapse
|
25
|
Perruzza L, Zagaglia C, Vitiello L, Sarshar M, Strati F, Pasqua M, Grassi F, Nicoletti M, Palamara AT, Ambrosi C, Scribano D. The Shigella flexneri virulence factor apyrase is released inside eukaryotic cells to hijack host cell fate. Microbiol Spectr 2023; 11:e0077523. [PMID: 37795996 PMCID: PMC10714728 DOI: 10.1128/spectrum.00775-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 08/19/2023] [Indexed: 10/06/2023] Open
Abstract
IMPORTANCE In this paper, we demonstrated that apyrase is released within the host cell cytoplasm during infection to target the intracellular ATP pool. By degrading intracellular ATP, apyrase contributes to prevent caspases activation, thereby inhibiting the activation of pyroptosis in infected cells. Our results show, for the first time, that apyrase is involved in the modulation of host cell survival, thereby aiding this pathogen to dampen the inflammatory response. This work adds a further piece to the puzzle of Shigella pathogenesis. Due to its increased spread worldwide, prevention and controlling strategies are urgently needed. Overall, this study highlighted apyrase as a suitable target for an anti-virulence therapy to tackle this pathogen.
Collapse
Affiliation(s)
- Lisa Perruzza
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona, Switzerland
- Humabs BioMed, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Carlo Zagaglia
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Laura Vitiello
- Laboratory of Flow Cytometry, IRCCS San Raffaele Roma, Rome, Italy
| | - Meysam Sarshar
- Research Laboratories, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Francesco Strati
- Mucosal Immunology Lab, Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Martina Pasqua
- Institute Pasteur Italy, Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, Rome, Italy
| | - Fabio Grassi
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Mauro Nicoletti
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Anna Teresa Palamara
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Laboratory Affiliated to Institute Pasteur Italia-Cenci Bolognetti Foundation, Rome, Italy
- Department Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Cecilia Ambrosi
- Department of Human Sciences and Quality of Life Promotion, San Raffaele University, Rome, Italy
- Laboratory of Microbiology of Chronic-Neurodegenerative Diseases, IRCCS San Raffaele Roma, Rome, Italy
| | - Daniela Scribano
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
26
|
Perlman M, Senger S, Verma S, Carey J, Faherty CS. A foundational approach to culture and analyze malnourished organoids. Gut Microbes 2023; 15:2248713. [PMID: 37724815 PMCID: PMC10512930 DOI: 10.1080/19490976.2023.2248713] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 08/07/2023] [Indexed: 09/21/2023] Open
Abstract
The gastrointestinal (GI) epithelium plays a major role in nutrient absorption, barrier formation, and innate immunity. The development of organoid-based methodology has significantly impacted the study of the GI epithelium, particularly in the fields of mucosal biology, immunity, and host-microbe interactions. Various effects on the GI epithelium, such as genetics and nutrition, impact patients and alter disease states. Thus, incorporating these effects into organoid-based models will facilitate a better understanding of disease progression and offer opportunities to evaluate therapeutic candidates. One condition that has a significant effect on the GI epithelium is malnutrition, and studying the mechanistic impacts of malnutrition would enhance our understanding of several pathologies. Therefore, the goal of this study was to begin to develop methodology to generate viable malnourished organoids with accessible techniques and resources that can be used for a wide array of mechanistic studies. By selectively limiting distinct macronutrient components of organoid media, we were able to successfully culture and evaluate malnourished organoids. Genetic and protein-based analyses were used to validate the approach and confirm the presence of known biomarkers of malnutrition. Additionally, as proof-of-concept, we utilized malnourished organoid-derived monolayers to evaluate the effect of malnourishment on barrier formation and the ability of the bacterial pathogen Shigella flexneri to infect the GI epithelium. This work serves as the basis for new and exciting techniques to alter the nutritional state of organoids and investigate the related impacts on the GI epithelium.
Collapse
Affiliation(s)
- Meryl Perlman
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Stefania Senger
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, MA, USA
| | - Smriti Verma
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA
| | - James Carey
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, MA, USA
| | - Christina S. Faherty
- Mucosal Immunology and Biology Research Center, Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
27
|
唐 智, 符 立, 刘 人, 陈 昱, 别 明, 王 保. [Mechanisms of Helicobacter pylori Intracellular Infection and Reflections Concerning Clinical Practice]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2023; 54:1300-1305. [PMID: 38162071 PMCID: PMC10752795 DOI: 10.12182/20231160401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Indexed: 01/03/2024]
Abstract
Helicobacter pylori (H. pylori), for a long time, has generally been considered an extracellular bacterium. However, recent findings have shown that H. pylori can gain entry into host cells, evade attacks from the host immune system and the killing ability of medication, form stable intracellular ecological niche, and achieve re-release into the extracellular environment, thus causing recurrent infections. H. pylori intracellular infection causes cellular signaling and metabolic alterations, which may be closely associated with the pathogenesis and progression of tumors, thereby presenting new challenges for clinical eradicative treatment of H. pylori. Herein, examining this issue from a clinical perspective, we reviewed reported findings on the mechanisms of how H. pylori achieved intracellular infection, including the breaching of the host cell biological barrier, immune evasion, and resistance to autophagy. In addition, we discussed our reflections and the prospects of important questions concerning H. pylori, including the clinical prevention and control strategy, intracellular derivation, and the damage to host cells.
