1
|
Tong D, He Y, Haile SA, Lee Z, Le LHM, Emery J, Wray-McCann G, Chonwerawong M, Philpott DJ, Hertzog PJ, Schneider P, Ferrero RL, Ying L. BAFF Blockade Attenuates B Cell MALT Formation in Conditional Nlrc5-Deficient Mice With Helicobacter felis Infection. Eur J Immunol 2025; 55:e202451355. [PMID: 39686777 DOI: 10.1002/eji.202451355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 11/28/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024]
Abstract
Helicobacter infection is a key cause of gastric B cell mucosa-associated lymphoid tissue (MALT) lymphoma. This study examined the role of B cell-activating factor (BAFF), a major driver of B cell proliferation and many B cell disorders, in this malignancy using a model in which conditional knockout mice for NOD-like receptor family CARD domain-containing 5 (Nlrc5) are infected with Helicobacter felis. Gastric BAFF production was significantly increased in H. felis-infected Nlrc5mø-KO mice compared to wild-type. Blocking BAFF signalling, before or after the onset of Helicobacter-induced gastritis, significantly reduced MALT development, with fewer gastric B cell follicles and reduced gland hyperplasia. BAFF blockade also reshaped the immune cell landscape in the stomach, resulting in fewer CD4+ T cells, Tregs, macrophages and dendritic cells. Using a cell culture model, we identified the protein-coding BAFF transcripts that are upregulated in NLRC5-deficient macrophages stimulated with either H. felis or the NLRC5 agonist, lipopolysaccharide. Among the upregulated variants, TNFSF13B (BAFF)-206 acts as a transcription factor and is reported to enhance BAFF production in autoimmune diseases and cancer. Altogether, these findings implicate the NLRC5-BAFF signalling axis in Helicobacter-induced B cell MALT lymphoma, highlighting BAFF inhibition as a potential therapeutic approach.
Collapse
Affiliation(s)
- Dongmei Tong
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Monash University, Melbourne, Victoria, Australia
| | - Yuqi He
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Shambel Araya Haile
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Monash University, Melbourne, Victoria, Australia
| | - Zoe Lee
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Lena H M Le
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Jack Emery
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Georgie Wray-McCann
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Michelle Chonwerawong
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Monash University, Melbourne, Victoria, Australia
| | - Dana J Philpott
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Paul J Hertzog
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Monash University, Melbourne, Victoria, Australia
| | - Pascal Schneider
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Richard L Ferrero
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Monash University, Melbourne, Victoria, Australia
- Biomedicine Discovery Institute, Department of Microbiology, Monash University, Melbourne, Victoria, Australia
| | - Le Ying
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Monash University, Melbourne, Victoria, Australia
- Department of Medicine, Monash University, Melbourne, Victoria, Australia
| |
Collapse
|
2
|
Ying L, Liu P, Ding Z, Wray‐McCann G, Emery J, Colon N, Le LHM, Tran LS, Xu P, Yu L, Philpott DJ, Tu Y, Cheah DMZ, Cheng CL, Lim ST, Ong CK, Ferrero RL. Anti-CD40L therapy prevents the formation of precursor lesions to gastric B-cell MALT lymphoma in a mouse model. J Pathol 2023; 259:402-414. [PMID: 36640261 PMCID: PMC10952994 DOI: 10.1002/path.6053] [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: 08/01/2022] [Revised: 12/21/2022] [Accepted: 01/11/2023] [Indexed: 01/15/2023]
Abstract
