1
|
Zhu L, Liang Y, Yang L, Yang Q, Yin J, Wang T, Xu X, Zhang Q. Helicobacter mastomyrinus infection induces autoimmune hepatitis in mice. J Transl Autoimmun 2025; 10:100275. [PMID: 39981114 PMCID: PMC11840492 DOI: 10.1016/j.jtauto.2025.100275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Accepted: 01/22/2025] [Indexed: 02/22/2025] Open
Abstract
Background Autoimmune hepatitis (AIH) is a chronic progressive liver disease caused by the immune system mistakenly attacking its own hepatocytes. The role of the gut microbiome in the pathogenesis and progression of AIH is of considerable significance. However, the dearth of suitable animal models has significantly constrained advancements in the pathogenesis and the development of therapeutic strategies for AIH. Helicobacter mastomyrinus (H. mastomyrinus, Hm) is a potentially zoonotic pathogenic microorganism capable of causing diseases of the enterohepatic system in rodent laboratory animals. Nevertheless, research on its role and mechanisms in causing liver disease is severely limited. Methods In this study, male BALB/c mice were infected with Hm isolate Hm-17, and were sacrificed at 4 w, 8 w, 14 w and 22 w after infection, respectively. The serum was collected for detecting a number of AIH indicators, including the aminotransferases level, IgG content and autoantibody level. Additionally, the liver tissue was examined for pathological analysis, fibrosis, bacterial content, and the distribution of immune cells. Results It was observed that the infection initially caused focal necrotizing hepatitis and subsequently progressed to interface hepatitis with lymphocyte/plasma cell infiltration, as well as hypergammaglobulinemia and autoantibody reactions, predominantly to Anti-nuclear and anti-smooth muscle antibodies. Furthermore, as the infection persisted, the mice exhibited a progressive increase in liver fibrosis and mild steatosis. Despite the maintenance of a low level of Hm colonization in the liver, there was a notable infiltrate of macrophages, T and B lymphocytes. In particular, the inflammatory foci in the Hm-infected liver were highly enriched for IL17+ cells. Conclusion The present study provides an animal model of immunological liver injury induced by Hm infection that exhibits main characteristics similar to those observed in AIH-1 patients. This model may serve as a novel animal model for the study of the pathogenesis and potential therapeutic strategies for human AIH.
Collapse
Affiliation(s)
- Liqi Zhu
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, China
| | - Yuanyuan Liang
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, China
| | - Linghan Yang
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, China
| | - Qihui Yang
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, China
| | - Jun Yin
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, China
| | - Tao Wang
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, China
| | - Xiangming Xu
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
- Taizhou University, Taizhou, China
| | - Quan Zhang
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, China
| |
Collapse
|
2
|
Zhang R, Zhang X, Lau HCH, Yu J. Gut microbiota in cancer initiation, development and therapy. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1283-1308. [PMID: 39821827 DOI: 10.1007/s11427-024-2831-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 12/12/2024] [Indexed: 01/19/2025]
Abstract
Cancer has long been associated with genetic and environmental factors, but recent studies reveal the important role of gut microbiota in its initiation and progression. Around 13% of cancers are linked to infectious agents, highlighting the need to identify the specific microorganisms involved. Gut microbiota can either promote or inhibit cancer growth by influencing oncogenic signaling pathways and altering immune responses. Dysbiosis can lead to cancer, while certain probiotics and their metabolites may help reestablish micro-ecological balance and improve anti-tumor immune responses. Research into targeted approaches that enhance therapy with probiotics is promising. However, the effects of probiotics in humans are complex and not yet fully understood. Additionally, methods to counteract harmful bacteria are still in development. Early clinical trials also indicate that modifying gut microbiota may help manage side effects of cancer treatments. Ongoing research is crucial to understand better how gut microbiota can be used to improve cancer prevention and treatment outcomes.
Collapse
Affiliation(s)
- Ruyi Zhang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiang Zhang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Harry Cheuk Hay Lau
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China
| | - Jun Yu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
3
|
Wang T, Meng X, Qian M, Jin S, Bao R, Zhu L, Zhang Q. Helicobacter hepaticus CdtB Triggers Colonic Mucosal Barrier Disruption in Mice via Epithelial Tight Junction Impairment Mediated by MLCK/pMLC2 Signaling Pathway. Vet Sci 2025; 12:174. [PMID: 40005934 PMCID: PMC11860670 DOI: 10.3390/vetsci12020174] [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: 01/10/2025] [Revised: 02/06/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Background:Helicobacter hepaticus (H. hepaticus) has been demonstrated to have clinical relevance to the development of colitis in rodents. H. hepaticus produces cytolethal distending toxins (CDTs), which are identified as the most important virulence factors to the pathogenicity of CDT-producing bacteria in animals. However, the precise relationship between CDTs of H. hepaticus and intestinal barrier dysfunction remains unclear. The objective of the present study was to ascertain the impact of CdtB, the active subunit of CDTs, on the colonic mucosal barrier during H. hepaticus infection. Materials and Methods: We investigated the infection of male BALB/c mice, intestinal organoids, and IEC-6 cell monolayers by H. hepaticus or CdtB-deficient H. hepaticus (ΔCdtB). A comprehensive histopathological examination was conducted, encompassing the assessment of H. hepaticus colonization, the levels of mRNA expression for inflammatory cytokines, the expression levels of tight junction proteins, and the related signaling pathways. Results: The results demonstrate that the presence of ΔCdtB led to a mitigation of the symptoms associated with H. hepaticus-induced colitis, as evidenced by colon length shortening and the colon histological inflammation score. In addition, the levels of pro-inflammatory cytokines were reduced in the ΔCdtB group. Moreover, a downward trend was observed in the phosphorylation levels of STAT3 and nuclear factor-κB (p65). In vitro, the presence of H. hepaticus resulted in a reduction in the expression of tight junction (TJ) markers (ZO-1 and occludin) and an impairment of the F-actin structure in either the intestinal epithelium or intestinal organoids. However, these effects were reversed by CdtB deletion. Concurrently, both ROS levels and apoptosis levels were found to be significantly reduced in cells treated with the ΔCdtB strain. Mechanistically, myosin light chain kinase (MLCK) activation was observed in the H. hepaticus-infected group in vivo, whereas the MLCK inhibitor ML-7 was found to reverse the CdtB-induced alterations in TJ proteins in IEC6 cells. Conclusions: The collective findings demonstrate that CdtB plays a pivotal role in the H. hepaticus-induced colonic mucosal barrier. This is achieved through the regulation of TJs via the MLCK/pMLC2 signaling pathway, which is linked to elevations in oxidative stress and inflammation within intestinal epithelial cells.
Collapse
Affiliation(s)
- Tao Wang
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Xiao Meng
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Miao Qian
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Shanhao Jin
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Ruoyu Bao
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Liqi Zhu
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Quan Zhang
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou 225009, China
- Jiangsu Transgenic Animal Pharmaceutical Engineering Research Center, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
4
|
Liqi Z, Yuanyuan L, Linghan Y, Jun Y, Tao W, Quan Z. Genome and pathogenicity analysis of Helicobacter mastomyrinus isolated from mice. Arch Microbiol 2025; 207:55. [PMID: 39939499 DOI: 10.1007/s00203-025-04254-x] [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: 08/09/2024] [Revised: 12/06/2024] [Accepted: 01/21/2025] [Indexed: 02/14/2025]
Abstract
The increasing attention given to the potential risk offered by enterohepatic Helicobacter species to the well-being of human beings and animals is of significant importance. Helicobacter mastomyrinus (H. mastomyrinus), a bacterium predominantly associated with rodents, has been implicated in liver and intestinal pathologies. Here, a strain of H. mastomyrinus, designated as Hm-17 (GenBank: CP145316.1), was isolated from asymptomatic C57BL/6 mice. Subsequently, an in-depth and comprehensive investigation was undertaken, which included genome sequencing analysis, micro-biochemical identification, evaluation of growth characteristic, cytotoxicity assessment, and testing of animal pathogenicity. The analysis of 16 S rRNA reveals a close phylogenetic relationship between H. mastomyrinus and H. canadensis. However, core-pan genome analysis and an evaluation of pathogenic factors indicates a more robust association between H. mastomyrinus and H. hepaticus. Cytotoxicity analysis revealed that Hm-17 exhibits robust cytolethal distending toxin (CDT) activity, inducing pronounced cellular swelling and death. Furthermore, Hm-17 infection in BALB/c mice results in rapid and characteristic focal necrotic hepatitis. Genome sequencing and pathogenicity analysis indicate that H. mastomyrinus isolates from asymptomatic mice possess significant pathogenic potential. These findings underscore the need for further investigation into the epidemiology and mechanisms of pathogenesis associated with this organism.
Collapse
Affiliation(s)
- Zhu Liqi
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
- International Research Laboratory of Prevention and Control of Important Animal Infectious Diseases and Zoonotic Diseases of Jiangsu Higher Education Institutions, Yangzhou University, Yangzhou, China
| | - Liang Yuanyuan
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Yang Linghan
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Yin Jun
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Wang Tao
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Zhang Quan
- College of Veterinary Medicine, Institute of Comparative Medicine, Yangzhou University, Yangzhou, China.
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China.
| |
Collapse
|
5
|
Permain J, Hock B, Eglinton T, Purcell R. Functional links between the microbiome and the molecular pathways of colorectal carcinogenesis. Cancer Metastasis Rev 2024; 43:1463-1474. [PMID: 39340753 PMCID: PMC11554747 DOI: 10.1007/s10555-024-10215-5] [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: 04/02/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024]
Abstract
Colorectal cancer (CRC) is a common cancer, with a concerning rise in early-onset CRC cases, signalling a shift in disease epidemiology. Whilst our understanding of the molecular underpinnings of CRC has expanded, the complexities underlying its initiation remain elusive, with emerging evidence implicating the microbiome in CRC pathogenesis. This review synthesizes current knowledge on the intricate interplay between the microbiome, tumour microenvironment (TME), and molecular pathways driving CRC carcinogenesis. Recent studies have reported how the microbiome may modulate the TME and tumour immune responses, consequently influencing cancer progression, and whilst specific bacteria have been linked with CRC, the underlying mechanisms remains poorly understood. By elucidating the functional links between microbial landscapes and carcinogenesis pathways, this review offers insights into how bacteria orchestrate diverse pathways of CRC development, shedding light on potential therapeutic targets and personalized intervention strategies.
Collapse
Affiliation(s)
- Jessica Permain
- Department of Surgery and Critical Care, University of Otago, Christchurch, New Zealand
| | - Barry Hock
- Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Timothy Eglinton
- Department of Surgery and Critical Care, University of Otago, Christchurch, New Zealand
| | - Rachel Purcell
- Department of Surgery and Critical Care, University of Otago, Christchurch, New Zealand.
| |
Collapse
|
6
|
Azzi-Martin L, Touffait-Calvez V, Everaert M, Jia R, Sifré E, Seeneevassen L, Varon C, Dubus P, Ménard A. Cytolethal Distending Toxin Modulates Cell Differentiation and Elicits Epithelial to Mesenchymal Transition. J Infect Dis 2024; 229:1688-1701. [PMID: 38416880 DOI: 10.1093/infdis/jiae105] [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: 08/07/2023] [Revised: 01/29/2024] [Accepted: 02/27/2024] [Indexed: 03/01/2024] Open
Abstract
BACKGROUND The bacterial genotoxin, cytolethal distending toxin (CDT), causes DNA damage in host cells, a risk factor for carcinogenesis. Previous studies have shown that CDT induces phenotypes reminiscent of epithelial to mesenchymal transition (EMT), a process involved in cancer initiation and progression. METHODS We investigated different steps of EMT in response to Helicobacter hepaticus CDT and its active CdtB subunit using in vivo and in vitro models. RESULTS Most of the steps of the EMT process were induced by CDT/CdtB and observed throughout the study in murine and epithelial cell culture models. CdtB induced cell-cell junction disassembly, causing individualization of cells and acquisition of a spindle-like morphology. The key transcriptional regulators of EMT (SNAIL and ZEB1) and some EMT markers were upregulated at both RNA and protein levels in response to CDT/CdtB. CdtB increased the expression and proteolytic activity of matrix metalloproteinases, as well as cell migration. A range of these results were confirmed in Helicobacter hepaticus-infected and xenograft murine models. In addition, colibactin, a genotoxic metabolite produced by Escherichia coli, induced EMT-like effects in cell culture. CONCLUSIONS Overall, these data show that infection with genotoxin-producing bacteria elicits EMT process activation, supporting their role in tumorigenesis.