Collapse
Affiliation(s)
- 智慧 唐
- 四川大学华西基础医学与法医学院 (成都 610041)West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - 立发 符
- 四川大学华西基础医学与法医学院 (成都 610041)West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - 人捷 刘
- 四川大学华西基础医学与法医学院 (成都 610041)West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - 昱作 陈
- 四川大学华西基础医学与法医学院 (成都 610041)West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - 明江 别
- 四川大学华西基础医学与法医学院 (成都 610041)West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China
- 四川大学华西公共卫生学院/四川大学华西第四医院 (成都 610041)West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu 610041, China
| | - 保宁 王
- 四川大学华西基础医学与法医学院 (成都 610041)West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu 610041, China
| |
Collapse
|
28
|
Miljkovic M, Lozano S, Castellote I, de Cózar C, Villegas-Moreno AI, Gamallo P, Jimenez-Alfaro Martinez D, Fernández-Álvaro E, Ballell L, Garcia GA. Novel inhibitors that target bacterial virulence identified via HTS against intra-macrophage survival of Shigella flexneri. mSphere 2023; 8:e0015423. [PMID: 37565760 PMCID: PMC10597453 DOI: 10.1128/msphere.00154-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 06/02/2023] [Indexed: 08/12/2023] Open
Abstract
Shigella flexneri is a facultative intracellular pathogen that causes shigellosis, a human diarrheal disease characterized by the destruction of the colonic epithelium. Novel antimicrobial compounds to treat infections are urgently needed due to the proliferation of bacterial antibiotic resistance and lack of new effective antimicrobials in the market. Our approach to find compounds that block the Shigella virulence pathway has three potential advantages: (i) resistance development should be minimized due to the lack of growth selection pressure, (ii) no resistance due to environmental antibiotic exposure should be developed since the virulence pathways are not activated outside of host infection, and (iii) the normal intestinal microbiota, which do not have the targeted virulence pathways, should be unharmed. We chose to utilize two phenotypic assays, inhibition of Shigella survival in macrophages and Shigella growth inhibition (minimum inhibitory concentration), to interrogate the 1.7 M compound screening collection subset of the GlaxoSmithKline drug discovery chemical library. A number of secondary assays on the hit compounds resulting from the primary screens were conducted, which, in combination with chemical, structural, and physical property analyses, narrowed the final hit list to 44 promising compounds for further drug discovery efforts. The rapid development of antibiotic resistance is a critical problem that has the potential of returning the world to a "pre-antibiotic" type of environment, where millions of people will die from previously treatable infections. One relatively newer approach to minimize the selection pressures for the development of resistance is to target virulence pathways. This is anticipated to eliminate any resistance selection pressure in environmental exposure to virulence-targeted antibiotics and will have the added benefit of not affecting the non-virulent microbiome. This paper describes the development and application of a simple, reproducible, and sensitive assay to interrogate an extensive chemical library in high-throughput screening format for activity against the survival of Shigella flexneri 2457T-nl in THP-1 macrophages. The ability to screen very large numbers of compounds in a reasonable time frame (~1.7 M compounds in ~8 months) distinguishes this assay as a powerful tool in further exploring new compounds with intracellular effect on S. flexneri or other pathogens with similar pathways of pathogenesis. The assay utilizes a luciferase reporter which is extremely rapid, simple, relatively inexpensive, and sensitive and possesses a broad linear range. The assay also utilized THP-1 cells that resemble primary monocytes and macrophages in morphology and differentiation properties. THP-1 cells have advantages over human primary monocytes or macrophages because they are highly plastic and their homogeneous genetic background minimizes the degree of variability in the cell phenotype (1). The intracellular and virulence-targeted selectivity of our methodology, determined via secondary screening, is an enormous advantage. Our main interest focuses on hits that are targeting virulence, and the most promising compounds with adequate physicochemical and drug metabolism and pharmacokinetic (DMPK) properties will be progressed to a suitable in vivo shigellosis model to evaluate the therapeutic potential of this approach. Additionally, compounds that act via a host-directed mechanism could be a promising source for further research given that it would allow a whole new, specific, and controlled approach to the treatment of diseases caused by some pathogenic bacteria.
Collapse
Affiliation(s)
- Marija Miljkovic
- Department of Medical Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan, USA
- GSK Global Health Unit, Madrid, Spain
| | | | | | | | | | | | | | | | | | - George A. Garcia
- Department of Medical Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
29
|
Misra P, Paunikar VM. Healthy Drinking Water as a Necessity in Developing Countries Like India: A Narrative review. Cureus 2023; 15:e47247. [PMID: 38022361 PMCID: PMC10654688 DOI: 10.7759/cureus.47247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Water is an indispensable part of human life. This article is an extensive review that focuses on the importance of water to sustain human life, the necessity of healthy, safe drinking water, and the health hazards of drinking untreated and contaminated water. We drink treated water thinking it to be safe without the knowledge that it, too, has harmful effects. Detrimental health effects due to water chlorination are mentioned in this article. The usage of nanoparticles for the treatment of water is an alternative to chlorination, but they are little in use as they are expensive. Transmission of waterborne diseases through drinking water is widespread in a developing country like India. A list of the pathogens contaminating drinking water is present in the review. Pathogens pollute water, and heavy metals and chemicals from industries, pesticides, pharmaceutical compounds, and radioactive waste also taint it. The harmful effects of metal and chemical toxicities on human health are discussed in this review. The government of India has launched several programmes to ensure clean, safe drinking water for all its residents. The study reflects on the treatment given to individuals suffering from waterborne diseases in India. Significant changes in health status in India have been seen recently after the execution of various government programmes to provide safe, healthy drinking water to all its residents.
Collapse
Affiliation(s)
- Purbasha Misra
- Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Vaishali M Paunikar
- Physiology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
30
|
Gomes MC, Brokatzky D, Bielecka MK, Wardle FC, Mostowy S. Shigella induces epigenetic reprogramming of zebrafish neutrophils. SCIENCE ADVANCES 2023; 9:eadf9706. [PMID: 37672585 PMCID: PMC10482349 DOI: 10.1126/sciadv.adf9706] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 08/03/2023] [Indexed: 09/08/2023]
Abstract
Trained immunity is a long-term memory of innate immune cells, generating an improved response upon reinfection. Shigella is an important human pathogen and inflammatory paradigm for which there is no effective vaccine. Using zebrafish larvae, we demonstrate that after Shigella training, neutrophils are more efficient at bacterial clearance. We observe that Shigella-induced protection is nonspecific and has differences with training by BCG and β-glucan. Analysis of histone ChIP-seq on trained neutrophils revealed that Shigella training deposits the active H3K4me3 mark on promoter regions of 1612 genes, dramatically changing the epigenetic landscape of neutrophils toward enhanced microbial recognition and mitochondrial ROS production. Last, we demonstrate that mitochondrial ROS plays a key role in enhanced antimicrobial activity of trained neutrophils. It is envisioned that signals and mechanisms we discover here can be used in other vertebrates, including humans, to suggest new therapeutic strategies involving neutrophils to control bacterial infection.
Collapse
Affiliation(s)
- Margarida C. Gomes
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| | - Dominik Brokatzky
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| | - Magdalena K. Bielecka
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| | - Fiona C. Wardle
- Randall Centre for Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, King's College London, UK
| | - Serge Mostowy
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
31
|
Trirocco R, Pasqua M, Tramonti A, Colonna B, Paiardini A, Prosseda G. Diffusible signal factors (DSFs) bind and repress VirF, the leading virulence activator of Shigella flexneri. Sci Rep 2023; 13:13170. [PMID: 37580399 PMCID: PMC10425336 DOI: 10.1038/s41598-023-40023-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 08/03/2023] [Indexed: 08/16/2023] Open
Abstract
Shigella, the aetiological agent of human bacillary dysentery, controls the expression of its virulence determinants through an environmentally stimulated cascade of transcriptional activators. VirF is the leading activator and is essential for proper virulence expression. In this work, we report on in vitro and in vivo experiments showing that two autoinducers of the DSF family, XcDSF and BDSF interact with the jelly roll module of VirF causing its inhibition and affecting the expression of the entire virulence system of Shigella, including its ability to invade epithelial cells. We propose a molecular model explaining how the binding of XcDSF and BDSF causes inhibition of VirF by preventing its dimerization. Overall, our experimental results suggest that XcDSF and BDSF may contribute to "colonisation resistance" in the human gut or, alternatively, may be exploited for the fine-tuning of Shigella virulence expression as the bacterium migrates from the lumen to approach the intestinal mucosa. Our findings also stress how a detailed understanding of the interaction of DSF ligands with VirF may contribute to the rational development of innovative antivirulence drugs to treat shigellosis.