Mucosa-associated lymphoid tissue (MALT) lymphoma is a B-cell tumour that develops over many decades in the stomachs of individuals with chronic Helicobacter pylori infection. We developed a new mouse model of human gastric MALT lymphoma in which mice with a myeloid-specific deletion of the innate immune molecule, Nlrc5, develop precursor B-cell lesions to MALT lymphoma at only 3 months post-Helicobacter infection versus 9-24 months in existing models. The gastric B-cell lesions in the Nlrc5 knockout mice had the histopathological features of the human disease, notably lymphoepithelial-like lesions, centrocyte-like cells, and were infiltrated by dendritic cells (DCs), macrophages, and T-cells (CD4+ , CD8+ and Foxp3+ ). Mouse and human gastric tissues contained immune cells expressing immune checkpoint receptor programmed death 1 (PD-1) and its ligand PD-L1, indicating an immunosuppressive tissue microenvironment. We next determined whether CD40L, overexpressed in a range of B-cell malignancies, may be a potential drug target for the treatment of gastric MALT lymphoma. Importantly, we showed that the administration of anti-CD40L antibody either coincident with or after establishment of Helicobacter infection prevented gastric B-cell lesions in mice, when compared with the control antibody treatment. Mice administered the CD40L antibody also had significantly reduced numbers of gastric DCs, CD8+ and Foxp3+ T-cells, as well as decreased gastric expression of B-cell lymphoma genes. These findings validate the potential of CD40L as a therapeutic target in the treatment of human gastric B-cell MALT lymphoma. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Le Ying
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVICAustralia
- Department of Molecular and Translational ScienceMonash UniversityClaytonVICAustralia
| | - Phoebe Liu
- Department of ImmunologyUniversity of TorontoTorontoONCanada
| | - Zhoujie Ding
- Department of Immunology and Pathology, Central Clinical SchoolMonash UniversityClaytonVICAustralia
- Department of Microbiology, Tumor and Cell BiologyKarolinska InstitutetSolnaSweden
| | - Georgie Wray‐McCann
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVICAustralia
| | - Jack Emery
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVICAustralia
| | - Nina Colon
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVICAustralia
| | - Lena HM Le
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVICAustralia
| | - Le Son Tran
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVICAustralia
| | - Ping Xu
- Department of Tea ScienceZhejiang UniversityHangzhouPR China
| | - Liang Yu
- Department of General Surgery, Shanghai General HospitalShanghai Jiao Tong University School of MedicineShanghaiPR China
| | - Dana J Philpott
- Department of ImmunologyUniversity of TorontoTorontoONCanada
| | - Yugang Tu
- Cell Signaling Technology, Inc.DanversMAUSA
| | - Daryl MZ Cheah
- Lymphoma Genomic Translational Research Laboratory, Cellular and Molecular ResearchNational Cancer Centre SingaporeSingaporeSingapore
| | - Chee L Cheng
- Department of PathologySingapore General HospitalSingaporeSingapore
| | - Soon T Lim
- Division of Medical OncologyNational Cancer Centre SingaporeSingaporeSingapore
- SingHealth Duke‐NUS Blood Cancer CentreSingaporeSingapore
- Office of EducationDuke‐NUS Medical SchoolSingaporeSingapore
| | - Choon K Ong
- Lymphoma Genomic Translational Research Laboratory, Cellular and Molecular ResearchNational Cancer Centre SingaporeSingaporeSingapore
- Cancer and Stem Cell Biology ProgramDuke‐NUS Medical SchoolSingaporeSingapore
- Genome Institute of SingaporeSingaporeSingapore
| | - Richard L Ferrero
- Centre for Innate Immunity and Infectious DiseasesHudson Institute of Medical ResearchClaytonVICAustralia
- Department of Molecular and Translational ScienceMonash UniversityClaytonVICAustralia
- Biomedicine Discovery Institute, Department of MicrobiologyMonash UniversityClaytonVICAustralia
| |
Collapse
|
3
|
Soudi H, Falsafi T, Mahboubi M, Gharavi S. Evaluation of Helicobacter pylori OipA protein as a vaccine candidate and propolis as an adjuvant in C57BL/6 mice. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2021; 24:1220-1230. [PMID: 35083009 PMCID: PMC8751746 DOI: 10.22038/ijbms.2021.56232.12579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 07/12/2021] [Indexed: 11/06/2022]
Abstract