Collapse
Affiliation(s)
- Lamia Azzi-Martin
- Bordeaux Institute of Oncology, UMR1312, INSERM, University of Bordeaux, Bordeaux, France
- Unité de Formation et de Recherche des Sciences Médicales, University of Bordeaux, Bordeaux, France
| | | | - Maude Everaert
- Bordeaux Institute of Oncology, UMR1312, INSERM, University of Bordeaux, Bordeaux, France
| | - Ruxue Jia
- Bordeaux Institute of Oncology, UMR1312, INSERM, University of Bordeaux, Bordeaux, France
| | - Elodie Sifré
- Bordeaux Institute of Oncology, UMR1312, INSERM, University of Bordeaux, Bordeaux, France
| | - Lornella Seeneevassen
- Bordeaux Institute of Oncology, UMR1312, INSERM, University of Bordeaux, Bordeaux, France
| | - Christine Varon
- Bordeaux Institute of Oncology, UMR1312, INSERM, University of Bordeaux, Bordeaux, France
- Unité de Formation et de Recherche des Sciences Médicales, University of Bordeaux, Bordeaux, France
| | - Pierre Dubus
- Bordeaux Institute of Oncology, UMR1312, INSERM, University of Bordeaux, Bordeaux, France
- Unité de Formation et de Recherche des Sciences Médicales, University of Bordeaux, Bordeaux, France
- Institut de Pathologie et de Biologie du Cancer, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Armelle Ménard
- Bordeaux Institute of Oncology, UMR1312, INSERM, University of Bordeaux, Bordeaux, France
| |
Collapse
|
7
|
Muthupalani S, Annamalai D, Feng Y, Ganesan SM, Ge Z, Whary MT, Nakagawa H, Rustgi AK, Wang TC, Fox JG. IL-1β transgenic mouse model of inflammation driven esophageal and oral squamous cell carcinoma. Sci Rep 2023; 13:12732. [PMID: 37543673 PMCID: PMC10404242 DOI: 10.1038/s41598-023-39907-8] [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: 03/11/2023] [Accepted: 08/02/2023] [Indexed: 08/07/2023] Open
Abstract
Chronic inflammation is integral to the development of esophageal adenocarcinoma (EAC) and esophageal squamous cell carcinoma (ESCC), although the latter has not been associated with reflux esophagitis. The L2-IL-1β transgenic mice, expressing human interleukin (IL)-1β in the oral, esophageal and forestomach squamous epithelia feature chronic inflammation and a stepwise development of Barrett's esophagus-like metaplasia, dysplasia and adenocarcinoma at the squamo-columnar junction. However, the functional consequences of IL-1β-mediated chronic inflammation in the oral and esophageal squamous epithelia remain elusive. We report for the first time that in addition to the previously described Barrett's esophagus-like metaplasia, the L2-IL-1β mice also develop squamous epithelial dysplasia with progression to squamous cell carcinoma (SCC) in the esophagus and the tongue. L2-IL-1β showed age-dependent progression of squamous dysplasia to SCC with approximately 40% (n = 49) and 23.5% (n = 17) incidence rates for esophageal and tongue invasive SCC respectively, by 12-15 months of age. Interestingly, SCC development and progression in L2-IL-1β was similar in both Germ Free (GF) and Specific Pathogen Free (SPF) conditions. Immunohistochemistry revealed a T cell predominant inflammatory profile with enhanced expression of Ki67, Sox2 and the DNA double-strand break marker, γ-H2AX, in the dysplastic squamous epithelia of L2-IL-1β mice. Pro-inflammatory cytokines, immunomodulatory players, chemoattractants for inflammatory cells (T cells, neutrophils, eosinophils, and macrophages) and oxidative damage marker, iNOS, were significantly increased in the esophageal and tongue tissues of L2-IL-1β mice. Our recent findings have expanded the translational utility of the IL-1β mouse model to aid in further characterization of the key pathways of inflammation driven BE and EAC as well as ESCC and Oral SCC.
Collapse
Affiliation(s)
- Sureshkumar Muthupalani
- Division of Comparative Medicine, Massachusetts Institute of Technology, 77 Massachusetts Avenue, 16-825C, Cambridge, MA, 02139, USA.
- StageBio, 5930 Main St, Mount Jackson, VA, 22842, USA.
| | - Damodaran Annamalai
- Division of Comparative Medicine, Massachusetts Institute of Technology, 77 Massachusetts Avenue, 16-825C, Cambridge, MA, 02139, USA
| | - Yan Feng
- Division of Comparative Medicine, Massachusetts Institute of Technology, 77 Massachusetts Avenue, 16-825C, Cambridge, MA, 02139, USA
| | - Suresh M Ganesan
- Division of Comparative Medicine, Massachusetts Institute of Technology, 77 Massachusetts Avenue, 16-825C, Cambridge, MA, 02139, USA
| | - Zhongming Ge
- Division of Comparative Medicine, Massachusetts Institute of Technology, 77 Massachusetts Avenue, 16-825C, Cambridge, MA, 02139, USA
| | - Mark T Whary
- Division of Comparative Medicine, Massachusetts Institute of Technology, 77 Massachusetts Avenue, 16-825C, Cambridge, MA, 02139, USA
| | - Hiroshi Nakagawa
- Division of Digestive and Liver Diseases and Herbert Irving Cancer Research Center, Columbia University College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Anil K Rustgi
- Division of Digestive and Liver Diseases and Herbert Irving Cancer Research Center, Columbia University College of Physicians and Surgeons, New York, NY, 10032, USA
| | - Timothy C Wang
- Division of Digestive and Liver Diseases and Herbert Irving Cancer Research Center, Columbia University College of Physicians and Surgeons, New York, NY, 10032, USA
| | - James G Fox
- Division of Comparative Medicine, Massachusetts Institute of Technology, 77 Massachusetts Avenue, 16-825C, Cambridge, MA, 02139, USA.
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
| |
Collapse
|
8
|
Kato I, Minkevitch J, Sun J. Oncogenic potential of Campylobacter infection in the gastrointestinal tract: narrative review. Scand J Gastroenterol 2023; 58:1453-1465. [PMID: 37366241 DOI: 10.1080/00365521.2023.2228954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/26/2023] [Accepted: 06/16/2023] [Indexed: 06/28/2023]
Abstract
BACKGROUND Campylobacter jejuni is the leading cause of zoonotic gastroenteritis. The other emerging group of Campylobacters spp. are part of human oral commensal, represented by C. concisus (CC), which has been recently linked to non-oral conditions. Although long-term gastrointestinal (GI) complications from these two groups of Campylobacters have been previously reviewed individually, overall impact of Campylobacter infection on GI carcinogenesis and their inflammatory precursor lesions has not been assessed collectively. AIMS To evaluate the available evidence concerning the association between Campylobacter infection/colonization and inflammatory bowel disease (IBD), reflux esophagitis/metaplasia colorectal cancer (CRC) and esophageal cancer (EC). METHODS We performed a comprehensive literature search of PubMed for relevant original publications and systematic reviews/meta-analyses of epidemiological and clinical studies. In addition, we gathered additional information concerning microbiological data, animal models and mechanistic data from in vitro studies. RESULTS Both retrospective and prospective studies on IBD showed relatively consistent increased risk associated with Campylobacter infection. Despite lack of supporting prospective studies, retrospective studies based on tissue/fecal microbiome revealed consistent enrichment of Campylobacter in CRC samples. Studies on EC precursor lesions (esophagitis and metaplasia) were generally supportive for the association with Campylobacter, while inconsistent observations on EC. Studies on both IBD and EC precursors suggested the predominant role of CC, but studies on CRC were not informative of species. CONCLUSIONS There is sufficient evidence calling for concerted effort in unveiling direct and indirect connection of this organism to colorectal and esophageal cancer in humans.
Collapse
Affiliation(s)
- Ikuko Kato
- Department of Oncology and Pathology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Julia Minkevitch
- Rosalind Franklin University of Medicine and Science, Chicago, IL, USA
| | - Jun Sun
- Department of Microbiology/Immunology, University of Illinois at Chicago (UIC), Chicago, IL, USA
- UIC Cancer Center, Chicago, IL, USA
| |
Collapse
|
9
|
Genotoxins: The Mechanistic Links between Escherichia coli and Colorectal Cancer. Cancers (Basel) 2023; 15:cancers15041152. [PMID: 36831495 PMCID: PMC9954437 DOI: 10.3390/cancers15041152] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 02/06/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023] Open
Abstract
Emerging evidence indicates bacterial infections contribute to the formation of cancers. Bacterial genotoxins are effectors that cause DNA damage by introducing single- and double-strand DNA breaks in the host cells. The first bacterial genotoxin cytolethal distending toxin (CDT) was a protein identified in 1987 in a pathogenic strain in Escherichia coli (E. coli) isolated from a young patient. The peptide-polyketide genotoxin colibactin is produced by the phylogenetic group B2 of E. coli. Recently, a protein produced by attaching/effacing (A/E) pathogens, including enteropathogenic and enterohemorrhagic E. coli (EPEC and EHEC) and their murine equivalent Citrobacter rodentium (CR), has been reported as a novel protein genotoxin, being injected via the type III secretion system (T3SS) into host cells and harboring direct DNA digestion activity with a catalytic histidine-aspartic acid dyad. These E. coli-produced genotoxins impair host DNA, which results in senescence or apoptosis of the target cells if the damage is beyond repair. Conversely, host cells can survive and proliferate if the genotoxin-induced DNA damage is not severe enough to kill them. The surviving cells may accumulate genomic instability and acquire malignant traits. This review presents the cellular responses of infection with the genotoxins-producing E. coli and discusses the current knowledge of the tumorigenic potential of these toxins.
Collapse
|
10
|
Hand TW, Overacre-Delgoffe AE. The complex immunological role of Helicobacter in modulating cancer. Trends Immunol 2022; 43:826-832. [PMID: 36041951 DOI: 10.1016/j.it.2022.08.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/02/2022] [Accepted: 08/04/2022] [Indexed: 10/15/2022]
Abstract
The gut microbiota has recently emerged as a unique mechanism of immunotherapeutic resistance or response within certain cancer patients. Certain adherent bacterial species that reside along the epithelial barrier within the gastrointestinal tract have been shown to be the most immunogenic and include several species within the Helicobacteraceae family. The role of these microbes in cancer remains controversial and varies according to species, immune status, and cancer type. Here, we hypothesize that the functional characteristics rather than the bacterial species of Helicobacteraceae dictate the type of immune response with either a benefit or a detriment to overall cancer progression.
Collapse
Affiliation(s)
- T W Hand
- University of Pittsburgh, Department of Immunology, Pittsburgh, PA, USA; Children's Hospital of Pittsburgh, RK Mellon Institute, Department of Pediatrics, Pittsburgh, PA, USA
| | - A E Overacre-Delgoffe
- University of Pittsburgh, Department of Immunology, Pittsburgh, PA, USA; Cancer Immunology and Immunotherapy Program, UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
| |
Collapse
|
11
|
Zhu L, Wu Z, Zhu C, Yin J, Huang Y, Feng J, Zhang Q. The Deletion of IL-17A Enhances Helicobacter hepaticus Colonization and Triggers Colitis. J Inflamm Res 2022; 15:2761-2773. [PMID: 35518840 PMCID: PMC9064063 DOI: 10.2147/jir.s359100] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 04/19/2022] [Indexed: 01/14/2023] Open
Abstract
Objective IL-17 is a key regulator of the inflammatory response, and as such, it is involved in the constraint and clearance of pathogens. The mechanism of IL-17 in the pathogenesis of inflammatory bowel disease (IBD) caused by microbial infection is still unclear. Helicobacter hepaticus infection can induce colitis in many mouse strains, and thus, it has been widely used in the study of IBD pathogenesis. Methods In this study, male C57BL/6, BALB/c, Il-10−/−, and Il-17a−/− mice were infected with H. hepaticus for several weeks. Histopathology, H. hepaticus colonization and distribution, expression of inflammatory cytokines and lysozyme, and distribution of mucus in proximal colon were examined. Results The colonic colonization of H. hepaticus was abnormally high in Il-17a−/− mice. H. hepaticus infection caused only mild to moderate colitis symptoms in Il-17a−/− mice, including low levels of lymphocyte infiltration, epithelial cell defects, goblet cell reduction, and crypt atrophy without obvious hyperplasia in the later stage of infection. Furthermore, many inflammatory genes were significantly increased in the proximal colon of H. hepaticus-infected Il-17a−/− mice compared with C57BL/6 mice. In addition, the reduction of colonic mucus and the down-regulation of ZO-1, Claudin-1, and IL-22 were observed in Il-17a−/− mice compared with C57BL/6 mice post H. hepaticus infection. Conclusion These results demonstrated that the deletion of IL-17A impaired the integrity of the intestinal epithelium, weakened the secretion of mucus, attenuated colonic mucosal regeneration, reduced the ability to resist microbial infection, and finally led to colitis caused by H. hepaticus.