Collapse
Affiliation(s)
- Rita Trirocco
- Institute Pasteur Italia, Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, p.le Aldo Moro 5, 00185, Rome, Italy
| | - Martina Pasqua
- Institute Pasteur Italia, Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, p.le Aldo Moro 5, 00185, Rome, Italy
| | - Angela Tramonti
- Institute of Molecular Biology and Pathology, National Research Council, Rome, Italy
| | - Bianca Colonna
- Institute Pasteur Italia, Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, p.le Aldo Moro 5, 00185, Rome, Italy
| | - Alessandro Paiardini
- Department of Biochemical Sciences, Sapienza University of Rome, p.le Aldo Moro 5, 00185, Rome, Italy
| | - Gianni Prosseda
- Institute Pasteur Italia, Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, p.le Aldo Moro 5, 00185, Rome, Italy.
| |
Collapse
|
32
|
Ascari A, Waters JK, Morona R, Eijkelkamp BA. Shigella flexneri Adapts to Niche-Specific Stresses through Modifications in Cell Envelope Composition and Decoration. ACS Infect Dis 2023; 9:1610-1621. [PMID: 37494550 DOI: 10.1021/acsinfecdis.3c00210] [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] [Indexed: 07/28/2023]
Abstract
Shigella flexneri is the primary causative agent of worldwide shigellosis. As the pathogen transverses the distinct niches of the gastrointestinal tract it necessitates dynamic adaptation strategies to mitigate host antimicrobials such as dietary fatty acids (FAs) and the bile salt, deoxycholate (DOC). This study investigates the dynamics of the S. flexneri cell envelope, by interrogating adaptations following FA or DOC exposure. We deciphered the effects of FAs and DOC on bacterial membrane fatty acid and lipopolysaccharide (LPS) compositions. We identified novel LPS-based strategies by the pathogen to support resistance to these host compounds. In particular, expression of S. flexneri very-long O antigen (VL-Oag) LPS was found to play a central role in stress mitigation, as VL-Oag protects against antimicrobial FAs, but its presence rendered S. flexneri susceptible to DOC stress. Collectively, this work underpins the importance for S. flexneri to maintain appropriate regulation of cell envelope constituents, in particular VL-Oag LPS, to adequately adapt to diverse stresses during infection.
Collapse
Affiliation(s)
- Alice Ascari
- School of Biological Sciences, Department of Molecular and Biomedical Science, Research Centre for Infectious Diseases, University of Adelaide, Adelaide 5005, South Australia, Australia
- Molecular Sciences and Technology, College of Science and Engineering, Flinders University, Adelaide 5042, South Australia, Australia
| | - Jack K Waters
- Molecular Sciences and Technology, College of Science and Engineering, Flinders University, Adelaide 5042, South Australia, Australia
| | - Renato Morona
- School of Biological Sciences, Department of Molecular and Biomedical Science, Research Centre for Infectious Diseases, University of Adelaide, Adelaide 5005, South Australia, Australia
| | - Bart A Eijkelkamp
- Molecular Sciences and Technology, College of Science and Engineering, Flinders University, Adelaide 5042, South Australia, Australia
| |
Collapse
|
33
|
Torraca V, Bielecka MK, Gomes MC, Brokatzky D, Busch‐Nentwich EM, Mostowy S. Zebrafish null mutants of Sept6 and Sept15 are viable but more susceptible to Shigella infection. Cytoskeleton (Hoboken) 2023; 80:266-274. [PMID: 36855298 PMCID: PMC10952258 DOI: 10.1002/cm.21750] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 01/21/2023] [Accepted: 02/24/2023] [Indexed: 03/02/2023]
Abstract
Septins are evolutionarily conserved GTP-binding proteins known for their roles in cell division and host defence against Shigella infection. Although septin group members are viewed to function as hetero-oligomeric complexes, the role of individual septins within these complexes or in isolation is poorly understood. Decades of work using mouse models has shown that some septins (including SEPT7) are essential for animal development, while others (including SEPT6) are dispensable, suggesting that some septins may compensate for the absence of others. The zebrafish genome encodes 19 septin genes, representing the full complement of septin groups described in mice and humans. In this report, we characterise null mutants for zebrafish Sept6 (a member of the SEPT6 group) and Sept15 (a member of the SEPT7 group) and test their role in zebrafish development and host defence against Shigella infection. We show that null mutants for Sept6 and Sept15 are both viable, and that expression of other zebrafish septins are not significantly affected by their mutation. Consistent with previous reports using knockdown of Sept2, Sept7b, and Sept15, we show that Sept6 and Sept15 are required for host defence against Shigella infection. These results highlight Shigella infection of zebrafish as a powerful system to study the role of individual septins in vivo.
Collapse
Affiliation(s)
- Vincenzo Torraca
- Department of Infection BiologyLondon School of Hygiene & Tropical MedicineLondonUK
- School of Life SciencesUniversity of WestminsterLondonUK
| | | | - Margarida C. Gomes
- Department of Infection BiologyLondon School of Hygiene & Tropical MedicineLondonUK
| | - Dominik Brokatzky
- Department of Infection BiologyLondon School of Hygiene & Tropical MedicineLondonUK
| | - Elisabeth M. Busch‐Nentwich
- Department of Medicine, Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID)University of CambridgeCambridgeUK
- School of Biological and Behavioural Sciences, Faculty of Science and EngineeringQueen Mary University of LondonLondonUK
| | - Serge Mostowy
- Department of Infection BiologyLondon School of Hygiene & Tropical MedicineLondonUK
| |
Collapse
|
34
|
Haidar-Ahmad N, Manigat FO, Silué N, Pontier SM, Campbell-Valois FX. A Tale about Shigella: Evolution, Plasmid, and Virulence. Microorganisms 2023; 11:1709. [PMID: 37512882 PMCID: PMC10383432 DOI: 10.3390/microorganisms11071709] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 06/23/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Shigella spp. cause hundreds of millions of intestinal infections each year. They target the mucosa of the human colon and are an important model of intracellular bacterial pathogenesis. Shigella is a pathovar of Escherichia coli that is characterized by the presence of a large invasion plasmid, pINV, which encodes the characteristic type III secretion system and icsA used for cytosol invasion and cell-to-cell spread, respectively. First, we review recent advances in the genetic aspects of Shigella, shedding light on its evolutionary history within the E. coli lineage and its relationship to the acquisition of pINV. We then discuss recent insights into the processes that allow for the maintenance of pINV. Finally, we describe the role of the transcription activators VirF, VirB, and MxiE in the major virulence gene regulatory cascades that control the expression of the type III secretion system and icsA. This provides an opportunity to examine the interplay between these pINV-encoded transcriptional activators and numerous chromosome-encoded factors that modulate their activity. Finally, we discuss novel chromosomal genes icaR, icaT, and yccE that are regulated by MxiE. This review emphasizes the notion that Shigella and E. coli have walked the fine line between commensalism and pathogenesis for much of their history.