OBJECTIVES Outer inflammatory protein A (OipA) is an essential adhesin of Helicobacter pylori. We aimed to evaluate the effects of a recombinant OipA in the induction of crucial cytokines as a vaccine candidate and propolis as an adjuvant in C57BL/6 mice. MATERIALS AND METHODS C57BL/6 mice were divided into nine groups according to the disposition of antigen and adjuvant and route of administration: subcutaneous (sc) or gavage. The administrated recombinant purified OipA and propolis concentrations were 10 μg/ml and 40 μg/ml, respectively. After vaccination, we measured expression levels of IFN-γ and IL-4 cytokine genes in the spleen cells of mice by real-time PCR. RESULTS All results were contrasted with the negative sample. By sc injection, the expression of INF-γ was increased 3.5 and 2.9-fold for OipA and OipA plus propolis, respectively. By gavage 4.4 and 11-fold increase was found for OipA and OipA plus propolis, respectively. The administration of propolis by gavage showed more increase than Sc injection concerning the production of INF-γ. The 11-fold increase for injection of OipA plus propolis by gavage was comparable OipA plus Freund's adjuvant injected subcutaneously. This result suggested an excellent immunological response toward OipA concerning the production of INF-γ in mice. In all cases there were no notable IL-4 production increases. CONCLUSION The results confirm the efficiency of OipA in induction of IFN-γ production, and thereby the cellular immune response. Propolis could be a suitable adjuvant.
Collapse
Affiliation(s)
- Hengameh Soudi
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Tahereh Falsafi
- Microbiology department, Faculty of Biological Sciences, Alzahra University, Tehran, Iran
| | - Mohaddeseh Mahboubi
- Medicinal Plants Research Department, Research and Development, TabibDaru Pharmaceutical Company, Kashan, Iran
| | - Sara Gharavi
- Biotechnology Department, Faculty of Biological Sciences, Alzahra University, Tehran, Iran
| |
Collapse
|
4
|
Chonwerawong M, Ferrand J, Chaudhry HM, Higgins C, Tran LS, Lim SS, Walker MM, Bhathal PS, Dev A, Moore GT, Sievert W, Jenkins BJ, D'Elios MM, Philpott DJ, Kufer TA, Ferrero RL. Innate Immune Molecule NLRC5 Protects Mice From Helicobacter-induced Formation of Gastric Lymphoid Tissue. Gastroenterology 2020; 159:169-182.e8. [PMID: 32169428 DOI: 10.1053/j.gastro.2020.03.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 02/05/2020] [Accepted: 03/03/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Helicobacter pylori induces strong inflammatory responses that are directed at clearing the infection, but if not controlled, these responses can be harmful to the host. We investigated the immune-regulatory effects of the innate immune molecule, nucleotide-binding oligomerization domain-like receptors (NLR) family CARD domain-containing 5 (NLRC5), in patients and mice with Helicobacter infection. METHODS We obtained gastric biopsies from 30 patients in Australia. We performed studies with mice that lack NLRC5 in the myeloid linage (Nlrc5møKO) and mice without Nlrc5 gene disruption (controls). Some mice were gavaged with H pylori SS1 or Helicobacter felis; 3 months later, stomachs, spleens, and sera were collected, along with macrophages derived from bone marrow. Human and mouse gastric tissues and mouse macrophages were analyzed by histology, immunohistochemistry, immunoblots, and quantitative polymerase chain reaction. THP-1 cells (human macrophages, controls) and NLRC5-/- THP-1 cells (generated by CRISPR-Cas9 gene editing) were incubated with Helicobacter and gene expression and production of cytokines were analyzed. RESULTS Levels of NLRC5 messenger RNA were significantly increased in gastric tissues from patients with H pylori infection, compared with patients without infection (P < .01), and correlated with gastritis severity (P < .05). H pylori bacteria induced significantly higher levels of chemokine and cytokine production by NLRC5-/- THP-1 macrophages than by control THP-1 cells (P < .05). After 3 months of infection with H felis, Nlrc5mø-KO mice developed gastric hyperplasia (P < .0001), splenomegaly (P < .0001), and increased serum antibody titers (P < .01), whereas control mice did not. Nlrc5mø-KO mice with chronic H felis infection had increased numbers of gastric B-cell follicles expressing CD19 (P < .0001); these follicles had features of mucosa-associated lymphoid tissue lymphoma. We identified B-cell-activating factor as a protein that promoted B-cell hyperproliferation in Nlrc5mø-KO mice. CONCLUSIONS NLRC5 is a negative regulator of gastric inflammation and mucosal lymphoid formation in response to Helicobacter infection. Aberrant NLRC5 signaling in macrophages can promote B-cell lymphomagenesis during chronic Helicobacter infection.