Collapse
Affiliation(s)
- Liqi Zhu
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, People's Republic of China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, 225009, People's Republic of China
| | - Zhihao Wu
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, People's Republic of China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, 225009, People's Republic of China
| | - Chen Zhu
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, People's Republic of China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, 225009, People's Republic of China
| | - Jun Yin
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, People's Republic of China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, 225009, People's Republic of China
| | - Yuzheng Huang
- National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, Jiangsu Institute of Parasitic Diseases, Wuxi, Jiangsu Province, 214064, People's Republic of China.,Public Health Research Center, Jiangnan University, Wuxi, Jiangsu Province, 214122, People's Republic of China
| | - Jie Feng
- Shanghai Laboratory Animal Research Center, Shanghai Quality Monitoring Center of Laboratory Animals, Shanghai, 201203, People's Republic of China
| | - Quan Zhang
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, 225009, People's Republic of China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, 225009, People's Republic of China
| |
Collapse
|
12
|
Contino KF, Yadav H, Shiozawa Y. The gut microbiota can be a potential regulator and treatment target of bone metastasis. Biochem Pharmacol 2022; 197:114916. [PMID: 35041811 PMCID: PMC8858876 DOI: 10.1016/j.bcp.2022.114916] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/10/2022] [Accepted: 01/10/2022] [Indexed: 02/08/2023]
Abstract
The gut microbiota, an often forgotten organ, have a tremendous impact on human health. It has long been known that the gut microbiota are implicated in cancer development, and more recently, the gut microbiota have been shown to influence cancer metastasis to distant organs. Although one of the most common sites of distant metastasis is the bone, and the skeletal system has been shown to be a subject of interactions with the gut microbiota to regulate bone homeostasis, little research has been done regarding how the gut microbiota control the development of bone metastasis. This review will discuss the mechanisms through which the gut microbiota and derived microbial compounds (i) regulate gastrointestinal cancer disease progression and metastasis, (ii) influence skeletal remodeling and potentially modulate bone metastasis, and (iii) affect and potentially enhance immunotherapeutic treatments for bone metastasis.
Collapse
Affiliation(s)
- Kelly F Contino
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA
| | - Hariom Yadav
- Department of Neurosurgery and Brain Repair and Institute for Microbiome, University of South Florida, Tampa, FL 33612, USA
| | - Yusuke Shiozawa
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University Health Sciences, Winston-Salem, NC 27157, USA.
| |
Collapse
|
13
|
Overacre-Delgoffe AE, Bumgarner HJ, Cillo AR, Burr AHP, Tometich JT, Bhattacharjee A, Bruno TC, Vignali DAA, Hand TW. Microbiota-specific T follicular helper cells drive tertiary lymphoid structures and anti-tumor immunity against colorectal cancer. Immunity 2021; 54:2812-2824.e4. [PMID: 34861182 PMCID: PMC8865366 DOI: 10.1016/j.immuni.2021.11.003] [Citation(s) in RCA: 160] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 07/19/2021] [Accepted: 11/04/2021] [Indexed: 02/06/2023]
Abstract
The composition of the intestinal microbiota is associated with both the development of tumors and the efficacy of anti-tumor immunity. Here, we examined the impact of microbiota-specific T cells in anti-colorectal cancer (CRC) immunity. Introduction of Helicobacter hepaticus (Hhep) in a mouse model of CRC did not alter the microbial landscape but increased tumor infiltration by cytotoxic lymphocytes and inhibited tumor growth. Anti-tumor immunity was independent of CD8+ T cells but dependent upon CD4+ T cells, B cells, and natural killer (NK) cells. Hhep colonization induced Hhep-specific T follicular helper (Tfh) cells, increased the number of colon Tfh cells, and supported the maturation of Hhep+ tumor-adjacent tertiary lymphoid structures. Tfh cells were necessary for Hhep-mediated tumor control and immune infiltration, and adoptive transfer of Hhep-specific CD4+ T cells to Tfh cell-deficient Bcl6fl/flCd4Cre mice restored anti-tumor immunity. Thus, introduction of immunogenic intestinal bacteria can promote Tfh-associated anti-tumor immunity in the colon, suggesting therapeutic approaches for the treatment of CRC.
Collapse
Affiliation(s)
- Abigail E Overacre-Delgoffe
- R.K. Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA 15224, USA; Department of Immunology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15261, USA
| | - Hannah J Bumgarner
- R.K. Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA 15224, USA; Department of Immunology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15261, USA; Graduate Program of Microbiology and Immunology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| | - Anthony R Cillo
- Department of Immunology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15261, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Ansen H P Burr
- R.K. Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA 15224, USA; Department of Immunology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15261, USA; Graduate Program of Microbiology and Immunology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15213, USA
| | - Justin T Tometich
- R.K. Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA 15224, USA
| | - Amrita Bhattacharjee
- R.K. Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA 15224, USA; Department of Immunology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15261, USA
| | - Tullia C Bruno
- Department of Immunology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15261, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Dario A A Vignali
- Department of Immunology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15261, USA; Tumor Microenvironment Center, UPMC Hillman Cancer Center, Pittsburgh, PA 15232, USA
| | - Timothy W Hand
- R.K. Mellon Institute for Pediatric Research, Pediatrics Department, Infectious Disease Section, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA 15224, USA; Department of Immunology, University of Pittsburgh, School of Medicine, Pittsburgh, PA 15261, USA.
| |
Collapse
|
14
|
Lai YR, Chang YF, Ma J, Chiu CH, Kuo ML, Lai CH. From DNA Damage to Cancer Progression: Potential Effects of Cytolethal Distending Toxin. Front Immunol 2021; 12:760451. [PMID: 34868002 PMCID: PMC8634426 DOI: 10.3389/fimmu.2021.760451] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/21/2021] [Indexed: 12/16/2022] Open
Abstract
Cytolethal distending toxin (CDT), one of the most important genotoxins, is produced by several gram-negative bacteria and is involved in bacterial pathogenesis. Recent studies have shown that bacteria producing this peculiar genotoxin target host DNA, which potentially contributes to development of cancer. In this review, we highlighted the recent studies focusing on the idea that CDT leads to DNA damage, and the cells with inappropriately repaired DNA continue cycling, resulting in cancer development. Understanding the detailed mechanisms of genotoxins that cause DNA damage might be useful for targeting potential markers that drive cancer progression and help to discover new therapeutic strategies to prevent diseases caused by pathogens.
Collapse
Affiliation(s)
- Yi-Ru Lai
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Fang Chang
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Jason Ma
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Cheng-Hsun Chiu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Molecular Infectious Disease Research Center, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Ming-Ling Kuo
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Division of Allergy, Asthma, and Rheumatology, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Chih-Ho Lai
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Molecular Infectious Disease Research Center, Department of Pediatrics, Chang Gung Memorial Hospital, Linkou, Taiwan.,Department of Microbiology, School of Medicine, China Medical University, Taichung, Taiwan.,Department of Nursing, Asia University, Taichung, Taiwan
| |
Collapse
|
15
|
Jeffery R, Ilott NE, Powrie F. Genetic and environmental factors shape the host response to Helicobacter hepaticus: insights into IBD pathogenesis. Curr Opin Microbiol 2021; 65:145-155. [PMID: 34883389 DOI: 10.1016/j.mib.2021.10.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 10/07/2021] [Accepted: 10/12/2021] [Indexed: 11/03/2022]
Abstract
Pathobionts are members of the gut microbiota with the capacity to cause disease when there is malfunctioning intestinal homeostasis. These organisms are thought to be major contributors to the pathogenesis of inflammatory bowel disease (IBD), a group of chronic inflammatory disorders driven by dysregulated responses towards the microbiota. Over two decades have passed since the discovery of Helicobacter hepaticus, a mouse pathobiont which causes colitis in the context of immune deficiency. During this time, we have developed a detailed understanding of the cellular players and cytokine networks which drive H. hepaticus immunopathology. However, we are just beginning to understand the microbial factors that enable H. hepaticus to interact with the host and influence colonic health and disease. Here we review key H. hepaticus-host interactions, their relevance to other exemplar pathobionts and how when maladapted they drive colitis. Further understanding of these pathways may offer new therapeutic approaches for IBD.
Collapse
Affiliation(s)
- Rebecca Jeffery
- Kennedy Institute of Rheumatology, University of Oxford, Old Road Campus, Roosevelt Drive, Headington, Oxford OX3 7FY, United Kingdom
| | - Nicholas E Ilott
- Kennedy Institute of Rheumatology, University of Oxford, Old Road Campus, Roosevelt Drive, Headington, Oxford OX3 7FY, United Kingdom
| | - Fiona Powrie
- Kennedy Institute of Rheumatology, University of Oxford, Old Road Campus, Roosevelt Drive, Headington, Oxford OX3 7FY, United Kingdom.
| |
Collapse
|
16
|
Varon C, Azzi-Martin L, Khalid S, Seeneevassen L, Ménard A, Spuul P. Helicobacters and cancer, not only gastric cancer? Semin Cancer Biol 2021; 86:1138-1154. [PMID: 34425210 DOI: 10.1016/j.semcancer.2021.08.007] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 08/11/2021] [Accepted: 08/16/2021] [Indexed: 12/11/2022]
Abstract
The Helicobacter genus actually comprises 46 validly published species divided into two main clades: gastric and enterohepatic Helicobacters. These bacteria colonize alternative sites of the digestive system in animals and humans, and contribute to inflammation and cancers. In humans, Helicobacter infection is mainly related to H. pylori, a gastric pathogen infecting more than half of the world's population, leading to chronic inflammation of the gastric mucosa that can evolve into two types of gastric cancers: gastric adenocarcinomas and gastric MALT lymphoma. In addition, H. pylori but also non-H. pylori Helicobacter infection has been associated with many extra-gastric malignancies. This review focuses on H. pylori and its role in gastric cancers and extra-gastric diseases, as well as malignancies induced by non-H. pylori Helicobacters. Their different virulence factors and their involvement in carcinogenesis is discussed. This review highlights the importance of both gastric and enterohepatic Helicobacters in gastrointestinal and liver cancers.
Collapse
Affiliation(s)
- Christine Varon
- Univ. Bordeaux, INSERM, UMR1053 Bordeaux Research in Translational Oncology, BaRITOn, Bordeaux, France
| | - Lamia Azzi-Martin
- Univ. Bordeaux, INSERM, UMR1053 Bordeaux Research in Translational Oncology, BaRITOn, Bordeaux, France; Univ. Bordeaux, UFR des Sciences Médicales, Bordeaux, France
| | - Sadia Khalid
- Tallinn University of Technology, Department of Chemistry and Biotechnology, Akadeemia RD 15, 12618, Tallinn, Estonia
| | - Lornella Seeneevassen
- Univ. Bordeaux, INSERM, UMR1053 Bordeaux Research in Translational Oncology, BaRITOn, Bordeaux, France
| | - Armelle Ménard
- Univ. Bordeaux, INSERM, UMR1053 Bordeaux Research in Translational Oncology, BaRITOn, Bordeaux, France
| | - Pirjo Spuul
- Tallinn University of Technology, Department of Chemistry and Biotechnology, Akadeemia RD 15, 12618, Tallinn, Estonia.
| |
Collapse
|
17
|
The Role of DNA Damage Response in Dysbiosis-Induced Colorectal Cancer. Cells 2021; 10:cells10081934. [PMID: 34440703 PMCID: PMC8391204 DOI: 10.3390/cells10081934] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/23/2021] [Accepted: 07/23/2021] [Indexed: 12/16/2022] Open
Abstract
The high incidence of colorectal cancer (CRC) in developed countries indicates a predominant role of the environment as a causative factor. Natural gut microbiota provides multiple benefits to humans. Dysbiosis is characterized by an unbalanced microbiota and causes intestinal damage and inflammation. The latter is a common denominator in many cancers including CRC. Indeed, in an inflammation scenario, cellular growth is promoted and immune cells release Reactive Oxygen Species (ROS) and Reactive Nitrogen Species (RNS), which cause DNA damage. Apart from that, many metabolites from the diet are converted into DNA damaging agents by microbiota and some bacteria deliver DNA damaging toxins in dysbiosis conditions as well. The interactions between diet, microbiota, inflammation, and CRC are not the result of a straightforward relationship, but rather a network of multifactorial interactions that deserve deep consideration, as their consequences are not yet fully elucidated. In this paper, we will review the influence of dysbiosis in the induction of DNA damage and CRC.
Collapse
|
18
|
Ochoa S, Collado L. Enterohepatic Helicobacter species - clinical importance, host range, and zoonotic potential. Crit Rev Microbiol 2021; 47:728-761. [PMID: 34153195 DOI: 10.1080/1040841x.2021.1924117] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The genus Helicobacter defined just over 30 years ago, is a highly diverse and fast-growing group of bacteria that are able to persistently colonize a wide range of animals. The members of this genus are subdivided into two groups with different ecological niches, associated pathologies, and phylogenetic relationships: the gastric Helicobacter (GH) and the enterohepatic Helicobacter (EHH) species. Although GH have been mostly studied, EHH species have become increasingly important as emerging human pathogens and potential zoonotic agents in the last years. This group of bacteria has been associated with the development of several diseases in humans from acute pathologies like gastroenteritis to chronic pathologies that include inflammatory bowel disease, and liver and gallbladder diseases. However, their reservoirs, as well as their routes of transmission, have not been well established yet. Therefore, this review summarizes the current knowledge of taxonomy, epidemiology, and clinical role of the EHH group.