Collapse
Affiliation(s)
- Nathaline Haidar-Ahmad
- Host-Microbe Interactions Laboratory, Centre for Chemical and Synthetic Biology, Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - France Ourida Manigat
- Host-Microbe Interactions Laboratory, Centre for Chemical and Synthetic Biology, Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Navoun Silué
- Host-Microbe Interactions Laboratory, Centre for Chemical and Synthetic Biology, Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Stéphanie M Pontier
- Centre de Recherche Santé Environnementale et Biodiversité de l'Outaouais (SEBO), CEGEP de l'Outaouais, Gatineau, QC J8Y 6M4, Canada
| | - François-Xavier Campbell-Valois
- Host-Microbe Interactions Laboratory, Centre for Chemical and Synthetic Biology, Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
- Centre for Infection, Immunity and Inflammation, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| |
Collapse
|
35
|
Wang M, Zeng J, Zhu Y, Chen X, Guo Q, Tan H, Cui B, Song S, Deng Y. A 4-Hydroxybenzoic Acid-Mediated Signaling System Controls the Physiology and Virulence of Shigella sonnei. Microbiol Spectr 2023; 11:e0483522. [PMID: 37036340 PMCID: PMC10269604 DOI: 10.1128/spectrum.04835-22] [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: 11/24/2022] [Accepted: 03/15/2023] [Indexed: 04/11/2023] Open
Abstract
Many bacteria use small molecules, such as quorum sensing (QS) signals, to perform intraspecies signaling and interspecies or interkingdom communication. Previous studies demonstrated that some bacteria regulate their physiology and pathogenicity by employing 4-hydroxybenzoic acid (4-HBA). Here, we report that 4-HBA controls biological functions, virulence, and anthranilic acid production in Shigella sonnei. The biosynthesis of 4-HBA is performed by UbiC (SSON_4219), which is a chorismate pyruvate-lyase that catalyzes the conversion of chorismate to 4-HBA. Deletion of ubiC caused S. sonnei to exhibit impaired phenotypes, including impaired biofilm formation, extracellular polysaccharide (EPS) production, and virulence. In addition, we found that 4-HBA controls the physiology and virulence of S. sonnei through the response regulator AaeR (SSON_3385), which contains a helix-turn-helix (HTH) domain and a LysR substrate-binding (LysR_substrate) domain. The same biological functions are controlled by AaeR and the 4-HBA signal, and 4-HBA-deficient mutant phenotypes were rescued by in trans expression of AaeR. We found that 4-HBA binds to AaeR and then enhances the binding of AaeR to the promoter DNA regions in target genes. Moreover, we revealed that 4-HBA from S. sonnei reduces the competitive fitness of Candida albicans by interfering with morphological transition. Together, our results suggested that the 4-HBA signaling system plays crucial roles in bacterial physiology and interkingdom communication. IMPORTANCE Shigella sonnei is an important pathogen in human intestines. Following previous findings that some bacteria employ 4-HBA as a QS signal to regulate biological functions, we demonstrate that 4-HBA controls the physiology and virulence of S. sonnei. This study is significant because it identifies both the signal synthase UbiC and receptor AaeR and unveils the signaling pathway of 4-HBA in S. sonnei. In addition, this study also supports the important role of 4-HBA in microbial cross talk, as 4-HBA strongly inhibits hyphal formation by Candida albicans. Together, our findings describe the dual roles of 4-HBA in both intraspecies signaling and interkingdom communication.
Collapse
Affiliation(s)
- Mingfang Wang
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Jia Zeng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Yu Zhu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Xiayu Chen
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Quan Guo
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Huihui Tan
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Binbin Cui
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Shihao Song
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
- School of Pharmaceutical Sciences, Hainan University, Haikou, China
| | - Yinyue Deng
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, China
- School of Pharmaceutical Sciences, Hainan University, Haikou, China
| |
Collapse
|
36
|
Baker S, Scott TA. Antimicrobial-resistant Shigella: where do we go next? Nat Rev Microbiol 2023:10.1038/s41579-023-00906-1. [PMID: 37188805 PMCID: PMC10184058 DOI: 10.1038/s41579-023-00906-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Affiliation(s)
- Stephen Baker
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK.
- International AIDS Vaccine Initiative, Chelsea and Westminster Hospital, London, UK.
| | - Timothy A Scott
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge Biomedical Campus, Cambridge, UK
| |
Collapse
|
37
|
Coluccia M, Béranger A, Trirocco R, Fanelli G, Zanzi F, Colonna B, Grossi M, Prosseda G, Pasqua M. Role of the MDR Efflux Pump AcrAB in Epithelial Cell Invasion by Shigella flexneri. Biomolecules 2023; 13:biom13050823. [PMID: 37238693 DOI: 10.3390/biom13050823] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/03/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
The tripartite complex AcrAB-TolC is the major RND pump in Escherichia coli and other Enterobacteriaceae, including Shigella, the etiological agent of bacillary dysentery. In addition to conferring resistance to many classes of antibiotics, AcrAB plays a role in the pathogenesis and virulence of several bacterial pathogens. Here, we report data demonstrating that AcrAB specifically contributes to Shigella flexneri invasion of epithelial cells. We found that deletion of both acrA and acrB genes causes reduced survival of S. flexneri M90T strain within Caco-2 epithelial cells and prevents cell-to-cell spread of the bacteria. Infections with single deletion mutant strains indicate that both AcrA and AcrB favor the viability of the intracellular bacteria. Finally, we were able to further confirm the requirement of the AcrB transporter activity for intraepithelial survival by using a specific EP inhibitor. Overall, the data from the present study expand the role of the AcrAB pump to an important human pathogen, such as Shigella, and add insights into the mechanism governing the Shigella infection process.
Collapse
Affiliation(s)
- Marco Coluccia
- Istituto Pasteur Italy, Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy
| | - Aude Béranger
- Istituto Pasteur Italy, Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy
| | - Rita Trirocco
- Istituto Pasteur Italy, Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy
| | - Giulia Fanelli
- Istituto Pasteur Italy, Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy
| | - Francesco Zanzi
- Istituto Pasteur Italy, Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy
| | - Bianca Colonna
- Istituto Pasteur Italy, Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy
| | - Milena Grossi
- Istituto Pasteur Italy, Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy
| | - Gianni Prosseda
- Istituto Pasteur Italy, Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy
| | - Martina Pasqua
- Istituto Pasteur Italy, Department of Biology and Biotechnologies "Charles Darwin", Sapienza University of Rome, 00185 Rome, Italy
| |
Collapse
|
38
|
Song J, Zhang Y, Zhu Y, Jin X, Li L, Wang C, Zhou Y, Li Y, Wang D, Hu M. Structural characterization and anti-osteoporosis effects of polysaccharide purified from Eucommia ulmoides Oliver cortex based on its modulation on bone metabolism. Carbohydr Polym 2023; 306:120601. [PMID: 36746570 DOI: 10.1016/j.carbpol.2023.120601] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/28/2022] [Accepted: 01/16/2023] [Indexed: 01/20/2023]
Abstract
EuOCP3, with a molecular weight of 38.1 kDa, is an acidic polysaccharide purified from Eucommia ulmoides Oliver cortex. Herein, we determined that the main backbone of EuOCP3 was predominantly composed of →4)-α-GalpA-(1 → 4)-α-GalpA-(1→, →4)-α-GalpA-(1 → 5)-α-Araf-(1→, →4)-α-GalpA-(1 → 2)-α-Rhap-(1→, and →4)-α-GalpA-(1 → 5)-α-Araf-(1 → 2)-α-Rhap-(1 → repeating blocks, which were connected by →2,3,5)-α-Araf-(1→. The side chains, substituted at C-2 and C-5 of →2,3,5)-α-Araf-(1→, contained T-β-Araf→ and T-β-Araf → 4)-α-GalpA-(1 → residues. In dexamethasone (Dex)-induced osteoporosis (OP) mice, EuOCP3 treatment restored cortical bone thickness, increased mineralized bone area, enhanced the number of osteoblasts, and decreased the number of osteoclasts on the surface of cortical bone. Combining analysis of gut microflora, serum metabolite profiles, and biological detection results, we demonstrated that EuOCP3 regulated the abundance of specific species within the gut microflora, such as g_Dorea and g_Prevotella, and ameliorated oxidative stress. In turn, enhancement of osteogenic function and restoration of bone metabolism via the extracellular signal-regulated kinase (ERK)/c-Jun N-terminal kinase (JNK)/nuclear factor erythroid-2 related factor 2 (Nrf2) signaling pathway was indicated. The current findings contribute to understanding the potential of EuOCP3 in anti-OP treatment.
