1
|
Bali P, Lozano-Pope I, Hernandez J, Estrada MV, Benner C, Obonyo M. Protocol to establish an accelerated murine model for Helicobacter-induced gastric cancer. STAR Protoc 2024; 5:103302. [PMID: 39264805 PMCID: PMC11416640 DOI: 10.1016/j.xpro.2024.103302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 07/22/2024] [Accepted: 08/16/2024] [Indexed: 09/14/2024] Open
Abstract
Helicobacter-induced gastric cancer progresses very slowly, even in animal models, making it difficult to study. Here, we present a protocol to establish an accelerated murine model for Helicobacter-induced gastric cancer. We describe steps for infecting mice with Helicobacter felis, harvesting gastric tissue, assessing disease severity by histopathologic scoring, and performing gene expression studies with RT-qPCR and RNA sequencing. The accelerated model shows rapid progression of the disease, with gastric precancerous lesions developing within 6 months post-infection with Helicobacter. For complete details on the use and execution of this protocol, please refer to Bali et al.1.
Collapse
Affiliation(s)
- Prerna Bali
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Ivonne Lozano-Pope
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Jonathan Hernandez
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Monica V Estrada
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA
| | - Christopher Benner
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Marygorret Obonyo
- Department of Medicine, School of Medicine, University of California, San Diego, La Jolla, CA, USA; Moores Cancer Center, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
2
|
Liu Z, Li H, Huang X, Liu Q. Animal Models of Helicobacter pylori Infection and Vaccines: Current Status and Future Prospects. Helicobacter 2024; 29:e13119. [PMID: 39108210 DOI: 10.1111/hel.13119] [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: 04/27/2024] [Revised: 07/10/2024] [Accepted: 07/23/2024] [Indexed: 01/02/2025]
Abstract
Helicobacter pylori infection causes chronic gastritis, ulcers, and gastric cancer, making it a threat to human health. Despite the use of antibiotic therapy, the global prevalence of H. pylori infection remains high, necessitating early eradication measures. Immunotherapy, especially vaccine development, is a promising solution in this direction, albeit the selection of an appropriate animal model is critical in efficient vaccine production. Accordingly, we conducted a literature, search and summarized the commonly used H. pylori strains, H. pylori infection-related animal models, and models for evaluating H. pylori vaccines. Based on factors such as the ability to replicate human diseases, strain compatibility, vaccine types, and eliciting of immune responses, we systematically compared the advantages and disadvantages of different animal models, to obtain the informed recommendations. In addition, we have proposed novel perspectives on H. pylori-related animal models to advance research and vaccine evaluation for the prevention and treatment of diseases such as gastric cancer.
Collapse
Affiliation(s)
- Zhili Liu
- Department of Medical Microbiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
- HuanKui Academy, Nanchang University, Nanchang, China
| | - He Li
- Department of Medical Microbiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xiaotian Huang
- Department of Medical Microbiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Qiong Liu
- Department of Medical Microbiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
3
|
Nguyen TKC, Do HDK, Nguyen TLP, Pham TT, Mach BN, Nguyen TC, Pham TL, Katsande PM, Hong HA, Duong HT, Phan AN, Cutting SM, Vu MT, Nguyen VD. Genomic and vaccine preclinical studies reveal a novel mouse-adapted Helicobacter pylori model for the hpEastAsia genotype in Southeast Asia. J Med Microbiol 2024; 73. [PMID: 38235783 DOI: 10.1099/jmm.0.001786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2024] Open
Abstract
Introduction. Helicobacter pylori infection is a major global health concern, linked to the development of various gastrointestinal diseases, including gastric cancer. To study the pathogenesis of H. pylori and develop effective intervention strategies, appropriate animal pathogen models that closely mimic human infection are essential.Gap statement. This study focuses on the understudied hpEastAsia genotype in Southeast Asia, a region marked by a high H. pylori infection rate. No mouse-adapted model strains has been reported previously. Moreover, it recognizes the urgent requirement for vaccines in developing countries, where overuse of antimicrobials is fuelling the emergence of resistance.Aim. This study aims to establish a novel mouse-adapted H. pylori model specific to the hpEastAsia genotype prevalent in Southeast Asia, focusing on comparative genomic and histopathological analysis of pathogens coupled with vaccine preclinical studies.Methodology. We collected and sequenced the whole genome of clinical strains of H. pylori from infected patients in Vietnam and performed comparative genomic analyses of H. pylori strains in Southeast Asia. In parallel, we conducted preclinical studies to assess the pathogenicity of the mouse-adapted H. pylori strain and the protective effect of a new spore-vectored vaccine candidate on male Mlac:ICR mice and the host immune response in a female C57BL/6 mouse model.Results. Genome sequencing and comparison revealed unique and common genetic signatures, antimicrobial resistance genes and virulence factors in strains HP22 and HP34; and supported clarithromycin-resistant HP34 as a representation of the hpEastAsia genotype in Vietnam and Southeast Asia. HP34-infected mice exhibited gastric inflammation, epithelial erosion and dysplastic changes that closely resembled the pathology observed in human H. pylori infection. Furthermore, comprehensive immunological characterization demonstrated a robust host immune response, including both mucosal and systemic immune responses. Oral vaccination with candidate vaccine formulations elicited a significant reduction in bacterial colonization in the model.Conclusion. Our findings demonstrate the successful development of a novel mouse-adapted H. pylori model for the hpEastAsia genotype in Vietnam and Southeast Asia. Our research highlights the distinctive genotype and pathogenicity of clinical H. pylori strains in the region, laying the foundation for targeted interventions to address this global health burden.