Collapse
Affiliation(s)
- Michelle Chonwerawong
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Victoria, Australia; Department of Molecular and Translational Science, Monash University, Victoria, Australia
| | - Jonathan Ferrand
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Victoria, Australia
| | - Hassan Mohammad Chaudhry
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Victoria, Australia
| | - Chloe Higgins
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Victoria, Australia
| | - Le Son Tran
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Victoria, Australia
| | - San Sui Lim
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Victoria, Australia
| | - Marjorie M Walker
- School of Medicine and Public Health, Faculty of Health and Medicine, The University of Newcastle, New South Wales, Australia; Department of Pathology, University of Melbourne, Victoria, Australia
| | - Prithi S Bhathal
- Department of Pathology, University of Melbourne, Victoria, Australia
| | - Anouk Dev
- Department of Medicine, Monash University, Monash Medical Centre, Victoria, Australia
| | - Gregory T Moore
- Department of Medicine, Monash University, Monash Medical Centre, Victoria, Australia
| | - William Sievert
- Department of Medicine, Monash University, Monash Medical Centre, Victoria, Australia; Dipartimento di Medicina Sperimentale e Clinica, University of Florence, Florence, Italy
| | - Brendan J Jenkins
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Victoria, Australia; Department of Molecular and Translational Science, Monash University, Victoria, Australia
| | - Mario M D'Elios
- Dipartimento di Medicina Sperimentale e Clinica, University of Florence, Florence, Italy
| | - Dana J Philpott
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Thomas A Kufer
- University of Hohenheim, Institute of Nutritional Medicine, Department of Immunology, Stuttgart, Germany
| | - Richard L Ferrero
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Victoria, Australia; Department of Molecular and Translational Science, Monash University, Victoria, Australia; Biomedicine Discovery Institute, Department of Microbiology, Monash University, Victoria, Australia.
| |
Collapse
|
5
|
Kuo SH, Wu MS, Yeh KH, Lin CW, Hsu PN, Chen LT, Cheng AL. Novel Insights of Lymphomagenesis of Helicobacter pylori-Dependent Gastric Mucosa-Associated Lymphoid Tissue Lymphoma. Cancers (Basel) 2019; 11:547. [PMID: 30999581 PMCID: PMC6520890 DOI: 10.3390/cancers11040547] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 04/09/2019] [Accepted: 04/12/2019] [Indexed: 02/06/2023] Open
Abstract
Gastric mucosa-associated lymphoid tissue (MALT) lymphoma is the most common subtype of gastric lymphoma. Most gastric MALT lymphomas are characterized by their association with the Helicobacter pylori (HP) infection and are cured by first-line HP eradication therapy (HPE). Several studies have been conducted to investigate why most gastric MALT lymphomas remain localized, are dependent on HP infection, and show HP-specific intratumoral T-cells (e.g., CD40-mediated signaling, T-helper-2 (Th2)-type cytokines, chemokines, costimulatory molecules, and FOXP3+ regulatory T-cells) and their communication with B-cells. Furthermore, the reason why the antigen stimuli of these intratumoral T-cells with tonic B-cell receptor signaling promote lymphomagenesis of gastric MALT lymphoma has also been investigated. In addition to the aforementioned mechanisms, it has been demonstrated that the translocated HP cytotoxin-associated gene A (CagA) can promote B-cell proliferation through the activation of Src homology-2 domain-containing phosphatase (SHP-2) phosphorylation-dependent signaling, extracellular-signal-regulated kinase (ERK), p38 mitogen-activated protein kinase (MAPK), B-cell lymphoma (Bcl)-2, and Bcl-xL. Furthermore, the expression of CagA and these CagA-signaling molecules is