Collapse
Affiliation(s)
- Sofia Ochoa
- Faculty of Sciences, Institute of Biochemistry and Microbiology, Universidad Austral de Chile, Valdivia, Chile.,ANID - Millennium Science Initiative Program - Millennium Nucleus in the Biology of the Intestinal Microbiota, Santiago, Chile
| | - Luis Collado
- Faculty of Sciences, Institute of Biochemistry and Microbiology, Universidad Austral de Chile, Valdivia, Chile.,ANID - Millennium Science Initiative Program - Millennium Nucleus in the Biology of the Intestinal Microbiota, Santiago, Chile
| |
Collapse
|
19
|
Lopez Chiloeches M, Bergonzini A, Frisan T. Bacterial Toxins Are a Never-Ending Source of Surprises: From Natural Born Killers to Negotiators. Toxins (Basel) 2021; 13:426. [PMID: 34204481 PMCID: PMC8235270 DOI: 10.3390/toxins13060426] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/07/2021] [Accepted: 06/14/2021] [Indexed: 02/07/2023] Open
Abstract
The idea that bacterial toxins are not only killers but also execute more sophisticated roles during bacteria-host interactions by acting as negotiators has been highlighted in the past decades. Depending on the toxin, its cellular target and mode of action, the final regulatory outcome can be different. In this review, we have focused on two families of bacterial toxins: genotoxins and pore-forming toxins, which have different modes of action but share the ability to modulate the host's immune responses, independently of their capacity to directly kill immune cells. We have addressed their immuno-suppressive effects with the perspective that these may help bacteria to avoid clearance by the host's immune response and, concomitantly, limit detrimental immunopathology. These are optimal conditions for the establishment of a persistent infection, eventually promoting asymptomatic carriers. This immunomodulatory effect can be achieved with different strategies such as suppression of pro-inflammatory cytokines, re-polarization of the immune response from a pro-inflammatory to a tolerogenic state, and bacterial fitness modulation to favour tissue colonization while preventing bacteraemia. An imbalance in each of those effects can lead to disease due to either uncontrolled bacterial proliferation/invasion, immunopathology, or both.
Collapse
Affiliation(s)
| | | | - Teresa Frisan
- Department of Molecular Biology and Umeå Centre for Microbial Research (UCMR), Umeå University, 901 87 Umeå, Sweden; (M.L.C.); (A.B.)
| |
Collapse
|
20
|
Tremblay W, Mompart F, Lopez E, Quaranta M, Bergoglio V, Hashim S, Bonnet D, Alric L, Mas E, Trouche D, Vignard J, Ferrand A, Mirey G, Fernandez-Vidal A. Cytolethal Distending Toxin Promotes Replicative Stress Leading to Genetic Instability Transmitted to Daughter Cells. Front Cell Dev Biol 2021; 9:656795. [PMID: 34026755 PMCID: PMC8138442 DOI: 10.3389/fcell.2021.656795] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 03/23/2021] [Indexed: 12/30/2022] Open
Abstract
The cytolethal distending toxin (CDT) is produced by several Gram-negative pathogenic bacteria. In addition to inflammation, experimental evidences are in favor of a protumoral role of CDT-harboring bacteria such as Escherichia coli, Campylobacter jejuni, or Helicobacter hepaticus. CDT may contribute to cell transformation in vitro and carcinogenesis in mice models, through the genotoxic action of CdtB catalytic subunit. Here, we investigate the mechanism of action by which CDT leads to genetic instability in human cell lines and colorectal organoids from healthy patients’ biopsies. We demonstrate that CDT holotoxin induces a replicative stress dependent on CdtB. The slowing down of DNA replication occurs mainly in late S phase, resulting in the expression of fragile sites and important chromosomic aberrations. These DNA abnormalities induced after CDT treatment are responsible for anaphase bridge formation in mitosis and interphase DNA bridge between daughter cells in G1 phase. Moreover, CDT-genotoxic potential preferentially affects human cycling cells compared to quiescent cells. Finally, the toxin induces nuclear distension associated to DNA damage in proliferating cells of human colorectal organoids, resulting in decreased growth. Our findings thus identify CDT as a bacterial virulence factor targeting proliferating cells, such as human colorectal progenitors or stem cells, inducing replicative stress and genetic instability transmitted to daughter cells that may therefore contribute to carcinogenesis. As some CDT-carrying bacterial strains were detected in patients with colorectal cancer, targeting these bacteria could be a promising therapeutic strategy.
Collapse
Affiliation(s)
- William Tremblay
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Florence Mompart
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Elisa Lopez
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Muriel Quaranta
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, UPS, Toulouse, France
| | - Valérie Bergoglio
- MCD, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Saleha Hashim
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Delphine Bonnet
- Department of Internale and Digestive Diseases, Pole Digestif, CHU Toulouse, Paul Sabatier University, Toulouse, France
| | - Laurent Alric
- Department of Internale and Digestive Diseases, Pole Digestif, CHU Toulouse, Paul Sabatier University, Toulouse, France
| | - Emmanuel Mas
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, UPS, Toulouse, France.,Unité de Gastroentérologie, Hépatologie, Nutrition, Diabétologie et Maladies Héréditaires du Métabolisme, Hôpital des Enfants, CHU de Toulouse, Toulouse, France
| | - Didier Trouche
- MCD, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Julien Vignard
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Audrey Ferrand
- IRSD, Université de Toulouse, INSERM, INRAE, ENVT, UPS, Toulouse, France
| | - Gladys Mirey
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France
| | - Anne Fernandez-Vidal
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UPS, Toulouse, France.,MCD, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse, France
| |
Collapse
|
21
|
Huang G, Boesze-Battaglia K, Walker LP, Zekavat A, Schaefer ZP, Blanke SR, Shenker BJ. The Active Subunit of the Cytolethal Distending Toxin, CdtB, Derived From Both Haemophilus ducreyi and Campylobacter jejuni Exhibits Potent Phosphatidylinositol-3,4,5-Triphosphate Phosphatase Activity. Front Cell Infect Microbiol 2021; 11:664221. [PMID: 33854985 PMCID: PMC8039388 DOI: 10.3389/fcimb.2021.664221] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 03/09/2021] [Indexed: 12/17/2022] Open
Abstract
Human lymphocytes exposed to Aggregatibacter actinomycetemcomitans (Aa) cytolethal distending toxin (Cdt) undergo cell cycle arrest and apoptosis. In previous studies, we demonstrated that the active Cdt subunit, CdtB, is a potent phosphatidylinositol (PI) 3,4,5-triphosphate phosphatase. Moreover, AaCdt-treated cells exhibit evidence of PI-3-kinase (PI-3K) signaling blockade characterized by reduced levels of PIP3, pAkt, and pGSK3β. We have also demonstrated that PI-3K blockade is a requisite of AaCdt-induced toxicity in lymphocytes. In this study, we extended our observations to include assessment of Cdts from Haemophilus ducreyi (HdCdt) and Campylobacter jejuni (CjCdt). We now report that the CdtB subunit from HdCdt and CjCdt, similar to that of AaCdt, exhibit potent PIP3 phosphatase activity and that Jurkat cells treated with these Cdts exhibit PI-3K signaling blockade: reduced levels of pAkt and pGSK3β. Since non-phosphorylated GSK3β is the active form of this kinase, we compared Cdts for dependence on GSK3β activity. Two GSK3β inhibitors were employed, LY2090314 and CHIR99021; both inhibitors blocked the ability of Cdts to induce cell cycle arrest. We have previously demonstrated that AaCdt induces increases in the CDK inhibitor, p21CIP1/WAF1, and, further, that this was a requisite for toxin-induced cell death via apoptosis. We now demonstrate that HdCdt and CjCdt also share this requirement. It is also noteworthy that p21CIP1/WAF1 was not involved in the ability of the three Cdts to induce cell cycle arrest. Finally, we demonstrate that, like AaCdt, HdCdt is dependent upon the host cell protein, cellugyrin, for its toxicity (and presumably internalization of CdtB); CjCdt was not dependent upon this protein. The implications of these findings as they relate to Cdt’s molecular mode of action are discussed.
Collapse
Affiliation(s)
- Grace Huang
- Department of Basic and Translational Sciences, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, United States
| | - Kathleen Boesze-Battaglia
- Department of Basic and Translational Sciences, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, United States
| | - Lisa P Walker
- Department of Basic and Translational Sciences, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, United States
| | - Ali Zekavat
- Department of Basic and Translational Sciences, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, United States
| | - Zachary P Schaefer
- Department of Microbiology, University of Illinois, Urbana, IL, United States
| | - Steven R Blanke
- Department of Microbiology, University of Illinois, Urbana, IL, United States.,Pathobiology Department, University of Illinois, Urbana, IL, United States.,Biomedical and Translational Sciences Department, University of Illinois, Urbana, IL, United States
| | - Bruce J Shenker
- Department of Basic and Translational Sciences, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, United States
| |
Collapse
|
22
|
Zhu L, Zhu C, Cao S, Zhang Q. Helicobacter hepaticus Induce Colitis in Male IL-10 -/- Mice Dependent by Cytolethal Distending Toxin B and via the Activation of Jak/Stat Signaling Pathway. Front Cell Infect Microbiol 2021; 11:616218. [PMID: 33777833 PMCID: PMC7994616 DOI: 10.3389/fcimb.2021.616218] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 02/09/2021] [Indexed: 01/08/2023] Open
Abstract
It has been well documented that cytolethal distending toxin (CDT) from Helicobacter hepaticus (H. hepaticus), Campylobacter jejuni (C. jejuni) and other Gram-negative intestinal pathogens is linked to the inflammatory bowel disease (IBD). However, the mechanisms underlying the progression of H. hepaticus induced colitis remains unclear. In this study, male B6.129P2-IL10tm1Cgn/J mice were infected by H. hepaticus and ΔCdtB H. hepaticus for 6, 12, 18, and 24 weeks. Histopathology, H. hepaticus colonization levels, expression of inflammatory cytokines, signaling pathways, and content of NO in proximal colon were examined. We found that Cytolethal distending toxin subunit B (CdtB) deletion had no influence on colonization ability of H. hepaticus in colon of B6.129P2-IL10tm1cgn/J mice, and there was no significant difference in abundance of colonic H. hepaticus over infection duration. H. hepaticus aggravated rectocele and proximal colonic inflammation, especially at 24 WPI, while ΔCdtB H. hepaticus could not cause significant symptom. Furthermore, mRNA levels of Il-6, Tnf-α, Il-1β, and iNOS significantly increased in the proximal colon of H. hepaticus-infected mice compared to ΔCdtB H. hepaticus infected group from 12 WPI to 24 WPI. In addition, the elevated content of NO and activated Stat3 and Jak2 in colon were observed in H. hepaticus infected mice. These data demonstrated that CdtB promote colitis development in male B6.129P2-IL10tm1Cgn/J mice by induction of inflammatory response and activation of Jak-Stat signaling pathway.
Collapse
Affiliation(s)
- Liqi Zhu
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Chen Zhu
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Shuyang Cao
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| | - Quan Zhang
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China.,Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| |
Collapse
|
23
|
García A, Fox JG. A One Health Perspective for Defining and Deciphering Escherichia coli Pathogenic Potential in Multiple Hosts. Comp Med 2021; 71:3-45. [PMID: 33419487 PMCID: PMC7898170 DOI: 10.30802/aalas-cm-20-000054] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 08/17/2020] [Accepted: 09/19/2020] [Indexed: 11/05/2022]
Abstract
E. coli is one of the most common species of bacteria colonizing humans and animals. The singularity of E. coli 's genus and species underestimates its multifaceted nature, which is represented by different strains, each with different combinations of distinct virulence factors. In fact, several E. coli pathotypes, or hybrid strains, may be associated with both subclinical infection and a range of clinical conditions, including enteric, urinary, and systemic infections. E. coli may also express DNA-damaging toxins that could impact cancer development. This review summarizes the different E. coli pathotypes in the context of their history, hosts, clinical signs, epidemiology, and control. The pathotypic characterization of E. coli in the context of disease in different animals, including humans, provides comparative and One Health perspectives that will guide future clinical and research investigations of E. coli infections.