Collapse
Affiliation(s)
- Thi Kim Cuc Nguyen
- Institute of Biotechnology and Environment, Nha Trang University, 2 Nguyen Dinh Chieu Street, Khanh Hoa, Vietnam
| | - Hoang Dang Khoa Do
- NTT Hi-tech Institute, Nguyen Tat Thanh University, 300A Nguyen Tat Thanh, Ho Chi Minh City, Vietnam
| | - Thi Lan Phuong Nguyen
- Institute of Vaccines and Biological Medicals (IVAC), 9 Pasteur Street, Nha Trang, Khanh Hoa, Vietnam
| | - Thu Thuy Pham
- Institute of Biotechnology and Environment, Nha Trang University, 2 Nguyen Dinh Chieu Street, Khanh Hoa, Vietnam
| | - Bao Ngoc Mach
- NTT Hi-tech Institute, Nguyen Tat Thanh University, 300A Nguyen Tat Thanh, Ho Chi Minh City, Vietnam
| | - Thi Chinh Nguyen
- Institute of Biotechnology and Environment, Nha Trang University, 2 Nguyen Dinh Chieu Street, Khanh Hoa, Vietnam
| | - Thi Lan Pham
- Institute of Biotechnology and Environment, Nha Trang University, 2 Nguyen Dinh Chieu Street, Khanh Hoa, Vietnam
| | - Paidamoyo M Katsande
- Department of Biological Sciences, Royal Holloway University of London, Egham, Surrey, TW20 0EX, UK
| | - Huynh Anh Hong
- Department of Biological Sciences, Royal Holloway University of London, Egham, Surrey, TW20 0EX, UK
| | - Huu Thai Duong
- Institute of Vaccines and Biological Medicals (IVAC), 9 Pasteur Street, Nha Trang, Khanh Hoa, Vietnam
| | - Anh N Phan
- School of Engineering, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK
| | - Simon M Cutting
- Department of Biological Sciences, Royal Holloway University of London, Egham, Surrey, TW20 0EX, UK
| | - Minh Thiet Vu
- NTT Hi-tech Institute, Nguyen Tat Thanh University, 300A Nguyen Tat Thanh, Ho Chi Minh City, Vietnam
| | - Van Duy Nguyen
- Institute of Biotechnology and Environment, Nha Trang University, 2 Nguyen Dinh Chieu Street, Khanh Hoa, Vietnam
- School of Engineering, Newcastle University, Newcastle upon Tyne, NE1 7RU, UK
| |
Collapse
|
4
|
Beduleva L, Fomina K, Sidorov A, Terentiev A, Ivanov P, Menshikov I. Rat Experimental Autoimmune Gastritis Model. Immunol Invest 2023; 52:1023-1038. [PMID: 37962068 DOI: 10.1080/08820139.2023.2283103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
BACKGROUND Autoimmune gastritis (AIG) is an autoimmune disease of the stomach characterized by the destruction of the oxyntic mucosa, which stops producing acid and becomes both functionally and morphologically atrophic. The pathogenic mechanisms behind the disease are still poorly understood. There is no early diagnosis and specific AIG therapy. To elucidate the pathogenesis of AIG, to search for early diagnostic markers, as well as to test new therapeutic approaches, an adequate and easily reproducible experimental model for autoimmune gastritis (EAG) is needed. Existing EAG models have some limitations, including slow development of signs, absence of advanced gastritis, irrational use of animals to obtain antigen. The aim was to find out whether it is possible to cause autoimmune gastritis similar to human disease in Wistar rats through immunization with a homologous gastric mucosa extract. METHODS Wistar rats were immunized with gastric mucosa extract. Histology studies and evaluation of serological parameters were performed 56 and 91 days later. RESULTS Destruction of oxyntic glands by infiltrating T lymphocytes were detected in rats on 56 and 91 days after initial immunization with gastric mucosa extract. Hyperplasia of enterochromaffin-like (ECL) cells was detected on the 91st day. Antral mucosa remained unchanged. CONCLUSION Wistar rats, immunized with gastric mucosa extract, developed EAG similar to human AIG. The advantages of received EAG model are the ease of obtaining, the rapid development of oxyntic mucosa damage, which may progress to ECL cell hyperplasia.
Collapse
Affiliation(s)
- Liubov Beduleva
- Laboratory of Molecular and Cell Immunology, Department of Immunology and Cell Biology, Udmurt State University, Izhevsk, Russian Federation
- Laboratory of Biocompatible Materials, Udmurt Federal Research Center UB RAS, Izhevsk, Russian Federation
| | - Kseniya Fomina
- Laboratory of Molecular and Cell Immunology, Department of Immunology and Cell Biology, Udmurt State University, Izhevsk, Russian Federation
- Laboratory of Biocompatible Materials, Udmurt Federal Research Center UB RAS, Izhevsk, Russian Federation
| | - Alexandr Sidorov
- Laboratory of Molecular and Cell Immunology, Department of Immunology and Cell Biology, Udmurt State University, Izhevsk, Russian Federation
- Laboratory of Biocompatible Materials, Udmurt Federal Research Center UB RAS, Izhevsk, Russian Federation
| | - Alexey Terentiev
- Laboratory of Molecular and Cell Immunology, Department of Immunology and Cell Biology, Udmurt State University, Izhevsk, Russian Federation
- Laboratory of Biocompatible Materials, Udmurt Federal Research Center UB RAS, Izhevsk, Russian Federation
| | - Pavel Ivanov
- Laboratory of Molecular and Cell Immunology, Department of Immunology and Cell Biology, Udmurt State University, Izhevsk, Russian Federation
| | - Igor Menshikov
- Laboratory of Molecular and Cell Immunology, Department of Immunology and Cell Biology, Udmurt State University, Izhevsk, Russian Federation
- Laboratory of Biocompatible Materials, Udmurt Federal Research Center UB RAS, Izhevsk, Russian Federation
| |
Collapse
|
5
|
Ansari S, Yamaoka Y. Animal Models and Helicobacter pylori Infection. J Clin Med 2022; 11:jcm11113141. [PMID: 35683528 PMCID: PMC9181647 DOI: 10.3390/jcm11113141] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 05/25/2022] [Accepted: 05/25/2022] [Indexed: 02/06/2023] Open
Abstract
Helicobacter pylori colonize the gastric mucosa of at least half of the world’s population. Persistent infection is associated with the development of gastritis, peptic ulcer disease, and an increased risk of gastric cancer and gastric-mucosa-associated lymphoid tissue (MALT) lymphoma. In vivo studies using several animal models have provided crucial evidence for understanding the pathophysiology of H. pylori-associated complications. Numerous animal models, such as Mongolian gerbils, transgenic mouse models, guinea pigs, and other animals, including non-human primates, are being widely used due to their persistent association in causing gastric complications. However, finding suitable animal models for in vivo experimentation to understand the pathophysiology of gastric cancer and MALT lymphoma is a complicated task. In this review, we summarized the most appropriate and latest information in the scientific literature to understand the role and importance of H. pylori infection animal models.