Collapse
Affiliation(s)
- Jiyu Song
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun 130021, China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun 130021, China.
| | - Yongfeng Zhang
- School of Life Sciences, Jilin University, Changchun 130012, China.
| | - Yanfeng Zhu
- School of Life Sciences, Jilin University, Changchun 130012, China.
| | - Xinghui Jin
- School of Life Sciences, Jilin University, Changchun 130012, China.
| | - Lanzhou Li
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China.
| | - Chunyue Wang
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China.
| | - Ying Zhou
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun 130021, China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun 130021, China.
| | - Yutong Li
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun 130021, China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun 130021, China.
| | - Di Wang
- School of Life Sciences, Jilin University, Changchun 130012, China; Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China.
| | - Min Hu
- Department of Orthodontics, Hospital of Stomatology, Jilin University, Changchun 130021, China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Changchun 130021, China.
| |
Collapse
|
39
|
CRISPR-Cas-Guided Mutagenesis of Chromosome and Virulence Plasmid in Shigella flexneri by Cytosine Base Editing. mSystems 2023; 8:e0104522. [PMID: 36541764 PMCID: PMC9948704 DOI: 10.1128/msystems.01045-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Shigella is a Gram-negative bacterium that invades the human gut epithelium. The resulting infection, shigellosis, is the deadliest bacterial diarrheal disease. Much of the information about the genes dictating the pathophysiology of Shigella, both on the chromosome and the virulence plasmid, was obtained by classical reverse genetics. However, technical limitations of the prevalent mutagenesis techniques restrict the generation of mutants in a single reaction to a small number, preventing large-scale targeted mutagenesis of Shigella and the subsequent assessment of phenotype. We adopted a CRISPR-Cas-dependent approach, where a nickase Cas9 and cytidine deaminase fusion is guided by single guide RNA (sgRNA) to introduce targeted C→T transitions, resulting in internal stop codons and premature termination of translation. In proof-of-principle experiments using an mCherry fluorescent reporter, we were able to generate loss-of-function mutants in both Escherichia coli and Shigella flexneri with up to 100% efficacy. Using a modified fluctuation assay, we determined that under optimized conditions, the frequency of untargeted mutations introduced by the Cas9-deaminase fusion was in the same range as spontaneous mutations, making our method a safe choice for bacterial mutagenesis. Furthermore, we programmed the method to mutate well-characterized chromosomal and plasmid-borne Shigella flexneri genes and found the mutant phenotype to be similar to those of the reported gene deletion mutants, with no apparent polar effects at the phenotype level. This method can be used in a 96-well-plate format to increase the throughput and generate an array of targeted loss-of-function mutants in a few days. IMPORTANCE Loss-of-function mutagenesis is critical in understanding the physiological role of genes. Therefore, high-throughput techniques to generate such mutants are important for facilitating the assessment of gene function at a pace that matches systems biology approaches. However, to our knowledge, no such method was available for generating an array of single gene mutants in an important enteropathogen-Shigella. This pathogen causes high morbidity and mortality in children, and antibiotic-resistant strains are quickly emerging. Therefore, determination of the function of unknown Shigella genes is of the utmost importance to develop effective strategies to control infections. Our present work will bridge this gap by providing a rapid method for generating loss-of-function mutants. The highly effective and specific method has the potential to be programmed to generate multiple mutants in a single, massively parallel reaction. By virtue of plasmid compatibility, this method can be extended to other members of Enterobacteriaceae.
Collapse
|
40
|
Mancini F, Alfini R, Caradonna V, Monaci V, Carducci M, Gasperini G, Piccioli D, Biagini M, Giannelli C, Rossi O, Pizza M, Micoli F. Exploring the Role of GMMA Components in the Immunogenicity of a 4-Valent Vaccine against Shigella. Int J Mol Sci 2023; 24:2742. [PMID: 36769063 PMCID: PMC9916818 DOI: 10.3390/ijms24032742] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/24/2023] [Accepted: 01/26/2023] [Indexed: 02/04/2023] Open
Abstract
Shigellosis is the leading cause of diarrheal disease, especially in children of low- and middle-income countries, and is often associated with anti-microbial resistance. Currently, there are no licensed vaccines widely available against Shigella, but several candidates based on the O-antigen (OAg) portion of lipopolysaccharides are in development. We have proposed Generalized Modules for Membrane Antigens (GMMA) as an innovative delivery system for OAg, and a quadrivalent vaccine candidate containing GMMA from S. sonnei and three prevalent S. flexneri serotypes (1b, 2a and 3a) is moving to a phase II clinical trial, with the aim to elicit broad protection against Shigella. GMMA are able to induce anti-OAg-specific functional IgG responses in animal models and healthy adults. We have previously demonstrated that antibodies against protein antigens are also generated upon immunization with S. sonnei GMMA. In this work, we show that a quadrivalent Shigella GMMA-based vaccine is able to promote a humoral response against OAg and proteins of all GMMA types contained in the investigational vaccine. Proteins contained in GMMA provide T cell help as GMMA elicit a stronger anti-OAg IgG response in wild type than in T cell-deficient mice. Additionally, we observed that only the trigger of Toll-like Receptor (TLR) 4 and not of TLR2 contributed to GMMA immunogenicity. In conclusion, when tested in mice, GMMA of a quadrivalent Shigella vaccine candidate combine both adjuvant and carrier activities which allow an increase in the low immunogenic properties of carbohydrate antigens.
Collapse
Affiliation(s)
- Francesca Mancini
- GSK Vaccines Institute for Global Health (GVGH), via Fiorentina 1, 53100 Siena, Italy
| | - Renzo Alfini
- GSK Vaccines Institute for Global Health (GVGH), via Fiorentina 1, 53100 Siena, Italy
| | - Valentina Caradonna
- GSK Vaccines Institute for Global Health (GVGH), via Fiorentina 1, 53100 Siena, Italy
| | - Valentina Monaci
- GSK Vaccines Institute for Global Health (GVGH), via Fiorentina 1, 53100 Siena, Italy
| | - Martina Carducci
- GSK Vaccines Institute for Global Health (GVGH), via Fiorentina 1, 53100 Siena, Italy
| | - Gianmarco Gasperini
- GSK Vaccines Institute for Global Health (GVGH), via Fiorentina 1, 53100 Siena, Italy
| | | | | | - Carlo Giannelli
- GSK Vaccines Institute for Global Health (GVGH), via Fiorentina 1, 53100 Siena, Italy
| | - Omar Rossi
- GSK Vaccines Institute for Global Health (GVGH), via Fiorentina 1, 53100 Siena, Italy
| | - Mariagrazia Pizza
- GSK Vaccines Institute for Global Health (GVGH), via Fiorentina 1, 53100 Siena, Italy
| | - Francesca Micoli
- GSK Vaccines Institute for Global Health (GVGH), via Fiorentina 1, 53100 Siena, Italy
| |
Collapse
|
41
|
Pokharel P, Dhakal S, Dozois CM. The Diversity of Escherichia coli Pathotypes and Vaccination Strategies against This Versatile Bacterial Pathogen. Microorganisms 2023; 11:344. [PMID: 36838308 PMCID: PMC9965155 DOI: 10.3390/microorganisms11020344] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 02/03/2023] Open
Abstract
Escherichia coli (E. coli) is a gram-negative bacillus and resident of the normal intestinal microbiota. However, some E. coli strains can cause diseases in humans, other mammals and birds ranging from intestinal infections, for example, diarrhea and dysentery, to extraintestinal infections, such as urinary tract infections, respiratory tract infections, meningitis, and sepsis. In terms of morbidity and mortality, pathogenic E. coli has a great impact on public health, with an economic cost of several billion dollars annually worldwide. Antibiotics are not usually used as first-line treatment for diarrheal illness caused by E. coli and in the case of bloody diarrhea, antibiotics are avoided due to the increased risk of hemolytic uremic syndrome. On the other hand, extraintestinal infections are treated with various antibiotics depending on the site of infection and susceptibility testing. Several alarming papers concerning the rising antibiotic resistance rates in E. coli strains have been published. The silent pandemic of multidrug-resistant bacteria including pathogenic E. coli that have become more difficult to treat favored prophylactic approaches such as E. coli vaccines. This review provides an overview of the pathogenesis of different pathotypes of E. coli, the virulence factors involved and updates on the major aspects of vaccine development against different E. coli pathotypes.