Collapse
Key Words
- aa, aggregative adherence
- a/e, attaching and effacing
- aepec, atypical epec
- afa, afimbrial adhesin
- aida-i, adhesin involved in diffuse adherence
- aiec, adherent invasive e. coli
- apec, avian pathogenic e. coli
- atcc, american type culture collection
- bfp, bundle-forming pilus
- cd, crohn disease
- cdt, cytolethal distending toxin gene
- clb, colibactin
- cnf, cytotoxic necrotizing factor
- cs, coli surface (antigens)
- daec, diffusely adhering e. coli
- db, dutch belted
- eae, e. coli attaching and effacing gene
- eaec, enteroaggregative e. coli
- eaf, epec adherence factor (plasmid)
- eahec, entero-aggregative-hemorrhagic e. coli
- east-1, enteroaggregative e. coli heat-stable enterotoxin
- e. coli, escherichia coli
- ed, edema disease
- ehec, enterohemorrhagic e. coli
- eiec, enteroinvasive e. coli
- epec, enteropathogenic e. coli
- esbl, extended-spectrum β-lactamase
- esp, e. coli secreted protein
- etec, enterotoxigenic e. coli
- expec, extraintestinal pathogenic e. coli
- fyua, yersiniabactin receptor gene
- gi, gastrointestinal
- hly, hemolysin
- hus, hemolytic uremic syndrome
- ibd, inflammatory bowel disease
- la, localized adherence
- lee, locus of enterocyte effacement
- lpf, long polar fimbriae
- lt, heat-labile (enterotoxin)
- mlst, multilocus sequence typing
- ndm, new delhi metallo-β-lactamase
- nzw, new zealand white
- pap, pyelonephritis-associated pilus
- pks, polyketide synthase
- sfa, s fimbrial adhesin
- slt, shiga-like toxin
- st, heat-stable (enterotoxin)
- stec, stx-producing e. coli
- stx, shiga toxin
- tepec, typical epec
- upec, uropathogenic e. coli
- uti, urinary tract infection
Collapse
Affiliation(s)
- Alexis García
- Molecular Sciences Research Center, University of Puerto Rico, San Juan, Puerto Rico; Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, Massachusetts; Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, Massachusetts;,
| | - James G Fox
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, Massachusetts
| |
Collapse
|
24
|
Hu S, Niu L, Wu L, Zhu X, Cai Y, Jin D, Yan L, Zhao F. Genomic analysis of Helicobacter himalayensis sp. nov. isolated from Marmota himalayana. BMC Genomics 2020; 21:826. [PMID: 33228534 PMCID: PMC7685656 DOI: 10.1186/s12864-020-07245-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 11/18/2020] [Indexed: 12/17/2022] Open
Abstract
Background Helicobacter himalayensis was isolated from Marmota himalayana in the Qinghai-Tibet Plateau, China, and is a new non-H. pylori species, with unclear taxonomy, phylogeny, and pathogenicity. Results A comparative genomic analysis was performed between the H. himalayensis type strain 80(YS1)T and other the genomes of Helicobacter species present in the National Center for Biotechnology Information (NCBI) database to explore the molecular evolution and potential pathogenicity of H. himalayensis. H. himalayensis 80(YS1)T formed a clade with H. cinaedi and H. hepaticus that was phylogenetically distant from H. pylori. The H. himalayensis genome showed extensive collinearity with H. hepaticus and H. cinaedi. However, it also revealed a low degree of genome collinearity with H. pylori. The genome of 80(YS1)T comprised 1,829,936 bp, with a 39.89% GC content, a predicted genomic island, and 1769 genes. Comparatively, H. himalayensis has more genes for functions in “cell wall/membrane/envelope biogenesis” and “coenzyme transport and metabolism” sub-branches than the other compared helicobacters, and its genome contained 42 virulence factors genes, including that encoding cytolethal distending toxin (CDT). Conclusions We characterized the H. himalayensis 80(YS1)T genome, its phylogenetic position, and its potential pathogenicity. However, further understanding of the pathogenesis of this potentially pathogenic bacterium is required, which might help to manage H. himalayensis-induced diseases. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-020-07245-y.
Collapse
Affiliation(s)
- Shoukui Hu
- Department of Clinical Laboratory, Peking University Shougang Hospital, Beijing, 100144, China
| | - Lina Niu
- Department of Pathogen Biology, School of Basic Medicine and Lifescience, Hainan Medical University, Haikou, 571101, China
| | - Lei Wu
- Department of Clinical Laboratory, Peking University Shougang Hospital, Beijing, 100144, China
| | - Xiaoxue Zhu
- Department of Clinical Laboratory, Peking University Shougang Hospital, Beijing, 100144, China
| | - Yu Cai
- Department of Clinical Laboratory, Peking University Shougang Hospital, Beijing, 100144, China
| | - Dong Jin
- State Key Laboratory of Infectious Disease Prevention and Control, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Changping, Beijing, 102206, China
| | - Linlin Yan
- Department of Clinical Laboratory, Peking University Shougang Hospital, Beijing, 100144, China.
| | - Fan Zhao
- Department of Clinical Laboratory, Peking University Shougang Hospital, Beijing, 100144, China.
| |
Collapse
|
25
|
Maier I, Liu J, Ruegger PM, Deutschmann J, Patsch JM, Helbich TH, Borneman J, Schiestl RH. Intestinal bacterial indicator phylotypes associate with impaired DNA double-stranded break sensors but augmented skeletal bone micro-structure. Carcinogenesis 2020; 41:483-489. [PMID: 31840161 DOI: 10.1093/carcin/bgz204] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 12/07/2019] [Accepted: 12/13/2019] [Indexed: 12/17/2022] Open
Abstract
Intestinal microbiota are considered a sensor for molecular pathways, which orchestrate energy balance, immune responses, and cell regeneration. We previously reported that microbiota restriction promoted higher levels of systemic radiation-induced genotoxicity, proliferative lymphocyte activation, and apoptotic polarization of metabolic pathways. Restricted intestinal microbiota (RM) that harbors increased abundance of Lactobacillus johnsonii (LBJ) has been investigated for bacterial communities that correlated radiation-induced genotoxicity. Indicator phylotypes were more abundant in RM mice and increased in prevalence after whole body irradiation in conventional microbiota (CM) mice, while none of the same ten most abundant phylotypes were different in abundance between CM mice before and after heavy ion irradiation. Muribaculum intestinale was detected highest in female small intestines in RM mice, which were lacking Ureaplasma felinum compared with males, and thus these bacteria could be contributing to the differential amounts of radiation-induced systemic genotoxicity between the CM and RM groups. Helicobacter rodentium and M.intestinale were found in colons in the radiation-resistant CM phenotype. While the expression of interferon-γ was elevated in the small intestine, and lower in blood in CM mice, high-linear energy transfer radiation reduced transforming growth factor-β with peripheral interleukin (IL)-17 in RM mice, particularly in females. We found that female RM mice showed improved micro-architectural bone structure and anti-inflammatory radiation response compared with CM mice at a delayed phase 6 weeks postexposure to particle radiation. However, microbiota restriction reduced inflammatory markers of tumor necrosis factor in marrow, when IL-17 was reduced by intraperitoneal injection of IL-17 neutralizing antibody.
Collapse
Affiliation(s)
- Irene Maier
- Department of Environmental Health Sciences, Fielding School of Public Health, University of California, Los Angeles 650 Charles E. Young Dr. South, Los Angeles, CA, USA
| | - Jared Liu
- Department of Environmental Health Sciences, Fielding School of Public Health, University of California, Los Angeles 650 Charles E. Young Dr. South, Los Angeles, CA, USA
| | - Paul M Ruegger
- Department of Microbiology and Plant Pathology, University of California, Riverside, CA, USA
| | - Julia Deutschmann
- Department for Radiologic Technology, University of Applied Sciences Wiener Neustadt for Business and Engineering Ltd., Lower Austria, Austria
| | - Janina M Patsch
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Währinger Gürtel,Vienna, Austria
| | - Thomas H Helbich
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Währinger Gürtel,Vienna, Austria
| | - James Borneman
- Department of Microbiology and Plant Pathology, University of California, Riverside, CA, USA
| | - Robert H Schiestl
- Department of Environmental Health Sciences, Fielding School of Public Health, University of California, Los Angeles 650 Charles E. Young Dr. South, Los Angeles, CA, USA.,Department of Pathology, University of California, Los Angeles, CA, USA
| |
Collapse
|
26
|
Abstract
Bacterial infections are increasingly being recognized as risk factors for the development of adenocarcinomas. The strong epidemiological evidence linking Helicobacter pylori infection to stomach cancer has paved the way to the demonstration that bacterial infections cause DNA damage in the host cells, initiating transformation. In this regard, the role of bacterial genotoxins has become more relevant. Salmonella enterica serovars Typhi and Paratyphi A have been clinically associated with gallbladder cancer. By harnessing the stem cell potential of cells from healthy human gallbladder explant, we regenerated and propagated the epithelium of this organ in vitro and used these cultures to model S. Paratyphi A infection. This study demonstrates the importance of the typhoid toxin, encoded only by these specific serovars, in causing genomic instability in healthy gallbladder cells, posing intoxicated cells at risk of malignant transformation. Carcinoma of the gallbladder (GBC) is the most frequent tumor of the biliary tract. Despite epidemiological studies showing a correlation between chronic infection with Salmonella enterica Typhi/Paratyphi A and GBC, the underlying molecular mechanisms of this fatal connection are still uncertain. The murine serovar Salmonella Typhimurium has been shown to promote transformation of genetically predisposed cells by driving mitogenic signaling. However, insights from this strain remain limited as it lacks the typhoid toxin produced by the human serovars Typhi and Paratyphi A. In particular, the CdtB subunit of the typhoid toxin directly induces DNA breaks in host cells, likely promoting transformation. To assess the underlying principles of transformation, we used gallbladder organoids as an infection model for Salmonella Paratyphi A. In this model, bacteria can invade epithelial cells, and we observed host cell DNA damage. The induction of DNA double-strand breaks after infection depended on the typhoid toxin CdtB subunit and extended to neighboring, non-infected cells. By cultivating the organoid derived cells into polarized monolayers in air-liquid interphase, we could extend the duration of the infection, and we observed an initial arrest of the cell cycle that does not depend on the typhoid toxin. Non-infected intoxicated cells instead continued to proliferate despite the DNA damage. Our study highlights the importance of the typhoid toxin in causing genomic instability and corroborates the epidemiological link between Salmonella infection and GBC.
Collapse
|
27
|
Longhi G, van Sinderen D, Ventura M, Turroni F. Microbiota and Cancer: The Emerging Beneficial Role of Bifidobacteria in Cancer Immunotherapy. Front Microbiol 2020; 11:575072. [PMID: 33013813 PMCID: PMC7507897 DOI: 10.3389/fmicb.2020.575072] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 08/17/2020] [Indexed: 12/15/2022] Open
Abstract
Many intestinal bacteria are believed to be involved in various inflammatory and immune processes that influence tumor etiology because of their metabolic properties and their ability to alter the microbiota homeostasis. Although many functions of the microbiota are still unclear, there is compelling experimental evidence showing that the intestinal microbiota is able to modulate carcinogenesis and the response to anticancer therapies, both in the intestinal tract and other body sites. Among the wide variety of gut-colonizing microorganisms, various species belonging to the Bifidobacterium genus are believed to elicit beneficial effects on human physiology and on the host-immune system. Recent findings, based on preclinical mouse models and on human clinical trials, have demonstrated the impact of gut commensals including bifidobacteria on the efficacy of tumor-targeting immunotherapy. Although the underlying molecular mechanisms remain obscure, bifidobacteria and other microorganisms have become a promising aid to immunotherapeutic procedures that are currently applied to treat cancer. The present review focuses on strategies to recruit the microbiome in order to enhance anticancer responses and develop therapies aimed at fighting the onset and progression of malignancies.
Collapse
Affiliation(s)
- Giulia Longhi
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Douwe van Sinderen
- Alimentary Pharmabotic Centre (APC) Microbiome Institute and School of Microbiology, Bioscience Institute, National University of Ireland, Cork, Ireland
| | - Marco Ventura
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy.,Microbiome Research Hub, University of Parma, Parma, Italy
| | - Francesca Turroni
- Laboratory of Probiogenomics, Department of Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy.,Microbiome Research Hub, University of Parma, Parma, Italy
| |
Collapse
|
28
|
Gut Microbiota and Colon Cancer: A Role for Bacterial Protein Toxins? Int J Mol Sci 2020; 21:ijms21176201. [PMID: 32867331 PMCID: PMC7504354 DOI: 10.3390/ijms21176201] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/25/2020] [Accepted: 08/26/2020] [Indexed: 02/06/2023] Open
Abstract
Accumulating evidence indicates that the human intestinal microbiota can contribute to the etiology of colorectal cancer. Triggering factors, including inflammation and bacterial infections, may favor the shift of the gut microbiota from a mutualistic to a pro-carcinogenic configuration. In this context, certain bacterial pathogens can exert a pro-tumoral activity by producing enzymatically-active protein toxins that either directly induce host cell DNA damage or interfere with essential host cell signaling pathways involved in cell proliferation, apoptosis, and inflammation. This review is focused on those toxins that, by mimicking carcinogens and cancer promoters, could represent a paradigm for bacterially induced carcinogenesis.
Collapse
|
29
|
Pons BJ, Loiseau N, Hashim S, Tadrist S, Mirey G, Vignard J. Functional Study of Haemophilus ducreyi Cytolethal Distending Toxin Subunit B. Toxins (Basel) 2020; 12:toxins12090530. [PMID: 32825080 PMCID: PMC7551728 DOI: 10.3390/toxins12090530] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/13/2020] [Accepted: 08/17/2020] [Indexed: 12/17/2022] Open
Abstract
The Cytolethal Distending Toxin (CDT) is produced by many Gram-negative pathogenic bacteria responsible for major foodborne diseases worldwide. CDT induces DNA damage and cell cycle arrest in host-cells, eventually leading to senescence or apoptosis. According to structural and sequence comparison, the catalytic subunit CdtB is suggested to possess both nuclease and phosphatase activities, carried by a single catalytic site. However, the impact of each activity on cell-host toxicity is yet to be characterized. Here, we analyze the consequences of cell exposure to different CDT mutated on key CdtB residues, focusing on cell viability, cell cycle defects, and DNA damage induction. A first class of mutant, devoid of any activity, targets putative catalytic (H160A), metal binding (D273R), and DNA binding residues (R117A-R144A-N201A). The second class of mutants (A163R, F156-T158, and the newly identified G114T), which gathers mutations on residues potentially involved in lipid substrate binding, has only partially lost its toxic effects. However, their defects are alleviated when CdtB is artificially introduced inside cells, except for the F156-T158 double mutant that is defective in nuclear addressing. Therefore, our data reveal that CDT toxicity is mainly correlated to CdtB nuclease activity, whereas phosphatase activity may probably be involved in CdtB intracellular trafficking.