Collapse
Affiliation(s)
- Shamshul Ansari
- Department of Environmental and Preventive Medicine, Oita University Faculty of Medicine, Yufu 879-5593, Oita, Japan;
| | - Yoshio Yamaoka
- Department of Environmental and Preventive Medicine, Oita University Faculty of Medicine, Yufu 879-5593, Oita, Japan;
- Department of Medicine, Gastroenterology and Hepatology Section, Baylor College of Medicine, Houston, TX 77030, USA
- Institute of Tropical Disease, Universitas Airlangga, Surabaya 60115, Indonesia
- Correspondence: ; Tel.: +81-97-586-5740
| |
Collapse
|
6
|
Dey TK, Karmakar BC, Sarkar A, Paul S, Mukhopadhyay AK. A Mouse Model of Helicobacter pylori Infection. Methods Mol Biol 2021; 2283:131-151. [PMID: 33765316 DOI: 10.1007/978-1-0716-1302-3_14] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Infection with Helicobacter pylori (H. pylori) is of great distress because of its vital role in the pathogenesis of chronic gastritis, peptic ulcers, and in the multi-step carcinogenic process of gastric cancer. The increasing antibiotic resistance pattern of H. pylori worldwide has prompted the World Health Organization to put this organism in the priority pathogens list. To study the disease biology, evaluation of drugs, treatment outcome and to come up with probable vaccination strategies, competent animal models that reproduce the signature of human infection are essential. Initial reports about animal colonization with H. pylori have shown significant heterogeneity, to such an extent that Barry Marshall, Nobel laureate for the discovery of H. pylori , infected himself with the bacterium to show its involvement in acute gastric illness. A paradigm-shift discovery of the H. pylori mouse-adapted strain SS1 has opened the avenues of research regarding the organism and its pathogenicity. Although the mouse model of H. pylori infection is being utilized all over the world, there are certain issues that need awareness and specific information to achieve successful, consistent colonization with symptoms resembling human. This chapter details an established and reliable protocol for the development of a competent mouse model for H. pylori infection leading to various gastro-intestinal diseases.
Collapse
Affiliation(s)
- Tanmoy Kumar Dey
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Bipul Chandra Karmakar
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Avijit Sarkar
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Sangita Paul
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India
| | - Asish Kumar Mukhopadhyay
- Division of Bacteriology, ICMR-National Institute of Cholera and Enteric Diseases, Kolkata, India.
| |
Collapse
|
7
|
Cytolethal distending toxin induces the formation of transient messenger-rich ribonucleoprotein nuclear invaginations in surviving cells. PLoS Pathog 2019; 15:e1007921. [PMID: 31568537 PMCID: PMC6824578 DOI: 10.1371/journal.ppat.1007921] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 11/01/2019] [Accepted: 06/18/2019] [Indexed: 01/26/2023] Open
Abstract
Humans are frequently exposed to bacterial genotoxins involved in digestive cancers, colibactin and Cytolethal Distending Toxin (CDT), the latter being secreted by many pathogenic bacteria. Our aim was to evaluate the effects induced by these genotoxins on nuclear remodeling in the context of cell survival. Helicobacter infected mice, coculture experiments with CDT- and colibactin-secreting bacteria and hepatic, intestinal and gastric cells, and xenograft mouse-derived models were used to assess the nuclear remodeling in vitro and in vivo. Our results showed that CDT and colibactin induced-nuclear remodeling can be associated with the formation of deep cytoplasmic invaginations in the nucleus of giant cells. These structures, observed both in vivo and in vitro, correspond to nucleoplasmic reticulum (NR). The core of the NR was found to concentrate ribosomes, proteins involved in mRNA translation, polyadenylated RNA and the main components of the complex mCRD involved in mRNA turnover. These structures are active sites of mRNA translation, correlated with a high degree of ploidy, and involve MAPK and calcium signaling. Additional data showed that insulation and concentration of these adaptive ribonucleoprotein particles within the nucleus are dynamic, transient and protect the cell until the genotoxic stress is relieved. Bacterial genotoxins-induced NR would be a privileged gateway for selected mRNA to be preferably transported therein for local translation. These findings offer new insights into the context of NR formation, a common feature of many cancers, which not only appears in response to therapies-induced DNA damage but also earlier in response to genotoxic bacteria. Humans are frequently exposed to bacterial genotoxins linked to cancers, colibactin and Cytolethal Distending Toxin (CDT). These genotoxins induce DNA damage and can promote formation of nucleoplasmic reticulum (NR), deeply invaginated in the nucleoplasm of giant nuclei both in vivo and in vitro. Our cellular models showed that these structures can be observed together with profound nuclear reorganization corresponding to remodeling of nuclear material. The core of the genotoxin-induced NRs concentrates protein production machinery of the cell as well as controlling elements of protein turnover. These genotoxin-induced dynamic structures may also be signaling hubs controlling mRNA turnover and translation of selected mRNAs and thus correspond to a privileged gateway for the synthesis of selected mRNA which are preferentially transported from the nucleus through pores and translated therein. These transient and reversible hubs allow the cell to pause and repair the DNA damage caused by bacterial genotoxins in order to maintain cell survival. As NR formation is a common feature of many cancers, similar mechanism could occur and contribute to the resistance of cancer cells to radiotherapies and some chemotherapies aimed at inducing DNA damage.
Collapse
|
8
|
CagL from Helicobacter pylori has ADP-ribosylation activity and exerts partial protective efficacy in mice. Arch Biochem Biophys 2017; 635:102-109. [DOI: 10.1016/j.abb.2017.10.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 10/20/2017] [Accepted: 10/26/2017] [Indexed: 12/26/2022]
|
9
|
Lozano-Pope I, Sharma A, Matthias M, Doran KS, Obonyo M. Effect of myeloid differentiation primary response gene 88 on expression profiles of genes during the development and progression of Helicobacter-induced gastric cancer. BMC Cancer 2017; 17:133. [PMID: 28201999 PMCID: PMC5310019 DOI: 10.1186/s12885-017-3114-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 02/04/2017] [Indexed: 12/15/2022] Open
Abstract
Background Gastric cancer is one of the most common and lethal type of cancer worldwide. Infection with Helicobacter pylori (H. pylori) is recognized as the major cause of gastric cancer. However, it remains unclear the mechanism by which Helicobacter infection leads to gastric cancer. Furthermore, the underlying molecular events involved during the progression of Helicobacter infection to gastric malignancy are not well understood. In previous studies, we demonstrated that that H. felis-infected Myd88−/− mice exhibited dramatic pathology and an accelerated progression to gastric dysplasia; however, the MyD88 downstream gene targets responsible for this pathology have not been described. This study was designed to identify MyD88-dependent genes involved in the progression towards gastric cancer during the course of Helicobacter infection. Methods Wild type (WT) and Myd88 deficient mice (Myd88−/−) were infected with H. felis for 25 and 47 weeks and global transcriptome analysis performed on gastric tissue using MouseWG-6 v2 expression BeadChips microarrays. Function and pathway enrichment analyses of statistically significant, differential expressed genes (p < 0.05) were performed using the Database for Annotation, Visualization and Integrated Discovery (DAVID) online tools. Results Helicobacter infection affected the transcriptional profile of more genes in Myd88−/− mice compared to WT mice. Infection of Myd88−/− mice resulted in the differential expression of 1,989 genes at 25 weeks (1031 up and 958 downregulated). At 47 weeks post-H.felis infection, 2,162 (1140 up and 1022 downregulated) were differentially expressed. The most significant differentially upregulated gene during Helicobacter infection in Myd88−/− mice was chitinase-like 4 (chil4), which is involved in tissue remodeling and wound healing. Other highly upregulated genes in H. felis-infected Myd88−/− mice included, Indoleamine 2,3-Dioxygenase 1 (Ido1), Guanylate binding protein 2 (Gbp2), ubiquitin D (Ubd), β2-Microglobulin (B2m), CD74 antigen (Cd74), which have been reported to promote cancer progression by enhancing angiogenesis, proliferation, migration, metastasis, invasion, and tumorigenecity. For downregulated genes, the highly expressed genes included, ATPase H+/K+ transporting, alpha subunit (Atp4a), Atp4b, Mucin 5 AC (Muc5ac), Apolipoprotein A-1 (Apoa1), and gastric intrinsic factor (Gif), whose optimal function is important in maintaining gastric hemostasis and lower expression has been associated with increased risk of gastric carcinogenesis. Conclusions These results provide a global transcriptional gene profile during the development and progression of Helicobacter-induced gastric cancer. The data show that our mouse model system is useful for identifying genes involved in gastric cancer progression. Electronic supplementary material The online version of this article (doi:10.1186/s12885-017-3114-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Arnika Sharma
- Department of Biology, San Diego State University, San Diego, CA, USA
| | - Michael Matthias
- Department of Medicine, University of California, La Jolla, CA, USA
| | - Kelly S Doran
- Department of Biology, San Diego State University, San Diego, CA, USA
| | - Marygorret Obonyo
- Department of Medicine, University of California, La Jolla, CA, USA.