Collapse
Affiliation(s)
- Pravil Pokharel
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique (INRS), 531 Boul des Prairies, Laval, QC H7V 1B7, Canada
- Centre de Recherche en Infectiologie Porcine et Avicole (CRIPA), Faculté de Médecine Vétérinaire, Université de Montréal Saint-Hyacinthe, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Sabin Dhakal
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique (INRS), 531 Boul des Prairies, Laval, QC H7V 1B7, Canada
- Centre de Recherche en Infectiologie Porcine et Avicole (CRIPA), Faculté de Médecine Vétérinaire, Université de Montréal Saint-Hyacinthe, Saint-Hyacinthe, QC J2S 2M2, Canada
| | - Charles M. Dozois
- Centre Armand-Frappier Santé Biotechnologie, Institut National de la Recherche Scientifique (INRS), 531 Boul des Prairies, Laval, QC H7V 1B7, Canada
- Centre de Recherche en Infectiologie Porcine et Avicole (CRIPA), Faculté de Médecine Vétérinaire, Université de Montréal Saint-Hyacinthe, Saint-Hyacinthe, QC J2S 2M2, Canada
- Pasteur Network, Laval, QC H7V 1B7, Canada
| |
Collapse
|
42
|
Pakbin B, Brück WM, Brück TB. Molecular Mechanisms of Shigella Pathogenesis; Recent Advances. Int J Mol Sci 2023; 24:2448. [PMID: 36768771 PMCID: PMC9917014 DOI: 10.3390/ijms24032448] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/21/2023] [Accepted: 01/23/2023] [Indexed: 01/28/2023] Open
Abstract
Shigella species are the main cause of bacillary diarrhoea or shigellosis in humans. These organisms are the inhabitants of the human intestinal tract; however, they are one of the main concerns in public health in both developed and developing countries. In this study, we reviewed and summarised the previous studies and recent advances in molecular mechanisms of pathogenesis of Shigella Dysenteriae and non-Dysenteriae species. Regarding the molecular mechanisms of pathogenesis and the presence of virulence factor encoding genes in Shigella strains, species of this bacteria are categorised into Dysenteriae and non-Dysenteriae clinical groups. Shigella species uses attachment, invasion, intracellular motility, toxin secretion and host cell interruption mechanisms, causing mild diarrhoea, haemorrhagic colitis and haemolytic uremic syndrome diseases in humans through the expression of effector delivery systems, protein effectors, toxins, host cell immune system evasion and iron uptake genes. The investigation of these genes and molecular mechanisms can help us to develop and design new methods to detect and differentiate these organisms in food and clinical samples and determine appropriate strategies to prevent and treat the intestinal and extraintestinal infections caused by these enteric pathogens.
Collapse
Affiliation(s)
- Babak Pakbin
- Werner Siemens Chair of Synthetic Biotechnology, Department of Chemistry, Technical University of Munich (TUM), Lichtenberg Str. 4, 85748 Garching bei München, Germany
- Institute for Life Technologies, University of Applied Sciences Western Switzerland Valais-Wallis, 1950 Sion, Switzerland
| | - Wolfram Manuel Brück
- Institute for Life Technologies, University of Applied Sciences Western Switzerland Valais-Wallis, 1950 Sion, Switzerland
| | - Thomas B. Brück
- Werner Siemens Chair of Synthetic Biotechnology, Department of Chemistry, Technical University of Munich (TUM), Lichtenberg Str. 4, 85748 Garching bei München, Germany
| |
Collapse
|
43
|
Roncaioli JL, Babirye JP, Chavez RA, Liu FL, Turcotte EA, Lee AY, Lesser CF, Vance RE. A hierarchy of cell death pathways confers layered resistance to shigellosis in mice. eLife 2023; 12:e83639. [PMID: 36645406 PMCID: PMC9876568 DOI: 10.7554/elife.83639] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 01/15/2023] [Indexed: 01/17/2023] Open
Abstract
Bacteria of the genus Shigella cause shigellosis, a severe gastrointestinal disease driven by bacterial colonization of colonic intestinal epithelial cells. Vertebrates have evolved programmed cell death pathways that sense invasive enteric pathogens and eliminate their intracellular niche. Previously we reported that genetic removal of one such pathway, the NAIP-NLRC4 inflammasome, is sufficient to convert mice from resistant to susceptible to oral Shigella flexneri challenge (Mitchell et al., 2020). Here, we investigate the protective role of additional cell death pathways during oral mouse Shigella infection. We find that the Caspase-11 inflammasome, which senses Shigella LPS, restricts Shigella colonization of the intestinal epithelium in the absence of NAIP-NLRC4. However, this protection is limited when Shigella expresses OspC3, an effector that antagonizes Caspase-11 activity. TNFα, a cytokine that activates Caspase-8-dependent apoptosis, also provides potent protection from Shigella colonization of the intestinal epithelium when mice lack both NAIP-NLRC4 and Caspase-11. The combined genetic removal of Caspases-1, -11, and -8 renders mice hyper-susceptible to oral Shigella infection. Our findings uncover a layered hierarchy of cell death pathways that limit the ability of an invasive gastrointestinal pathogen to cause disease.