Collapse
Affiliation(s)
| | | | | | | | - Gladys Mirey
- Correspondence: (G.M.); (J.V.); Tel.: +33-582-066-338 (G.M.)
| | - Julien Vignard
- Correspondence: (G.M.); (J.V.); Tel.: +33-582-066-338 (G.M.)
| |
Collapse
|
30
|
Tsvetikova SA, Koshel EI. Microbiota and cancer: host cellular mechanisms activated by gut microbial metabolites. Int J Med Microbiol 2020; 310:151425. [PMID: 32423739 DOI: 10.1016/j.ijmm.2020.151425] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 03/25/2020] [Accepted: 04/13/2020] [Indexed: 12/13/2022] Open
|
31
|
Silbergleit M, Vasquez AA, Miller CJ, Sun J, Kato I. Oral and intestinal bacterial exotoxins: Potential linked to carcinogenesis. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 171:131-193. [PMID: 32475520 DOI: 10.1016/bs.pmbts.2020.02.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Growing evidence suggests that imbalances in resident microbes (dysbiosis) can promote chronic inflammation, immune-subversion, and production of carcinogenic metabolites, thus leading to neoplasia. Yet, evidence to support a direct link of individual bacteria species to human sporadic cancer is still limited. This chapter focuses on several emerging bacterial toxins that have recently been characterized for their potential oncogenic properties toward human orodigestive cancer and the presence of which in human tissue samples has been documented. These include cytolethal distending toxins produced by various members of gamma and epsilon Proteobacteria, Dentilisin from mammalian oral Treponema, Pasteurella multocida toxin, two Fusobacterial toxins, FadA and Fap2, Bacteroides fragilis toxin, colibactin, cytotoxic necrotizing factors and α-hemolysin from Escherichia coli, and Salmonella enterica AvrA. It was clear that these bacterial toxins have biological activities to induce several hallmarks of cancer. Some toxins directly interact with DNA or chromosomes leading to their breakdowns, causing mutations and genome instability, and others modulate cell proliferation, replication and death and facilitate immune evasion and tumor invasion, prying specific oncogene and tumor suppressor pathways, such as p53 and β-catenin/Wnt. In addition, most bacterial toxins control tumor-promoting inflammation in complex and diverse mechanisms. Despite growing laboratory evidence to support oncogenic potential of selected bacterial toxins, we need more direct evidence from human studies and mechanistic data from physiologically relevant experimental animal models, which can reflect chronic infection in vivo, as well as take bacterial-bacterial interactions among microbiome into consideration.
Collapse
Affiliation(s)
| | - Adrian A Vasquez
- Department of Civil and Environmental Engineering, Wayne State University, Healthy Urban Waters, Detroit, MI, United States
| | - Carol J Miller
- Department of Civil and Environmental Engineering, Wayne State University, Healthy Urban Waters, Detroit, MI, United States
| | - Jun Sun
- Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Ikuko Kato
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, United States; Department of Pathology, Wayne State University School of Medicine, Detroit, MI, United States.
| |
Collapse
|
32
|
Péré-Védrenne C, He W, Azzi-Martin L, Prouzet-Mauléon V, Buissonnière A, Cardinaud B, Lehours P, Mégraud F, Grosset CF, Ménard A. The Nuclear Remodeling Induced by Helicobacter Cytolethal Distending Toxin Involves MAFB Oncoprotein. Toxins (Basel) 2020; 12:toxins12030174. [PMID: 32178359 PMCID: PMC7150770 DOI: 10.3390/toxins12030174] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/25/2020] [Accepted: 03/02/2020] [Indexed: 12/12/2022] Open
Abstract
Enterohepatic Helicobacters, such as Helicobacter hepaticus and Helicobacter pullorum, are associated with several intestinal and hepatic diseases. Their main virulence factor is the cytolethal distending toxin (CDT). In the present study, whole genome microarray-based identification of differentially expressed genes was performed in vitro in HT-29 intestinal cells while following the ectopic expression of the active CdtB subunit of H. hepaticus CDT. A CdtB-dependent upregulation of the V-maf musculoaponeurotic fibrosarcoma oncogene homolog B (MAFB) gene encoding the MAFB oncoprotein was found, as well as the CdtB-dependent regulation of several MAFB target genes. The transduction and coculture experiments confirmed MAFB mRNA and protein induction in response to CDT and its CdtB subunit in intestinal and hepatic cell lines. An analysis of MAFB protein subcellular localization revealed a strong nuclear and perinuclear localization in the CdtB-distended nuclei in intestinal and hepatic cells. MAFB was also detected at the cell periphery of the CdtB-induced lamellipodia in some cells. The silencing of MAFB changed the cellular response to CDT with the formation of narrower lamellipodia, a reduction of the increase in nucleus size, and the formation of less γH2AX foci, the biomarker for DNA double-strand breaks. Taken together, these data show that the CDT of enterohepatic Helicobacters modulates the expression of the MAFB oncoprotein, which is translocated in the nucleus and is associated with the remodeling of the nuclei and actin cytoskeleton.
Collapse
Affiliation(s)
- Christelle Péré-Védrenne
- Université de Bordeaux, INSERM—Institut National de la Santé et de la Recherche Médicale, BaRITOn—Bordeaux Research in Translational Oncology, UMR1053, 33076 Bordeaux, France; (C.P.-V.); (W.H.); (L.A.-M.); (A.B.); (P.L.); (F.M.)
| | - Wencan He
- Université de Bordeaux, INSERM—Institut National de la Santé et de la Recherche Médicale, BaRITOn—Bordeaux Research in Translational Oncology, UMR1053, 33076 Bordeaux, France; (C.P.-V.); (W.H.); (L.A.-M.); (A.B.); (P.L.); (F.M.)
| | - Lamia Azzi-Martin
- Université de Bordeaux, INSERM—Institut National de la Santé et de la Recherche Médicale, BaRITOn—Bordeaux Research in Translational Oncology, UMR1053, 33076 Bordeaux, France; (C.P.-V.); (W.H.); (L.A.-M.); (A.B.); (P.L.); (F.M.)
| | - Valérie Prouzet-Mauléon
- Université de Bordeaux, TBMCore, CRISP’edit, TBMcore CNRS-Centre National de la Recherche Scientifique UMS3427/INSERM—Institut National de la Santé et de la Recherche Médicale US005, 33076 Bordeaux, France;
- Université de Bordeaux, INSERM—Institut National de la Santé et de la Recherche Médicale, ACTION, U1218, Institut Bergonié, 33076 Bordeaux, France;
| | - Alice Buissonnière
- Université de Bordeaux, INSERM—Institut National de la Santé et de la Recherche Médicale, BaRITOn—Bordeaux Research in Translational Oncology, UMR1053, 33076 Bordeaux, France; (C.P.-V.); (W.H.); (L.A.-M.); (A.B.); (P.L.); (F.M.)
| | - Bruno Cardinaud
- Université de Bordeaux, INSERM—Institut National de la Santé et de la Recherche Médicale, ACTION, U1218, Institut Bergonié, 33076 Bordeaux, France;
- Bordeaux INP, ENSTBB, F-33000 Bordeaux, France
| | - Philippe Lehours
- Université de Bordeaux, INSERM—Institut National de la Santé et de la Recherche Médicale, BaRITOn—Bordeaux Research in Translational Oncology, UMR1053, 33076 Bordeaux, France; (C.P.-V.); (W.H.); (L.A.-M.); (A.B.); (P.L.); (F.M.)
- CHU Pellegrin, National Reference Center for Campylobacters and Helicobacters, 33076 Bordeaux, France
| | - Francis Mégraud
- Université de Bordeaux, INSERM—Institut National de la Santé et de la Recherche Médicale, BaRITOn—Bordeaux Research in Translational Oncology, UMR1053, 33076 Bordeaux, France; (C.P.-V.); (W.H.); (L.A.-M.); (A.B.); (P.L.); (F.M.)
- CHU Pellegrin, National Reference Center for Campylobacters and Helicobacters, 33076 Bordeaux, France
| | - Christophe F. Grosset
- Université de Bordeaux, INSERM—Institut National de la Santé et de la Recherche Médicale, BMGIC—Biotherapy of Genetic Diseases, Inflammatory Disorders and Cancer, U1035, miRCaDe Team, 33076 Bordeaux, France;
| | - Armelle Ménard
- Université de Bordeaux, INSERM—Institut National de la Santé et de la Recherche Médicale, BaRITOn—Bordeaux Research in Translational Oncology, UMR1053, 33076 Bordeaux, France; (C.P.-V.); (W.H.); (L.A.-M.); (A.B.); (P.L.); (F.M.)
- CHU Pellegrin, National Reference Center for Campylobacters and Helicobacters, 33076 Bordeaux, France
- Correspondence: ; Tel.: +33-(0)-5-5757-1288
| |
Collapse
|
33
|
Shenker BJ, Walker LM, Zekavat Z, Ojcius DM, Huang PR, Boesze-Battaglia K. Cytolethal distending toxin-induced release of interleukin-1β by human macrophages is dependent upon activation of glycogen synthase kinase 3β, spleen tyrosine kinase (Syk) and the noncanonical inflammasome. Cell Microbiol 2020; 22:e13194. [PMID: 32068949 DOI: 10.1111/cmi.13194] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 01/09/2020] [Accepted: 01/24/2020] [Indexed: 12/19/2022]
Abstract
Cytolethal distending toxins (Cdt) are a family of toxins produced by several human pathogens which infect mucocutaneous tissue and induce inflammatory disease. We have previously demonstrated that the Aggregatibacter actinomycetemcomitans Cdt induces a pro-inflammatory response from human macrophages which involves activation of the NLRP3 inflammasome. We now demonstrate that in addition to activating caspase-1 (canonical inflammasome), Cdt treatment leads to caspase-4 activation and involvement of the noncanonical inflammasome. Cdt-treated cells exhibit pyroptosis characterised by cleavage of gasdermin-D (GSDMD), release of HMGB1 at 24 hr and LDH at 48 hr. Inhibition of either the canonical (caspase-1) or noncanonical (caspase-4) inflammasome blocks both Cdt-induced release of IL-1β and induction of pyroptosis. Analysis of upstream events indicates that Cdt induces Syk phosphorylation (activation); furthermore, blockade of Syk expression and inhibition of pSyk activity inhibit both Cdt-induced cytokine release and pyroptosis. Finally, we demonstrate that increases in pSyk are dependent upon Cdt-induced activation of GSK3β. These studies advance our understanding of Cdt function and provide new insight into the virulence potential of Cdt in mediating the pathogenesis of disease caused by Cdt-producing organisms such as A. actinomycetemcomitans.
Collapse
Affiliation(s)
- Bruce J Shenker
- Department of Pathology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania, USA
| | - Lisa M Walker
- Department of Pathology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania, USA
| | - Zeyed Zekavat
- Department of Pathology, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania, USA
| | - David M Ojcius
- Department of Biomedical Sciences, University of the Pacific Arthur A. Dugoni School of Dentistry, San Francisco, California, USA
| | - Pei-Rong Huang
- Center for Molecular and Clinical Immunology, Chang Gung University, Taoyuan, Taiwan
| | - Kathleen Boesze-Battaglia
- Department of Biochemistry, University of Pennsylvania School of Dental Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
34
|
Martin OC, Frisan T. Bacterial Genotoxin-Induced DNA Damage and Modulation of the Host Immune Microenvironment. Toxins (Basel) 2020; 12:E63. [PMID: 31973033 PMCID: PMC7076804 DOI: 10.3390/toxins12020063] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 01/14/2020] [Accepted: 01/18/2020] [Indexed: 01/10/2023] Open
Abstract
: Bacterial genotoxins (BTGX) induce DNA damage, which results in senescence or apoptosis of the target cells if not properly repaired. Three BTGXs have been identified: the cytolethal distending toxin (CDT) family produced by several Gram-negative bacteria, the typhoid toxin produced by several Salmonella enterica serovars, and colibactin, a peptide-polyketide, produced mainly by the phylogenetic group B2 Escherichia coli. The cellular responses induced by BTGXs resemble those of well-characterized carcinogenic agents, and several lines of evidence indicate that bacteria carrying genotoxin genes can contribute to tumor development under specific circumstances. Given their unusual mode of action, it is still enigmatic why these effectors have been acquired by microbes and what is their role in the context of the biology of the producing bacterium, since it is unlikely that their primary purpose is to induce/promote cancer in the mammalian host. In this review, we will discuss the possibility that the DNA damage induced by BTGX modulates the host immune response, acting as immunomodulator, leading to the establishment of a suitable niche for the producing bacterium. We will further highlight open questions that remain to be solved regarding the biology of this unusual family of bacterial toxins.