| |
Collapse
|
10
|
Chonwerawong M, Avé P, Huerre M, Ferrero RL. Interferon-γ promotes gastric lymphoid follicle formation but not gastritis in Helicobacter-infected BALB/c mice. Gut Pathog 2016; 8:61. [PMID: 27895717 PMCID: PMC5117576 DOI: 10.1186/s13099-016-0142-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 11/14/2016] [Indexed: 12/13/2022] Open
Abstract
Background Mouse infection studies have shown that interferon-γ (IFN-γ), a T helper 1 (Th1) cytokine, is required for the development of severe pathology induced by chronic Helicobacter infection. This finding is largely based on studies performed using mice that have polarised Th1 responses i.e. C57BL/6 animals. The current work aims to investigate the role of IFN-γ in Helicobacter-induced inflammation in BALB/c mice which have Th2-polarised immune responses. Results At 7 months post-infection with Helicobacter felis, IFN-γ deficiency in BALB/c mice had no significant effect on H. felis colonisation levels in the gastric mucosa, nor on humoral responses, or gastritis severity. Ifng−/− animals with chronic H. felis infection did, however, develop significantly fewer lymphoid follicle lesions, as well as increased IL-4 splenocyte responses, when compared with infected Ifng+/+ mice (P = 0.015 and P = 0.0004, respectively). Conclusions The work shows that in mice on a BALB/c background, IFN-γ is not required for bacterial clearance, antibody responses, nor gastric inflammation. Conversely, IFN-γ appears to play a role in the development of gastric lymphoid follicles, which are precursor lesions to mucosa-associated lymphoid tissue (MALT) lymphoma. This study highlights the importance of mouse host background on the susceptibility to Helicobacter-induced pathologies.
Collapse
Affiliation(s)
- Michelle Chonwerawong
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Monash University, 27-31 Wright Street, Clayton, VIC 3123 Australia
| | - Patrick Avé
- Unité de Recherche et d'Expertise Histotechnologie et Pathologie, Institut Pasteur, 25-28 Rue du Dr Roux, 75724 Paris, France ; Unité de Histopathologie Humaine et Modèles Animaux, Institut Pasteur, 25-28 Rue du Dr Roux, 75724 Paris, France
| | - Michel Huerre
- Unité de Recherche et d'Expertise Histotechnologie et Pathologie, Institut Pasteur, 25-28 Rue du Dr Roux, 75724 Paris, France ; Département de Pathologie, Institut Curie, 26 Rue d'Ulm, 75248 Paris, France
| | - Richard L Ferrero
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Monash University, 27-31 Wright Street, Clayton, VIC 3123 Australia ; Department of Microbiology, Monash University, Clayton, VIC Australia
| |
Collapse
|
11
|
Banerjee A, Basu M, Blanchard TG, Chintalacharuvu SR, Guang W, Lillehoj EP, Czinn SJ. Early Molecular Events in Murine Gastric Epithelial Cells Mediated by Helicobacter pylori CagA. Helicobacter 2016; 21:395-404. [PMID: 26817586 PMCID: PMC4965344 DOI: 10.1111/hel.12300] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Murine models of Helicobacter pylori infection are used to study host-pathogen interactions, but lack of severe gastritis in this model has limited its usefulness in studying pathogenesis. We compared the murine gastric epithelial cell line GSM06 to the human gastric epithelial AGS cell line to determine whether similar events occur when cultured with H. pylori. MATERIALS AND METHODS The lysates of cells infected with H. pylori isolates or an isogenic cagA-deficient mutant were assessed for translocation and phosphorylation of CagA and for activation of stress pathway kinases by immunoblot. RESULTS Phosphorylated CagA was detected in both cell lines within 60 minutes. Phospho-ERK 1/2 was present within several minutes and distinctly present in GSM06 cells at 60 minutes. Similar results were obtained for phospho-JNK, although the 54 kDa phosphoprotein signal was dominant in AGS, whereas the lower molecular weight band was dominant in GSM06 cells. CONCLUSION These results demonstrate that early events in H. pylori pathogenesis occur within mouse epithelial cells similar to human cells and therefore support the use of the mouse model for the study of acute CagA-associated host cell responses. These results also indicate that reduced disease in H. pylori-infected mice may be due to lack of the Cag PAI, or by differences in the mouse response downstream of the initial activation events.