Collapse
Affiliation(s)
- Justin L Roncaioli
- Division of Immunology & Molecular Medicine, Department of Molecular & Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Janet Peace Babirye
- Division of Immunology & Molecular Medicine, Department of Molecular & Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Roberto A Chavez
- Division of Immunology & Molecular Medicine, Department of Molecular & Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Fitty L Liu
- Division of Immunology & Molecular Medicine, Department of Molecular & Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Elizabeth A Turcotte
- Division of Immunology & Molecular Medicine, Department of Molecular & Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Angus Y Lee
- Cancer Research Laboratory, University of California, BerkeleyBerkeleyUnited States
| | - Cammie F Lesser
- Department of Microbiology, Harvard Medical SchoolBostonUnited States
- Broad Institute of Harvard and MITCambridgeUnited States
- Department of Medicine, Division of Infectious Diseases, Massachusetts General HospitalBostonUnited States
| | - Russell E Vance
- Division of Immunology & Molecular Medicine, Department of Molecular & Cell Biology, University of California, BerkeleyBerkeleyUnited States
- Cancer Research Laboratory, University of California, BerkeleyBerkeleyUnited States
- Immunotherapeutics and Vaccine Research Initiative, University of California, BerkeleyBerkeleyUnited States
- Howard Hughes Medical Institute, University of California, BerkeleyBerkeleyUnited States
| |
Collapse
|
44
|
Han S, Zhuang J, Pan Y, Wu W, Ding K. Different Characteristics in Gut Microbiome between Advanced Adenoma Patients and Colorectal Cancer Patients by Metagenomic Analysis. Microbiol Spectr 2022; 10:e0159322. [PMID: 36453905 PMCID: PMC9769752 DOI: 10.1128/spectrum.01593-22] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 10/25/2022] [Indexed: 12/03/2022] Open
Abstract
The occurrence and development of colorectal cancer (CRC) and advanced adenoma (AA) are closely related to the gut microbiome, and AA has a high cancerization progression rate to CRC. Current studies have revealed that bacteriological analysis cannot identify CRC from AA. The objective was to explore microbial targets that could identify CRC and AA from a microecological perspective and to figure out the best way to identify CRC based on fecal microbes. The metagenomic sequencing data were used to describe the gut microbiome profile and analyze the differences between microbial abundance and microbial single nucleotide polymorphism (SNP) characteristics in AA and CRC patients. It was found that there were no significant differences in the diversity between the two groups. The abundance of bacteria (e.g., Firmicutes, Clostridia, and Blautia), fungi (Hypocreales), archaea (Methanosarcina, Methanoculleus, and Methanolacinia), and viruses (Alphacoronavirus, Sinsheimervirus, and Gammaretrovirus) differed between AA and CRC patients. Multiple machine-learning algorithms were used to establish prediction models, aiming to identify CRC and AA. The accuracy of the random forest (RF) model based on the gut microbiome was 86.54%. Nevertheless, the accuracy of SNP was 92.31% in identifying CRC from AA. In conclusion, using microbial SNP was the best method to identify CRC, it was superior to using the gut microbiome, and it could provide new targets for CRC screening. IMPORTANCE There are differences in characteristic microorganisms between AA and CRC. However, current studies have indicated that bacteriological analysis cannot identify CC from AA, and thus, we wondered if there were some other targets that could be used to identify CRC from AA in the gut microbiome. The differences of SNPs in the gut microbiota of intraindividuals were significantly smaller than those of interindividuals. In addition, compared with intestinal microbes, SNP was less affected by time with certain stability. It was discovered that microbial SNP was better than the gut microbiome for identifying CRC from AA. Therefore, screening characteristic microbial SNP could provide a new research direction for identifying CRC from AA.
Collapse
Affiliation(s)
- Shuwen Han
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang Provincial Clinical Research Center for Cancer, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Medical Oncology, Huzhou Central Hospital, Huzhou, Zhejiang, China
| | - Jing Zhuang
- Department of Medical Oncology, Huzhou Central Hospital, Huzhou, Zhejiang, China
| | - Yuefen Pan
- Department of Medical Oncology, Huzhou Central Hospital, Huzhou, Zhejiang, China
| | - Wei Wu
- Department of Medical Oncology, Huzhou Central Hospital, Huzhou, Zhejiang, China
| | - Kefeng Ding
- Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Zhejiang Provincial Clinical Research Center for Cancer, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
45
|
Lim HJ, Wubben JM, Garcia CP, Cruz-Gomez S, Deng J, Mak JY, Hachani A, Anderson RJ, Painter GF, Goyette J, Amarasinghe SL, Ritchie ME, Roquilly A, Fairlie DP, Gaus K, Rossjohn J, Villadangos JA, McWilliam HE. A specialized tyrosine-based endocytosis signal in MR1 controls antigen presentation to MAIT cells. J Cell Biol 2022; 221:213489. [PMID: 36129434 PMCID: PMC9499830 DOI: 10.1083/jcb.202110125] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 06/23/2022] [Accepted: 09/01/2022] [Indexed: 12/13/2022] Open
Abstract
MR1 is a highly conserved microbial immune-detection system in mammals. It captures vitamin B-related metabolite antigens from diverse microbes and presents them at the cell surface to stimulate MR1-restricted lymphocytes including mucosal-associated invariant T (MAIT) cells. MR1 presentation and MAIT cell recognition mediate homeostasis through host defense and tissue repair. The cellular mechanisms regulating MR1 cell surface expression are critical to its function and MAIT cell recognition, yet they are poorly defined. Here, we report that human MR1 is equipped with a tyrosine-based motif in its cytoplasmic domain that mediates low affinity binding with the endocytic adaptor protein 2 (AP2) complex. This interaction controls the kinetics of MR1 internalization from the cell surface and minimizes recycling. We propose MR1 uses AP2 endocytosis to define the duration of antigen presentation to MAIT cells and the detection of a microbial metabolic signature by the immune system.
Collapse
Affiliation(s)
- Hui Jing Lim
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute of Infection and Immunity, Melbourne, Victoria, Australia
| | - Jacinta M. Wubben
- Infection and Immunity Program and The Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute Monash University, Clayton, Victoria, Australia
| | - Cristian Pinero Garcia
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, The University of New South Wales, Sydney, Australia
| | - Sebastian Cruz-Gomez
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute of Infection and Immunity, Melbourne, Victoria, Australia
| | - Jieru Deng
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute of Infection and Immunity, Melbourne, Victoria, Australia
| | - Jeffrey Y.W. Mak
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Abderrahman Hachani
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute of Infection and Immunity, Melbourne, Victoria, Australia
| | - Regan J. Anderson
- Ferrier Research Institute, Victoria University of Wellington, Wellington, New Zealand
| | - Gavin F. Painter
- Ferrier Research Institute, Victoria University of Wellington, Wellington, New Zealand
| | - Jesse Goyette
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, The University of New South Wales, Sydney, Australia
| | - Shanika L. Amarasinghe
- Epigenetics and Development Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Matthew E. Ritchie
- Epigenetics and Development Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Antoine Roquilly
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute of Infection and Immunity, Melbourne, Victoria, Australia
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064; F-44000, Nantes, France
| | - David P. Fairlie
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Katharina Gaus
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, The University of New South Wales, Sydney, Australia
| | - Jamie Rossjohn
- Infection and Immunity Program and The Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute Monash University, Clayton, Victoria, Australia
- Institute of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff, UK
| | - Jose A. Villadangos
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute of Infection and Immunity, Melbourne, Victoria, Australia
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
- Jose A. Villadangos:
| | - Hamish E.G. McWilliam
- Department of Microbiology and Immunology, The University of Melbourne, The Peter Doherty Institute of Infection and Immunity, Melbourne, Victoria, Australia
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
- Correspondence to Hamish E.G. McWilliam:
| |
Collapse
|
46
|
Effect of diet on gut microbiota diversity in mandarin ducks (Aix galericulata) revealed by Illumina high-throughput sequencing. Arch Microbiol 2022; 204:725. [DOI: 10.1007/s00203-022-03333-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 11/03/2022] [Accepted: 11/10/2022] [Indexed: 11/27/2022]
|
47
|
Roles of Two-Component Signal Transduction Systems in Shigella Virulence. Biomolecules 2022; 12:biom12091321. [PMID: 36139160 PMCID: PMC9496106 DOI: 10.3390/biom12091321] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 11/17/2022] Open
Abstract
Two-component signal transduction systems (TCSs) are widespread types of protein machinery, typically consisting of a histidine kinase membrane sensor and a cytoplasmic transcriptional regulator that can sense and respond to environmental signals. TCSs are responsible for modulating genes involved in a multitude of bacterial functions, including cell division, motility, differentiation, biofilm formation, antibiotic resistance, and virulence. Pathogenic bacteria exploit the capabilities of TCSs to reprogram gene expression according to the different niches they encounter during host infection. This review focuses on the role of TCSs in regulating the virulence phenotype of Shigella, an intracellular pathogen responsible for severe human enteric syndrome. The pathogenicity of Shigella is the result of the complex action of a wide number of virulence determinants located on the chromosome and on a large virulence plasmid. In particular, we will discuss how five TCSs, EnvZ/OmpR, CpxA/CpxR, ArcB/ArcA, PhoQ/PhoP, and EvgS/EvgA, contribute to linking environmental stimuli to the expression of genes related to virulence and fitness within the host. Considering the relevance of TCSs in the expression of virulence in pathogenic bacteria, the identification of drugs that inhibit TCS function may represent a promising approach to combat bacterial infections.