Collapse
Affiliation(s)
- Océane C.B. Martin
- Univ. Bordeaux, INSERM, UMR1053 Bordeaux Research in Translational Oncology, BaRITOn, 33320 Bordeaux, France;
| | - Teresa Frisan
- Department of Cell and Molecular Biology Karolinska Institutet, 17177 Stockholm, Sweden
- Umeå Center for Microbial Research (UCMR), Umeå University, 90187 Umeå, Sweden
- Department of Molecular Biology, Umeå University, 90187 Umeå, Sweden
| |
Collapse
|
35
|
Dichotomous regulation of group 3 innate lymphoid cells by nongastric Helicobacter species. Proc Natl Acad Sci U S A 2019; 116:24760-24769. [PMID: 31740609 DOI: 10.1073/pnas.1908128116] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Intestinal innate lymphoid cells (ILCs) contribute to the protective immunity and homeostasis of the gut, and the microbiota are critically involved in shaping ILC function. However, the role of the gut microbiota in regulating ILC development and maintenance still remains elusive. Here, we identified opposing effects on ILCs by two Helicobacter species, Helicobacter apodemus and Helicobacter typhlonius, isolated from immunocompromised mice. We demonstrated that the introduction of both Helicobacter species activated ILCs and induced gut inflammation; however, these Helicobacter species negatively regulated RORγt+ group 3 ILCs (ILC3s), especially T-bet+ ILC3s, and diminished their proliferative capacity. Thus, these findings underscore a previously unknown dichotomous regulation of ILC3s by Helicobacter species, and may serve as a model for further investigations to elucidate the host-microbe interactions that critically sustain the maintenance of intestinal ILC3s.
Collapse
|
36
|
Cytolethal Distending Toxin Subunit B: A Review of Structure-Function Relationship. Toxins (Basel) 2019; 11:toxins11100595. [PMID: 31614800 PMCID: PMC6832162 DOI: 10.3390/toxins11100595] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/08/2019] [Accepted: 10/09/2019] [Indexed: 01/27/2023] Open
Abstract
The Cytolethal Distending Toxin (CDT) is a bacterial virulence factor produced by several Gram-negative pathogenic bacteria. These bacteria, found in distinct niches, cause diverse infectious diseases and produce CDTs differing in sequence and structure. CDTs have been involved in the pathogenicity of the associated bacteria by promoting persistent infection. At the host-cell level, CDTs cause cell distension, cell cycle block and DNA damage, eventually leading to cell death. All these effects are attributable to the catalytic CdtB subunit, but its exact mode of action is only beginning to be unraveled. Sequence and 3D structure analyses revealed similarities with better characterized proteins, such as nucleases or phosphatases, and it has been hypothesized that CdtB exerts a biochemical activity close to those enzymes. Here, we review the relationships that have been established between CdtB structure and function, particularly by mutation experiments on predicted key residues in different experimental systems. We discuss the relevance of these approaches and underline the importance of further study in the molecular mechanisms of CDT toxicity, particularly in the context of different pathological conditions.
Collapse
|
37
|
Han X, Huang T, Han J. Cytokines derived from innate lymphoid cells assist Helicobacter hepaticus to aggravate hepatocellular tumorigenesis in viral transgenic mice. Gut Pathog 2019; 11:23. [PMID: 31123503 PMCID: PMC6521485 DOI: 10.1186/s13099-019-0302-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 04/27/2019] [Indexed: 02/08/2023] Open
Abstract
Background Recently, intestinal microbiome has been involved in hepatic diseases due to the immunologic and metabolic communication between liver and intestine. Initiation of hepatocellular carcinoma (HCC) frequently attributes to conspiracy between immune cells and infectious carcinogens. Here, the hypothesis that the tumorigenesis of HCC with HBV infection will be aggravated by specific intestinal bacteria was verified in viral transgenic mouse models. Methods Comparative 16S rRNA sequencing was adopted to observe the intestinal enrichment of Helicobacter hepaticus in HCC. Oral administration of Helicobacter hepaticus was carried out to evaluate its hepatic carcinogenic effect in HBV transgenic mice or wildtype C57BL/6. The livers of experimental mice were collected and examined for the degree of tumorigenesis. Results We found that Helicobacter hepaticus more likely colonized at lower colon of HBV-infected mice with HCC, compared with C57BL/6 and HBV-infected mice without neoplasm. Pretreatment of Helicobacter hepaticus in transgenic mice aggravated tumor formation, with higher incidence, more tumor nodule and higher serum AFP. Then, a cytokines expression patterns with inclined IFN-γ, IFN-γR1, IL-17 and IL-23 was found in HBV-infected mice with Helicobacter hepaticus. Furthermore, innate lymphoid cells, especially Th17 and NK cells which can secret IL-17 and IFN-γ respectively, might be recruited by Helicobacter hepaticus cooperated with HBV. Besides, increased expression of CD69, NKG2D and IFN-γ showed activation of cytokine production in intrahepatic NK cells. Finally, IFN-γ decreased E-cadherin expression through p-STAT1 pathway, resulting in epithelial–mesenchymal transition with inclined expression of Snail2, SIP1 and CXCR4 in vitro. p-STAT1 inhibitor was able to reverse the expression of E-cadherin and EMT resulted from IFN-γ function on HBsAg-positive hepatocytes. Conclusions Helicobacter hepaticus generate a detrimental immune microenvironment by IFN-γ/p-STAT1 axis which can promote the tumorigenesis of hepatitis B via recruiting innate lymphoid cells. Electronic supplementary material The online version of this article (10.1186/s13099-019-0302-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiao Han
- 1Department of Experiment, Tumor Hospital Affiliated to Guangxi Medical University, 71# Hedi Road, Nanning, 530021 China
| | - Tianren Huang
- 1Department of Experiment, Tumor Hospital Affiliated to Guangxi Medical University, 71# Hedi Road, Nanning, 530021 China
| | - Junqing Han
- 2Department of Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong University, 324# Jingwu Road, Jinan, 250021 China
| |
Collapse
|
38
|
Ge Z, Feng Y, Sheh A, Muthupalani S, Gong G, Chawanthayatham S, Essigmann JM, Fox JG. Mutagenicity of Helicobacter hepaticus infection in the lower bowel mucosa of 129/SvEv Rag2 -/- Il10 -/- gpt delta mice is influenced by sex. Int J Cancer 2019; 145:1042-1054. [PMID: 30977112 DOI: 10.1002/ijc.32332] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 04/05/2019] [Indexed: 12/29/2022]
Abstract
Inflammatory bowel disease and colonic tumors induced by Helicobacter hepaticus (Hh) infection in susceptible mouse strains are utilized to dissect the mechanisms underlying similar human diseases. In our study, infection with genotoxic cytolethal distending toxin-producing Hh in 129/SvEv Rag2-/- Il10-/- gpt delta (RagIl10gpt) mice of both sexes for 21 weeks induced significantly more severe cecal and colonic pathology compared to uninfected controls. The mutation frequencies in the infected RagIl10gpt males were 2.1-fold higher for the cecum and 1.7-fold higher for the colon than male RagIl10gpt controls. In addition, there was a 12.5-fold increase of G:C-to-T:A transversions in the colon of Hh-infected males compared to controls. In contrast, there was no statistical significance in mutation frequencies between infected female Rag2Il10gpt mice and controls. Moreover, Hh infection in RagIl10gpt males significantly up-regulated transcription of Tnfα and iNos, and decreased mRNA levels of cecal Atm compared to the infected females; there was no significant difference in mRNA levels of Il-22, Il-17A, Ifnγ and Atr between the infected males and females. Significantly higher levels of cecal and colonic iNos expression and γH2AX-positive epithelial cells (a biomarker for double-strand DNA breaks [DSB]) in Hh-infected Rag2Il10gpt males vs. Hh-infected females were noted. Finally, Hh infection and associated inflammation increased levels of intestinal mucosa-associated genotoxic colibactin-producing pks+ Escherichia coli. Elevated Tnfα and iNos responses and bacterial genotoxins, in concert with suppression of the DSB repair responses, may have promoted mutagenesis in the lower bowel mucosa of Hh-infected male RagIl10gpt mice.
Collapse
Affiliation(s)
- Zhongming Ge
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yan Feng
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Alexander Sheh
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Guanyu Gong
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - John M Essigmann
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - James G Fox
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
39
|
He Z, Gharaibeh RZ, Newsome RC, Pope JL, Dougherty MW, Tomkovich S, Pons B, Mirey G, Vignard J, Hendrixson DR, Jobin C. Campylobacter jejuni promotes colorectal tumorigenesis through the action of cytolethal distending toxin. Gut 2019; 68:289-300. [PMID: 30377189 PMCID: PMC6352414 DOI: 10.1136/gutjnl-2018-317200] [Citation(s) in RCA: 267] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 09/12/2018] [Accepted: 10/04/2018] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Campylobacter jejuni produces a genotoxin, cytolethal distending toxin (CDT), which has DNAse activity and causes DNA double-strand breaks. Although C. jejuni infection has been shown to promote intestinal inflammation, the impact of this bacterium on carcinogenesis has never been examined. DESIGN Germ-free (GF) ApcMin/+ mice, fed with 1% dextran sulfate sodium, were used to test tumorigenesis potential of CDT-producing C. jejuni. Cells and enteroids were exposed to bacterial lysates to determine DNA damage capacity via γH2AX immunofluorescence, comet assay and cell cycle assay. To examine the interplay of CDT-producing C. jejuni, gut microbiome and host in tumorigenesis, colonic RNA-sequencing and faecal 16S rDNA sequencing were performed. Rapamycin was administrated to investigate the prevention of CDT-producing C. jejuni-induced tumorigenesis. RESULTS GF ApcMin/+ mice colonised with human clinical isolate C. jejuni81-176 developed significantly more and larger tumours when compared with uninfected mice. C. jejuni with a mutated cdtB subunit, mutcdtB, attenuated C. jejuni-induced tumorigenesis in vivo and decreased DNA damage response in cells and enteroids. C. jejuni infection induced expression of hundreds of colonic genes, with 22 genes dependent on the presence of cdtB. The C. jejuni-infected group had a significantly different microbial gene expression profile compared with the mutcdtB group as shown by metatranscriptomic data, and different microbial communities as measured by 16S rDNA sequencing. Finally, rapamycin could diminish the tumorigenic capability of C. jejuni. CONCLUSION Human clinical isolate C. jejuni 81-176 promotes colorectal cancer and induces changes in microbial composition and transcriptomic responses, a process dependent on CDT production.