Collapse
Affiliation(s)
- Aditi Banerjee
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Malini Basu
- Department of Microbiology, Rastraguru Surendra Nath College, Barrackpore, India
| | | | | | - Wei Guang
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Erik P Lillehoj
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Steven J Czinn
- University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
12
|
Philipson CW, Bassaganya-Riera J, Viladomiu M, Kronsteiner B, Abedi V, Hoops S, Michalak P, Kang L, Girardin SE, Hontecillas R. Modeling the Regulatory Mechanisms by Which NLRX1 Modulates Innate Immune Responses to Helicobacter pylori Infection. PLoS One 2015; 10:e0137839. [PMID: 26367386 PMCID: PMC4569576 DOI: 10.1371/journal.pone.0137839] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 08/22/2015] [Indexed: 12/15/2022] Open
Abstract
Helicobacter pylori colonizes half of the world’s population as the dominant member of the gastric microbiota resulting in a lifelong chronic infection. Host responses toward the bacterium can result in asymptomatic, pathogenic or even favorable health outcomes; however, mechanisms underlying the dual role of H. pylori as a commensal versus pathogenic organism are not well characterized. Recent evidence suggests mononuclear phagocytes are largely involved in shaping dominant immunity during infection mediating the balance between host tolerance and succumbing to overt disease. We combined computational modeling, bioinformatics and experimental validation in order to investigate interactions between macrophages and intracellular H. pylori. Global transcriptomic analysis on bone marrow-derived macrophages (BMDM) in a gentamycin protection assay at six time points unveiled the presence of three sequential host response waves: an early transient regulatory gene module followed by sustained and late effector responses. Kinetic behaviors of pattern recognition receptors (PRRs) are linked to differential expression of spatiotemporal response waves and function to induce effector immunity through extracellular and intracellular detection of H. pylori. We report that bacterial interaction with the host intracellular environment caused significant suppression of regulatory NLRC3 and NLRX1 in a pattern inverse to early regulatory responses. To further delineate complex immune responses and pathway crosstalk between effector and regulatory PRRs, we built a computational model calibrated using time-series RNAseq data. Our validated computational hypotheses are that: 1) NLRX1 expression regulates bacterial burden in macrophages; and 2) early host response cytokines down-regulate NLRX1 expression through a negative feedback circuit. This paper applies modeling approaches to characterize the regulatory role of NLRX1 in mechanisms of host tolerance employed by macrophages to respond to and/or to co-exist with intracellular H. pylori.
Collapse
Affiliation(s)
- Casandra W. Philipson
- Center for Modeling Immunity to Enteric Pathogens, Virginia Bioinformatics Institute at Virginia Tech, Blacksburg, VA, United States of America
- Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute at Virginia Tech, Blacksburg, VA, United States of America
| | - Josep Bassaganya-Riera
- Center for Modeling Immunity to Enteric Pathogens, Virginia Bioinformatics Institute at Virginia Tech, Blacksburg, VA, United States of America
- Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute at Virginia Tech, Blacksburg, VA, United States of America
- Virginia-Maryland Regional College of Veterinary Medicine, Blacksburg, VA, United States of America
| | - Monica Viladomiu
- Center for Modeling Immunity to Enteric Pathogens, Virginia Bioinformatics Institute at Virginia Tech, Blacksburg, VA, United States of America
- Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute at Virginia Tech, Blacksburg, VA, United States of America
| | - Barbara Kronsteiner
- Center for Modeling Immunity to Enteric Pathogens, Virginia Bioinformatics Institute at Virginia Tech, Blacksburg, VA, United States of America
- Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute at Virginia Tech, Blacksburg, VA, United States of America
| | - Vida Abedi
- Center for Modeling Immunity to Enteric Pathogens, Virginia Bioinformatics Institute at Virginia Tech, Blacksburg, VA, United States of America
- Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute at Virginia Tech, Blacksburg, VA, United States of America
| | - Stefan Hoops
- Center for Modeling Immunity to Enteric Pathogens, Virginia Bioinformatics Institute at Virginia Tech, Blacksburg, VA, United States of America
- Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute at Virginia Tech, Blacksburg, VA, United States of America
| | - Pawel Michalak
- Center for Modeling Immunity to Enteric Pathogens, Virginia Bioinformatics Institute at Virginia Tech, Blacksburg, VA, United States of America
| | - Lin Kang
- Center for Modeling Immunity to Enteric Pathogens, Virginia Bioinformatics Institute at Virginia Tech, Blacksburg, VA, United States of America
| | - Stephen E. Girardin
- Laboratory of Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Raquel Hontecillas
- Center for Modeling Immunity to Enteric Pathogens, Virginia Bioinformatics Institute at Virginia Tech, Blacksburg, VA, United States of America
- Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute at Virginia Tech, Blacksburg, VA, United States of America
- * E-mail:
| |
Collapse
|
13
|
Semper RP, Mejías-Luque R, Groß C, Anderl F, Müller A, Vieth M, Busch DH, Prazeres da Costa C, Ruland J, Groß O, Gerhard M. Helicobacter pylori-induced IL-1β secretion in innate immune cells is regulated by the NLRP3 inflammasome and requires the cag pathogenicity island. THE JOURNAL OF IMMUNOLOGY 2014; 193:3566-76. [PMID: 25172489 DOI: 10.4049/jimmunol.1400362] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Infection with the gram-negative bacterium Helicobacter pylori is the most prevalent chronic bacterial infection, affecting ∼50% of the world's population, and is the main risk factor of gastric cancer. The proinflammatory cytokine IL-1β plays a crucial role in the development of gastric tumors and polymorphisms in the IL-1 gene cluster leading to increased IL-1β production have been associated with increased risk for gastric cancer. To be active, pro-IL-1β must be cleaved by the inflammasome, an intracellular multiprotein complex implicated in physiological and pathological inflammation. Recently, H. pylori was postulated to activate the inflammasome in murine bone marrow-derived dendritic cells; however, the molecular mechanisms as well as the bacterial virulence factor acting as signal 2 activating the inflammasome remain elusive. In this study, we analyzed the inflammasome complex regulating IL-1β upon H. pylori infection as well as the molecular mechanisms involved. Our results indicate that H. pylori-induced IL-1β secretion is mediated by activation of the nucleotide-binding oligomerization domain-like receptor family, pyrin domain-containing 3 inflammasome. We also show that reactive oxygen species, potassium efflux, and lysosomal destabilization are the main cellular mechanisms responsible of nucleotide-binding oligomerization domain family, pyrin domain-containing 3 inflammasome activation upon H. pylori infection, and identify vacuolating cytotoxin A and cag pathogenicity island as the bacterial virulence determinants involved. Moreover, in vivo experiments indicate an important role for the inflammasome in the onset and establishment of H. pylori infection and in the subsequent inflammatory response of the host.