Collapse
|
48
|
Li S, Han X, Upadhyay I, Zhang W. Characterization of Functional B-Cell Epitopes at the Amino Terminus of Shigella Invasion Plasmid Antigen B (IpaB). Appl Environ Microbiol 2022; 88:e0038422. [PMID: 35856689 PMCID: PMC9361828 DOI: 10.1128/aem.00384-22] [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: 03/01/2022] [Accepted: 06/29/2022] [Indexed: 01/22/2023] Open
Abstract
Shigella invasion plasmid antigen B (IpaB) plays an important role in causing shigellosis. While IpaB's protein structure, contribution to disease mechanism, and protective immunity against Shigella infection have been well studied, the significance of individual antigenic domains, especially at the N terminus, has not been systematically characterized. In an attempt to identify IpaB protein functional epitopes and to construct an optimized polyvalent multiepitope fusion antigen (MEFA) immunogen for development of a protein-based cross protective Shigella vaccine, in this study, we in silico identified immunodominant B-cell epitopes from the IpaB N terminus, fused each epitope to carrier protein CsaB (the major subunit of enterotoxigenic Escherichia coli CS4 adhesin) for epitope fusion proteins, immunized mice with each epitope fusion protein, examined IpaB-specific antibody responses, and assessed antibody functional activity against Shigella bacterial invasion. A total of 10 B-cell continuous epitopes were identified from IpaB N terminus, and after being fused to carrier protein CsaB, each epitope induced anti-IpaB IgG responses in the intramuscularly immunized mice. While in vitro antibody invasion inhibition assays demonstrated that antibodies derived from each identified epitope were functional, epitopes 1 (LAKILASTELGDNTIQAA), 2 (HSTSNILIPELKAPKSL), and 4 (QARQQKNLEFSDKI) induced antibodies to inhibit Shigella sonnei and Shigella flexneri invasion at levels similar to those of recombinant IpaB protein, suggesting that these three IpaB epitopes can be used potentially as IpaB-representing antigens to induce protective anti-IpaB antibodies and for construction of an epitope-based polyvalent MEFA protein immunogen for Shigella vaccine development. IMPORTANCE Currently, there are no effective measures for control or prevention of Shigella infection, the most common cause of diarrhea in children 3 to 5 years of age in developing countries. Challenges in developing Shigella vaccines include virulence heterogeneity among species and serotypes. To overcome virulence heterogeneity challenge and to develop a protein-based multivalent Shigella vaccine, we targeted a panel of virulence factors, including invasion plasmid antigens, identified functional antigenic domains or epitopes as representative antigens, and applied the novel epitope- and structure-based vaccinology platform multiepitope fusion antigen (MEFA) to integrate functional antigenic domains or epitopes into a backbone immunogen to produce a polyvalent immunogen for cross protective antibodies. Identification of functional IpaB epitopes from this study enhances our understanding of IpaB immunogenicity and allows us to directly utilize IpaB epitopes for construction of a cross protective polyvalent Shigella immunogen and to accelerate development of a protein-based Shigella vaccine.
Collapse
Affiliation(s)
- Siqi Li
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Xinfeng Han
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Sichuan Agricultural University College of Veterinary Medicine, Chengdu, Sichuan, People’s Republic of China
| | - Ipshita Upadhyay
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Weiping Zhang
- Department of Pathobiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
49
|
Bullones-Bolaños A, Bernal-Bayard J, Ramos-Morales F. The NEL Family of Bacterial E3 Ubiquitin Ligases. Int J Mol Sci 2022; 23:7725. [PMID: 35887072 PMCID: PMC9320238 DOI: 10.3390/ijms23147725] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/08/2022] [Accepted: 07/11/2022] [Indexed: 12/16/2022] Open
Abstract
Some pathogenic or symbiotic Gram-negative bacteria can manipulate the ubiquitination system of the eukaryotic host cell using a variety of strategies. Members of the genera Salmonella, Shigella, Sinorhizobium, and Ralstonia, among others, express E3 ubiquitin ligases that belong to the NEL family. These bacteria use type III secretion systems to translocate these proteins into host cells, where they will find their targets. In this review, we first introduce type III secretion systems and the ubiquitination process and consider the various ways bacteria use to alter the ubiquitin ligation machinery. We then focus on the members of the NEL family, their expression, translocation, and subcellular localization in the host cell, and we review what is known about the structure of these proteins, their function in virulence or symbiosis, and their specific targets.
Collapse
Affiliation(s)
| | | | - Francisco Ramos-Morales
- Departamento de Genética, Facultad de Biología, Universidad de Sevilla, 41012 Sevilla, Spain; (A.B.-B.); (J.B.-B.)
| |
Collapse
|
50
|
Abstract
Pyroptosis, a regulated form of pro-inflammatory cell death, is characterised by cell lysis and by the release of cytokines, damage- and pathogen-associated molecular patterns. It plays an important role during bacterial infection, where it can promote an inflammatory response and eliminate the replicative niche of intracellular pathogens. Recent work, using a variety of bacterial pathogens, has illuminated the versatility of pyroptosis, revealing unexpected and important concepts underlying host defence. In this Review, we overview the molecular mechanisms underlying pyroptosis and discuss their role in host defence, from the single cell to the whole organism. We focus on recent studies using three cellular microbiology paradigms - Mycobacterium tuberculosis, Salmonella Typhimurium and Shigella flexneri - that have transformed the field of pyroptosis. We compare insights discovered in tissue culture, zebrafish and mouse models, highlighting the advantages and disadvantages of using these complementary infection models to investigate pyroptosis and for modelling human infection. Moving forward, we propose that in-depth knowledge of pyroptosis obtained from complementary infection models can better inform future studies using higher vertebrates, including humans, and help develop innovative host-directed therapies to combat bacterial infection.
Collapse
Affiliation(s)
- Dominik Brokatzky
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | - Serge Mostowy
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| |
Collapse
|