Collapse
Affiliation(s)
- Zhen He
- Department of Medicine, University of Florida, Gainesville, Florida, USA,Department of Colorectal Surgery, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Raad Z Gharaibeh
- Department of Medicine, University of Florida, Gainesville, Florida, USA
| | - Rachel C Newsome
- Department of Medicine, University of Florida, Gainesville, Florida, USA
| | - Jllian L Pope
- Department of Medicine, University of Florida, Gainesville, Florida, USA
| | | | - Sarah Tomkovich
- Department of Medicine, University of Florida, Gainesville, Florida, USA
| | - Benoit Pons
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, ENVT, INP- Purpan, UPS, Toulouse, France
| | - Gladys Mirey
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, ENVT, INP- Purpan, UPS, Toulouse, France
| | - Julien Vignard
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, ENVT, INP- Purpan, UPS, Toulouse, France
| | - David R Hendrixson
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Christian Jobin
- Department of Medicine, University of Florida, Gainesville, Florida, USA,Department of Anatomy and Cell Biology, University of Florida, Gainesville, Florida, USA,Department of Infectious Diseases and Immunology, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
40
|
Mannion A, Shen Z, Fox JG. Comparative genomics analysis to differentiate metabolic and virulence gene potential in gastric versus enterohepatic Helicobacter species. BMC Genomics 2018; 19:830. [PMID: 30458713 PMCID: PMC6247508 DOI: 10.1186/s12864-018-5171-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 10/15/2018] [Indexed: 02/08/2023] Open
Abstract
Background The genus Helicobacter are gram-negative, microaerobic, flagellated, mucus-inhabiting bacteria associated with gastrointestinal inflammation and classified as gastric or enterohepatic Helicobacter species (EHS) according to host species and colonization niche. While there are over 30 official species, little is known about the physiology and pathogenic mechanisms of EHS, which account for most in the genus, as well as what genetic factors differentiate gastric versus EHS, given they inhabit different hosts and colonization niches. The objective of this study was to perform a whole-genus comparative analysis of over 100 gastric versus EHS genomes in order to identify genetic determinants that distinguish these Helicobacter species and provide insights about their evolution/adaptation to different hosts, colonization niches, and mechanisms of virulence. Results Whole-genome phylogeny organized Helicobacter species according to their presumed gastric or EHS classification. Analysis of orthologs revealed substantial heterogeneity in physiological and virulence-related genes between gastric and EHS genomes. Metabolic reconstruction predicted that unlike gastric species, EHS appear asaccharolytic and dependent on amino/organic acids to fuel metabolism. Additionally, gastric species lack de novo biosynthetic pathways for several amino acids and purines found in EHS and instead rely on environmental uptake/salvage pathways. Comparison of virulence factor genes between gastric and EHS genomes identified overlapping yet distinct profiles and included canonical cytotoxins, outer membrane proteins, secretion systems, and survival factors. Conclusions The major differences in predicted metabolic function suggest gastric species and EHS may have evolved for survival in the nutrient-rich stomach versus the nutrient-devoid environments, respectively. Contrasting virulence factor gene profiles indicate gastric species and EHS may utilize different pathogenic mechanisms to chronically infect hosts and cause inflammation and tissue damage. The findings from this study provide new insights into the genetic differences underlying gastric versus EHS and support the need for future experimental studies to characterize these pathogens. Electronic supplementary material The online version of this article (10.1186/s12864-018-5171-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anthony Mannion
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Zeli Shen
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - James G Fox
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
41
|
van der Mee-Marquet NL, Bénéjat L, Diene SM, Lemaignen A, Gaïa N, Smet A, Haesebrouck F, Cherkaoui A, Ducournau A, Lacomme S, Gontier E, Bernard L, Mégraud F, Goudeau A, Lehours P, François P. A Potential New Human Pathogen Belonging to Helicobacter Genus, Identified in a Bloodstream Infection. Front Microbiol 2017; 8:2533. [PMID: 29326675 PMCID: PMC5741639 DOI: 10.3389/fmicb.2017.02533] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 12/05/2017] [Indexed: 01/20/2023] Open
Abstract
We isolated from aerobic and anaerobic blood culture bottles from a febrile patient, a Helicobacter-like Gram negative, rod-shaped bacterium that MALDI-TOF MS failed to identify. Blood agar cultures incubated in a microaerobic atmosphere revealed a motile Gram negative rod, which was oxidase, catalase, nitrate reductase, esterase, and alkaline phosphatase positive. It grew at 42°C with no detectable urease activity. Antimicrobial susceptibility testing showed that the organism was susceptible to beta-lactams, gentamicin, erythromycin, and tetracycline but resistant to ciprofloxacin. Electronic microscopy analysis revealed a 3 × 0.5 μm curved rod bacterium harboring two sheathed amphitrichous flagella. Whole genome sequencing revealed a genome 1,708,265 base-pairs long with a GC content of 37.80% and a total of 1,697 coding sequences. The genomic analyses using the nucleotide sequences of the 16S rRNA gene, hsp60 and gyrB genes, as well as the GyrA protein sequence, and the results of Average Nucleotide Identity and in silico DNA-DNA hybridization suggest evidence for a novel Helicobacter species close to Helicobacter equorum and belonging to the group of enterohepatic Helicobacter species. As soon as the particular peptide mass fingerprint of this pathogen is added to the spectral databases, MALDI-TOF MS technology will improve its identification from clinical specimens, especially in case of “sterile infection”. We propose to associate the present strain with the Latin name of the place of isolation; Caesarodunum (Tours, France) and suggest “Helicobacter caesarodunensis” for further description of this new bacterium.
Collapse
Affiliation(s)
- Nathalie L van der Mee-Marquet
- Service de Bactériologie, Virologie et Hygiène, Hôpital Trousseau, Réseau des Hygiénistes du Centre, CPIAS Centre Val de Loire, Centre Hospitalier Régional Universitaire, and UMR 1282 Infectiologie Santé Publique, Université François-Rabelais, Tours, France
| | - Lucie Bénéjat
- Laboratoire de Bactériologie, Centre National de Référence des Campylobacters et des Hélicobacters, Bordeaux, France
| | - Seydina M Diene
- Faculté de Médecine et de Pharmacie, URMITE, Aix-Marseille Université, UMR 63, Centre National de la Recherche Scientifique 7278, IRD 198, Institut National de la Santé et de la Recherche Médicale, IHU-Méditerranée Infection, Marseille, France
| | - Adrien Lemaignen
- Service de Médecine Interne et des Maladies Infectieuses, Centre Hospitalier Régional Universitaire, Hôpital Bretonneau, Tours, France
| | - Nadia Gaïa
- Genomic Research Laboratory, University Hospital of Geneva, Geneva, Switzerland
| | - Annemieke Smet
- Laboratory Experimental Medicine and Pediatrics, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Freddy Haesebrouck
- Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Abdessalam Cherkaoui
- Bacteriology Laboratory, Division of Laboratory Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Astrid Ducournau
- Laboratoire de Bactériologie, Centre National de Référence des Campylobacters et des Hélicobacters, Bordeaux, France
| | - Sabrina Lacomme
- Bordeaux Imaging Center, Imagerie Electronique, UMS 3420 Centre National de la Recherche Scientifique US4 Institut National de la Santé et de la Recherche Médicale Université de Bordeaux, Bordeaux, France
| | - Etienne Gontier
- Bordeaux Imaging Center, Imagerie Electronique, UMS 3420 Centre National de la Recherche Scientifique US4 Institut National de la Santé et de la Recherche Médicale Université de Bordeaux, Bordeaux, France
| | - Louis Bernard
- Service de Médecine Interne et des Maladies Infectieuses, Centre Hospitalier Régional Universitaire, Hôpital Bretonneau, Tours, France
| | - Francis Mégraud
- Laboratoire de Bactériologie, Centre National de Référence des Campylobacters et des Hélicobacters, Bordeaux, France
| | - Alain Goudeau
- Service de Bactériologie, Virologie et Hygiène, Hôpital Trousseau, Réseau des Hygiénistes du Centre, CPIAS Centre Val de Loire, Centre Hospitalier Régional Universitaire, and UMR 1282 Infectiologie Santé Publique, Université François-Rabelais, Tours, France
| | - Philippe Lehours
- Laboratoire de Bactériologie, Centre National de Référence des Campylobacters et des Hélicobacters, Bordeaux, France
| | - Patrice François
- Genomic Research Laboratory, University Hospital of Geneva, Geneva, Switzerland
| |
Collapse
|
42
|
Péré-Védrenne C, Flahou B, Loke MF, Ménard A, Vadivelu J. Other Helicobacters, gastric and gut microbiota. Helicobacter 2017; 22 Suppl 1. [PMID: 28891140 DOI: 10.1111/hel.12407] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The current article is a review of the most important and relevant literature published in 2016 and early 2017 on non-Helicobacter pylori Helicobacter infections in humans and animals, as well as interactions between H. pylori and the microbiota of the stomach and other organs. Some putative new Helicobacter species were identified in sea otters, wild boars, dogs, and mice. Many cases of Helicobacter fennelliae and Helicobacter cinaedi infection have been reported in humans, mostly in immunocompromised patients. Mouse models have been used frequently as a model to investigate human Helicobacter infection, although some studies have investigated the pathogenesis of Helicobacters in their natural host, as was the case for Helicobacter suis infection in pigs. Our understanding of both the gastric and gut microbiome has made progress and, in addition, interactions between H. pylori and the microbiome were demonstrated to go beyond the stomach. Some new approaches of preventing Helicobacter infection or its related pathologies were investigated and, in this respect, the probiotic properties of Saccharomyces, Lactobacillus and Bifidobacterium spp. were confirmed.
Collapse
Affiliation(s)
- Christelle Péré-Védrenne
- INSERM, UMR1053 Bordeaux Research in Translational Oncology, BaRITOn, Bordeaux, France.,University of Bordeaux, Bacteriology Laboratory, Bordeaux, France
| | - Bram Flahou
- Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Mun Fai Loke
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Armelle Ménard
- INSERM, UMR1053 Bordeaux Research in Translational Oncology, BaRITOn, Bordeaux, France.,University of Bordeaux, Bacteriology Laboratory, Bordeaux, France
| | - Jamuna Vadivelu
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
43
|
Péré-Védrenne C, Prochazkova-Carlotti M, Rousseau B, He W, Chambonnier L, Sifré E, Buissonnière A, Dubus P, Mégraud F, Varon C, Ménard A. The Cytolethal Distending Toxin Subunit CdtB of Helicobacter hepaticus Promotes Senescence and Endoreplication in Xenograft Mouse Models of Hepatic and Intestinal Cell Lines. Front Cell Infect Microbiol 2017; 7:268. [PMID: 28713773 PMCID: PMC5491915 DOI: 10.3389/fcimb.2017.00268] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 06/06/2017] [Indexed: 12/15/2022] Open
Abstract
Cytolethal distending toxins (CDTs) are common among pathogenic bacteria of the human and animal microbiota. CDTs exert cytopathic effets, via their active CdtB subunit. No clear description of those cytopathic effects has been reported at the cellular level in the target organs in vivo. In the present study, xenograft mouse models of colon and liver cell lines were set up to study the effects of the CdtB subunit of Helicobacter hepaticus. Conditional transgenic cell lines were established, validated in vitro and then engrafted into immunodeficient mice. After successful engraftment, mice were treated with doxycyclin to induce the expression of transgenes (red fluorescent protein, CdtB, and mutated CdtB). For both engrafted cell lines, results revealed a delayed tumor growth and a reduced tumor weight in CdtB-expressing tumors compared to controls. CdtB-derived tumors showed γ-H2AX foci formation, an increase in apoptosis, senescence, p21 and Ki-67 nuclear antigen expression. No difference in proliferating cells undergoing mitosis (phospho-histone H3) was observed. CdtB intoxication was also associated with an overexpression of cytokeratins in cells at the invasive front of the tumor as well as an increase in ploidy. All these features are hallmarks of endoreplication, as well as aggressiveness in cancer. These effects were dependent on the histidine residue at position 265 of the CdtB, underlying the importance of this residue in CdtB catalytic activity. Taken together, these data indicate that the CdtB triggers senescence and cell endoreplication leading to giant polyploid cells in these xenograft mouse models.
Collapse
Affiliation(s)
- Christelle Péré-Védrenne
- Institut National de la Santé et de la Recherche Médicale, UMR1053 Bordeaux Research in Translational Oncology, BaRITOnBordeaux, France
- UMR1053 Bordeaux Research in Translational Oncology, BaRITOn, Université de BordeauxBordeaux, France
| | - Martina Prochazkova-Carlotti
- Institut National de la Santé et de la Recherche Médicale, UMR1053 Bordeaux Research in Translational Oncology, BaRITOnBordeaux, France
- UMR1053 Bordeaux Research in Translational Oncology, BaRITOn, Université de BordeauxBordeaux, France
| | - Benoit Rousseau
- Service Commun des Animaleries, Université de BordeauxBordeaux, France
| | - Wencan He
- Institut National de la Santé et de la Recherche Médicale, UMR1053 Bordeaux Research in Translational Oncology, BaRITOnBordeaux, France
- UMR1053 Bordeaux Research in Translational Oncology, BaRITOn, Université de BordeauxBordeaux, France
| | - Lucie Chambonnier
- Institut National de la Santé et de la Recherche Médicale, UMR1053 Bordeaux Research in Translational Oncology, BaRITOnBordeaux, France
- UMR1053 Bordeaux Research in Translational Oncology, BaRITOn, Université de BordeauxBordeaux, France
| | - Elodie Sifré
- Institut National de la Santé et de la Recherche Médicale, UMR1053 Bordeaux Research in Translational Oncology, BaRITOnBordeaux, France
- UMR1053 Bordeaux Research in Translational Oncology, BaRITOn, Université de BordeauxBordeaux, France
| | - Alice Buissonnière
- Institut National de la Santé et de la Recherche Médicale, UMR1053 Bordeaux Research in Translational Oncology, BaRITOnBordeaux, France
- UMR1053 Bordeaux Research in Translational Oncology, BaRITOn, Université de BordeauxBordeaux, France
| | - Pierre Dubus
- Institut National de la Santé et de la Recherche Médicale, UMR1053 Bordeaux Research in Translational Oncology, BaRITOnBordeaux, France
- UMR1053 Bordeaux Research in Translational Oncology, BaRITOn, Université de BordeauxBordeaux, France
- CHU Hôpitaux de Bordeaux, Pôle Biologie et Pathologie, Service de Biologie des TumeursBordeaux, France
| | - Francis Mégraud
- Institut National de la Santé et de la Recherche Médicale, UMR1053 Bordeaux Research in Translational Oncology, BaRITOnBordeaux, France
- UMR1053 Bordeaux Research in Translational Oncology, BaRITOn, Université de BordeauxBordeaux, France
| | - Christine Varon
- Institut National de la Santé et de la Recherche Médicale, UMR1053 Bordeaux Research in Translational Oncology, BaRITOnBordeaux, France
- UMR1053 Bordeaux Research in Translational Oncology, BaRITOn, Université de BordeauxBordeaux, France
| | - Armelle Ménard
- Institut National de la Santé et de la Recherche Médicale, UMR1053 Bordeaux Research in Translational Oncology, BaRITOnBordeaux, France
- UMR1053 Bordeaux Research in Translational Oncology, BaRITOn, Université de BordeauxBordeaux, France
| |
Collapse
|