Collapse
Affiliation(s)
- Raphaela P Semper
- Institut für Medizinische Mikrobiologie, Immunologie und Hygiene, Technische Universität München, 81675 Munich, Germany
| | - Raquel Mejías-Luque
- Institut für Medizinische Mikrobiologie, Immunologie und Hygiene, Technische Universität München, 81675 Munich, Germany
| | - Christina Groß
- Institut für Klinische Chemie und Pathobiochemie, Klinikum Rechts der Isar, Technische Universität München, 81675 Munich, Germany
| | - Florian Anderl
- Institut für Medizinische Mikrobiologie, Immunologie und Hygiene, Technische Universität München, 81675 Munich, Germany
| | - Anne Müller
- Institute of Molecular Cancer Research, University of Zürich, 8057 Zürich, Switzerland
| | - Michael Vieth
- Institut für Pathologie, Klinikum Bayreuth, 95445 Bayreuth, Germany; and
| | - Dirk H Busch
- Institut für Medizinische Mikrobiologie, Immunologie und Hygiene, Technische Universität München, 81675 Munich, Germany; German Centre for Infection Research, Partner Site Munich, 81675 Munich, Germany
| | - Clarissa Prazeres da Costa
- Institut für Medizinische Mikrobiologie, Immunologie und Hygiene, Technische Universität München, 81675 Munich, Germany
| | - Jürgen Ruland
- Institut für Klinische Chemie und Pathobiochemie, Klinikum Rechts der Isar, Technische Universität München, 81675 Munich, Germany; German Centre for Infection Research, Partner Site Munich, 81675 Munich, Germany
| | - Olaf Groß
- Institut für Klinische Chemie und Pathobiochemie, Klinikum Rechts der Isar, Technische Universität München, 81675 Munich, Germany
| | - Markus Gerhard
- Institut für Medizinische Mikrobiologie, Immunologie und Hygiene, Technische Universität München, 81675 Munich, Germany; German Centre for Infection Research, Partner Site Munich, 81675 Munich, Germany
| |
Collapse
|
14
|
Molecular Mechanism of Gastric Carcinogenesis in Helicobacter pylori-Infected Rodent Models. Diseases 2014. [DOI: 10.3390/diseases2020168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
|
15
|
Winter JA, Letley DP, Cook KW, Rhead JL, Zaitoun AAM, Ingram RJM, Amilon KR, Croxall NJ, Kaye PV, Robinson K, Atherton JC. A role for the vacuolating cytotoxin, VacA, in colonization and Helicobacter pylori-induced metaplasia in the stomach. J Infect Dis 2014; 210:954-63. [PMID: 24625807 PMCID: PMC4136800 DOI: 10.1093/infdis/jiu154] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Carriage of Helicobacter pylori strains producing more active (s1/i1) forms of VacA is strongly associated with gastric adenocarcinoma. To our knowledge, we are the first to determine effects of different polymorphic forms of VacA on inflammation and metaplasia in the mouse stomach. Bacteria producing the less active s2/i2 form of VacA colonized mice more efficiently than mutants null for VacA or producing more active forms of it, providing the first evidence of a positive role for the minimally active s2/i2 toxin. Strains producing more active toxin forms induced more severe and extensive metaplasia and inflammation in the mouse stomach than strains producing weakly active (s2/i2) toxin. We also examined the association in humans, controlling for cagPAI status. In human gastric biopsy specimens, the vacA i1 allele was strongly associated with precancerous intestinal metaplasia, with almost complete absence of intestinal metaplasia in subjects infected with i2-type strains, even in a vacA s1, cagA+ background.
Collapse
Affiliation(s)
- Jody A Winter
- Nottingham Digestive Diseases Biomedical Research Unit, School of Medicine, University of Nottingham
| | - Darren P Letley
- Nottingham Digestive Diseases Biomedical Research Unit, School of Medicine, University of Nottingham
| | - Katherine W Cook
- Nottingham Digestive Diseases Biomedical Research Unit, School of Medicine, University of Nottingham
| | - Joanne L Rhead
- Nottingham Digestive Diseases Biomedical Research Unit, School of Medicine, University of Nottingham
| | - Abed A M Zaitoun
- Department of Pathology, Nottingham University Hospitals NHS Trust, Queen's Medical Centre Campus, United Kingdom
| | - Richard J M Ingram
- Nottingham Digestive Diseases Biomedical Research Unit, School of Medicine, University of Nottingham
| | - Karin R Amilon
- Nottingham Digestive Diseases Biomedical Research Unit, School of Medicine, University of Nottingham
| | - Nicola J Croxall
- Nottingham Digestive Diseases Biomedical Research Unit, School of Medicine, University of Nottingham
| | - Phillip V Kaye
- Department of Pathology, Nottingham University Hospitals NHS Trust, Queen's Medical Centre Campus, United Kingdom
| | - Karen Robinson
- Nottingham Digestive Diseases Biomedical Research Unit, School of Medicine, University of Nottingham
| | - John C Atherton
- Nottingham Digestive Diseases Biomedical Research Unit, School of Medicine, University of Nottingham
| |
Collapse
|
16
|
Helicobacter pylori infection in a pig model is dominated by Th1 and cytotoxic CD8+ T cell responses. Infect Immun 2013; 81:3803-13. [PMID: 23897614 DOI: 10.1128/iai.00660-13] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Helicobacter pylori infection is the leading cause for peptic ulcer disease and gastric adenocarcinoma. Mucosal T cell responses play an important role in mediating H. pylori-related gastric immunopathology. While induced regulatory T (iTreg) cells are required for chronic colonization without disease, T helper 1 (Th1) effector responses are associated with lower bacterial loads at the expense of gastric pathology. Pigs were inoculated with either H. pylori strain SS1 or J99. Phenotypic and functional changes in peripheral blood mononuclear cell (PBMC) populations were monitored weekly, and mucosal immune responses and bacterial loads were assessed up to 2 months postinfection. Both H. pylori strains elicited a Th1 response characterized by increased percentages of CD4(+)Tbet(+) cells and elevated gamma interferon (IFN-γ) mRNA in PBMCs. A subset of CD8(+) T cells expressing Tbet and CD16 increased following infection. Moreover, a significant increase in perforin and granzyme mRNA expression was observed in PBMCs of infected pigs, indicating a predominant cytotoxic immune response. Infiltration of B cells, myeloid cells, T cells expressing Treg- and Th17-associated transcription factors, and cytotoxic T cells was found in the gastric lamina propria of both infected groups. Interestingly, based on bacterial reisolation data, strain SS1 showed greater capacity to colonize and/or persist in the gastric mucosa than did strain J99. This novel pig model of infection closely mimics human gastric pathology and presents a suitable avenue for studying effector and regulatory responses toward H. pylori described in humans.