closely associated with the HP-dependence of gastric MALT lymphomas (completely respond to first-line HPE). In this article, we summarize evidence of the classical theory of HP-reactive T-cells and the new paradigm of direct interaction between HP and B-cells that contributes to the HP-dependent lymphomagenesis of gastric MALT lymphomas. Although the role of first-line HPE in the treatment of HP-negative gastric MALT lymphoma remains uncertain, several case series suggest that a proportion of HP-negative gastric MALT lymphomas remains antibiotic-responsive and is cured by HPE. Considering the complicated interaction between microbiomes and the genome/epigenome, further studies on the precise mechanisms of HP- and other bacteria-directed lymphomagenesis in antibiotic-responsive gastric MALT lymphomas are warranted.
Collapse
Affiliation(s)
- Sung-Hsin Kuo
- Department of Oncology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei 100, Taiwan.
- Cancer Research Center, National Taiwan University College of Medicine, Taipei 100, Taiwan.
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei 100, Taiwan.
- National Taiwan University Cancer Center, National Taiwan University College of Medicine, Taipei 106, Taiwan.
| | - Ming-Shiang Wu
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei 100, Taiwan.
| | - Kun-Huei Yeh
- Department of Oncology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei 100, Taiwan.
- Cancer Research Center, National Taiwan University College of Medicine, Taipei 100, Taiwan.
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei 100, Taiwan.
- National Taiwan University Cancer Center, National Taiwan University College of Medicine, Taipei 106, Taiwan.
| | - Chung-Wu Lin
- Department of Pathology, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei 100, Taiwan.
| | - Ping-Ning Hsu
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei 100, Taiwan.
| | - Li-Tzong Chen
- National Institute of Cancer Research, National Health Research Institutes, Tainan 704, Taiwan.
- Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
- Department of Internal Medicine, National Cheng-Kung University Hospital, Tainan 704, Taiwan.
| | - Ann-Lii Cheng
- Cancer Research Center, National Taiwan University College of Medicine, Taipei 100, Taiwan.
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei 100, Taiwan.
- National Taiwan University Cancer Center, National Taiwan University College of Medicine, Taipei 106, Taiwan.
- Department of Internal Medicine, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei 100, Taiwan.
| |
Collapse
|
6
|
Abstract
Helicobacter pylori is usually acquired in early childhood and the infection persists lifelong without causing symptoms. In a small of cases, the infection leads to gastric or duodenal ulcer disease, or gastric cancer. Why disease occurs in these individuals remains unclear, however the host response is known to play a very important part. Understanding the mechanisms involved in maintaining control over the immune and inflammatory response is therefore extremely important. Vaccines against H. pylori have remained elusive but are desperately needed for the prevention of gastric carcinogenesis. This review focuses on research findings which may prove useful in the development of prognostic tests for gastric cancer development, therapeutic agents to control immunopathology, and effective vaccines.
Collapse
Affiliation(s)
- Karen Robinson
- Nottingham Digestive Diseases Biomedical Research Centre, School of Medicine, University of Nottingham, Nottingham, UK
| | - Kazuyo Kaneko
- Nottingham Digestive Diseases Biomedical Research Centre, School of Medicine, University of Nottingham, Nottingham, UK
| | - Leif Percival Andersen
- Department of Clinical Microbiology, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark
| |
Collapse
|
7
|
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
|