Collapse
|
17
|
Hitkova I, Yuan G, Anderl F, Gerhard M, Kirchner T, Reu S, Röcken C, Schäfer C, Schmid RM, Vogelmann R, Ebert MPA, Burgermeister E. Caveolin-1 protects B6129 mice against Helicobacter pylori gastritis. PLoS Pathog 2013; 9:e1003251. [PMID: 23592983 PMCID: PMC3623771 DOI: 10.1371/journal.ppat.1003251] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2012] [Accepted: 02/04/2013] [Indexed: 02/07/2023] Open
Abstract
Caveolin-1 (Cav1) is a scaffold protein and pathogen receptor in the mucosa of the gastrointestinal tract. Chronic infection of gastric epithelial cells by Helicobacter pylori (H. pylori) is a major risk factor for human gastric cancer (GC) where Cav1 is frequently down-regulated. However, the function of Cav1 in H. pylori infection and pathogenesis of GC remained unknown. We show here that Cav1-deficient mice, infected for 11 months with the CagA-delivery deficient H. pylori strain SS1, developed more severe gastritis and tissue damage, including loss of parietal cells and foveolar hyperplasia, and displayed lower colonisation of the gastric mucosa than wild-type B6129 littermates. Cav1-null mice showed enhanced infiltration of macrophages and B-cells and secretion of chemokines (RANTES) but had reduced levels of CD25+ regulatory T-cells. Cav1-deficient human GC cells (AGS), infected with the CagA-delivery proficient H. pylori strain G27, were more sensitive to CagA-related cytoskeletal stress morphologies (“humming bird”) compared to AGS cells stably transfected with Cav1 (AGS/Cav1). Infection of AGS/Cav1 cells triggered the recruitment of p120 RhoGTPase-activating protein/deleted in liver cancer-1 (p120RhoGAP/DLC1) to Cav1 and counteracted CagA-induced cytoskeletal rearrangements. In human GC cell lines (MKN45, N87) and mouse stomach tissue, H. pylori down-regulated endogenous expression of Cav1 independently of CagA. Mechanistically, H. pylori activated sterol-responsive element-binding protein-1 (SREBP1) to repress transcription of the human Cav1 gene from sterol-responsive elements (SREs) in the proximal Cav1 promoter. These data suggested a protective role of Cav1 against H. pylori-induced inflammation and tissue damage. We propose that H. pylori exploits down-regulation of Cav1 to subvert the host's immune response and to promote signalling of its virulence factors in host cells. Infection with the bacterium Helicobacter pylori (H. pylori) mainly affects children in the developing countries who are at risk to progress to gastric cancer (GC) as adults after many years of persistent infection, especially with strains which are positive for the oncogenic virulence factor CagA. Eradication of H. pylori by antibiotics is a treatment of choice but may also alter the susceptibility to allergies and other tumor types. Thus, novel diagnostic or prognostic markers are needed which detect early molecular changes in the stomach mucosa during the transition of chronic inflammation to cancer. In our study, we found that the tumor suppressor caveolin-1 (Cav1) is reduced upon infection with H. pylori, and CagA was sufficient but not necessary for this down-regulation. Loss of Cav1 was caused by H. pylori-dependent activation of sterol-responsive element-binding protein-1 (SREBP1), and this event abolished the interaction of Cav1 with p120 RhoGTPase-activating protein/deleted in liver cancer-1 (p120RhoGAP/DLC1), a second bona fide tumor suppressor in gastric tissue. Conclusively, Cav1 and DLC1 may constitute novel molecular markers in the H. pylori-infected gastric mucosa before neoplastic transformation of the epithelium.
Collapse
Affiliation(s)
- Ivana Hitkova
- Department of Medicine II, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Gang Yuan
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Florian Anderl
- Institute of Medical Microbiology, Immunology and Hygiene, Technische Universität München, München, Germany
| | - Markus Gerhard
- Institute of Medical Microbiology, Immunology and Hygiene, Technische Universität München, München, Germany
- German Centre for Infection Research (DZIF), München, Germany
| | - Thomas Kirchner
- Institute of Pathology, Klinikum der Universität München, München, Germany
| | - Simone Reu
- Institute of Pathology, Klinikum der Universität München, München, Germany
| | - Christoph Röcken
- Department of Pathology, Christian-Albrechts Universität, Kiel, Germany
| | - Claus Schäfer
- Department of Medicine II, Klinikum der Universität München, München, Germany
| | - Roland M. Schmid
- Department of Medicine II, Klinikum rechts der Isar, Technische Universität München, München, Germany
| | - Roger Vogelmann
- Department of Medicine II, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Matthias P. A. Ebert
- Department of Medicine II, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Elke Burgermeister
- Department of Medicine II, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- * E-mail:
| |
Collapse
|
18
|
Siegl S, Uhlig S. Using the one-lung method to link p38 to pro-inflammatory gene expression during overventilation in C57BL/6 and BALB/c mice. PLoS One 2012; 7:e41464. [PMID: 22848503 PMCID: PMC3404097 DOI: 10.1371/journal.pone.0041464] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 06/21/2012] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION The mechanisms of ventilator-induced lung injury (VILI), including the role of MAP kinases, are frequently studied in different mouse strains. A useful model for such studies is the isolated perfused mouse lung. As a further development we present the one-lung method that permits to continue perfusion and ventilation of the right lung after removal of the left lung. This method was used to compare the effect of high pressure ventilation (HPV) on pro-inflammatory signaling events in two widely used mouse strains (C57BL/6, BALB/c) and to further define the role of p38 in VILI. METHODS Lungs were perfused and ventilated for 30 min under control conditions before they were randomized to low (8 cm H(2)O) or high (25 cm H(2)O) pressure ventilation (HPV) for 210 min, with the left lung being removed after 180 min. In the left lung we measured the phosphorylation of p38, JNK, ERK and Akt kinase, and in the right lung gene expression and protein concentrations of Il1b, Il6, Tnf, Cxcl1, Cxcl2, and Areg. RESULTS Lung mechanics and kinase activation were similar in both mouse strains. HPV increased all genes (except Tnf in BALB/c) and all mediators in both strains. The gene expression of mRNA for Il1b, Il6, Cxcl1 and Cxcl2 was higher in BALB/c mice. Backward regression of the kinase data at t = 180 min with the gene and protein expression data at t = 240 min suggested that p38 controls HPV-induced gene expression, but not protein production. This hypothesis was confirmed in experiments with the p38-kinase inhibitor SB203580. CONCLUSIONS The one-lung method is useful for mechanistic studies in the lungs. While C57BL/6 show diminished pro-inflammatory responses during HPV, lung mechanics and mechanotransduction processes appear to be similar in both mouse strains. Finally, the one-lung method allowed us to link p38 to gene expression during VILI.
Collapse
Affiliation(s)
- Stephanie Siegl
- Institute of Pharmacology and Toxicology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Stefan Uhlig
- Institute of Pharmacology and Toxicology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
19
|
Nedrud JG, Czinn SJ, Ding H, Zagorski BM, Redline RW, Twaddell W, Blanchard TG. Lack of genetic influence on the innate inflammatory response to helicobacter infection of the gastric mucosa. Front Immunol 2012; 3:181. [PMID: 22783255 PMCID: PMC3389333 DOI: 10.3389/fimmu.2012.00181] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Accepted: 06/12/2012] [Indexed: 12/31/2022] Open
Abstract
Helicobacter pylori (H. pylori) is a bacterial pathogen that resides at the gastric mucosa and has a world-wide prevalence of over 50%. Infection usually lasts for the life of the host, and although all infected individuals will develop histologic gastritis only a subset will develop symptomatic gastritis, peptic ulcer disease, gastric MALT lymphoma, or gastric adenocarcinoma. The bacterial and host factors that determine clinical outcome and influence the development of widely varying diseases have not been elucidated. We compared disease in Helicobacter-infected severe combined immunodeficient (SCID) mice on different genetic backgrounds with their corresponding immunocompetent partners to determine if the genetics of the host significantly impacts the innate inflammatory outcome, independent of variations in bacterial virulence factors. BALB/c SCID and C57BL/6 SCID mice developed equivalent histologic gastritis by 8 weeks of infection. Immunocompetent BALB/c mice and C57BL/6 mice developed significantly lower or higher degrees of inflammation respectively. Innate inflammation in immunodeficient mice on the C57BL/6 background remained low even in the absence of the regulatory cytokine IL-10. These results demonstrate that adaptive immunity is not required for the generation of low level inflammation in response to Helicobacter infection and that the degree of inflammation is consistent among different genetic backgrounds. Additionally, this inflammation is limited even in the absence of regulatory T cells.
Collapse
Affiliation(s)
- John G Nedrud
- Department of Pathology, Case Western Reserve University Cleveland, OH, USA
| | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
Standardization of bacterial culture is crucial for in vivo experiments addressing Helicobacter/host -interaction. Here we present methods for bacteria culture and infection of mice.
Collapse
Affiliation(s)
- Brian M Gray
- Unit for Laboratory Animal Medicine, Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| | | |
Collapse
|
21
|
Helicobacter pylori exploits cholesterol-rich microdomains for induction of NF-kappaB-dependent responses and peptidoglycan delivery in epithelial cells. Infect Immun 2010; 78:4523-31. [PMID: 20713621 DOI: 10.1128/iai.00439-10] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Infection with Helicobacter pylori cag pathogenicity island (cagPAI)-positive strains is associated with more destructive tissue damage and an increased risk of severe disease. The cagPAI encodes a type IV secretion system (TFSS) that delivers the bacterial effector molecules CagA and peptidoglycan into the host cell cytoplasm, thereby inducing responses in host cells. It was previously shown that interactions between CagL, present on the TFSS pilus, and host α(5)β(1) integrin molecules were critical for CagA translocation and the induction of cytoskeletal rearrangements in epithelial cells. As the α(5)β(1) integrin is found in cholesterol-rich microdomains (known as lipid rafts), we hypothesized that these domains may also be involved in the induction of proinflammatory responses mediated by NOD1 recognition of H. pylori peptidoglycan. Indeed, not only did methyl-β-cyclodextrin depletion of cholesterol from cultured epithelial cells have a significant effect on the levels of NF-κB and interleukin-8 (IL-8) responses induced by H. pylori bacteria with an intact TFSS (P < 0.05), but it also interfered with TFSS-mediated peptidoglycan delivery to cells. Both of these effects could be restored by cholesterol replenishment of the cells. Furthermore, we demonstrated for the first time the involvement of α(5)β(1) integrin in the induction of proinflammatory responses by H. pylori. Taking the results together, we propose that α(5)β(1) integrin, which is associated with cholesterol-rich microdomains at the host cell surface, is required for NOD1 recognition of peptidoglycan and subsequent induction of NF-κB-dependent responses to H. pylori. These data implicate cholesterol-rich microdomains as a novel platform for TFSS-dependent delivery of bacterial products to cytosolic pathogen recognition molecules.
Collapse
|
22
|
Natural antibody to conserved targets of Haemophilus influenzae limits colonization of the murine nasopharynx. Infect Immun 2009; 77:3458-65. [PMID: 19451240 DOI: 10.1128/iai.01564-08] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Nasopharyngeal colonization represents the initial interaction between Haemophilus influenzae and its human host. Factors that influence bacterial carriage likely affect transmission and incidence of infection. Therefore, we investigated host factors involved in limiting H. influenzae colonization in BALB/c mice, as colonization can be established in this genetic background. Unlike what is observed in the C57BL/6 background, initial colonization of BALB/c mice was mainly limited by adaptive immune components. This effect on colonization did not require either CD4- or CD8-positive T cells. Instead, initial colonization by genetically diverse strains was limited by preexisting natural antibody with a lesser contribution of complement activity and the presence of neutrophils. Natural serum immunoglobulin from mice was able to bind to the bacterial surface and exhibited complement-dependent bactericidal activity against these genetically diverse H. influenzae strains. Moreover, natural immunoglobulin G (IgG) recognizing these strains was detected at the nasopharyngeal mucosal surface. This antibody-mediated effect required exposure to the normal mouse microbial flora, since mice raised under germfree (GF) conditions showed increased levels of H. influenzae colonization that were not limited by adaptive immunity. In addition, serum IgG from GF mice exhibited less surface binding to H. influenzae, suggesting that natural antibody, induced through prior exposure to the microbial flora, mediated the observed reduction in initial colonization. The broad effect of natural IgG against genetically diverse isolates suggests the presence of conserved species-wide protective targets of antibody.
Collapse
|
23
|
Mita M, Satoh M, Shimada A, Okajima M, Azuma S, Suzuki JS, Sakabe K, Hara S, Himeno S. Metallothionein is a crucial protective factor against Helicobacter pylori-induced gastric erosive lesions in a mouse model. Am J Physiol Gastrointest Liver Physiol 2008; 294:G877-84. [PMID: 18239062 DOI: 10.1152/ajpgi.00251.2007] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Infection with the gastric pathogen Helicobacter pylori (H. pylori) causes chronic gastritis, peptic ulcer, and gastric adenocarcinoma. These diseases are associated with production of reactive oxygen species (ROS) from infiltrated macrophages and neutrophiles in inflammatory sites. Metallothionein (MT) is a low-molecular-weight, cysteine-rich protein that can act not only as a metal-binding protein, but also as a ROS scavenger. In the present study, we examined the role of MT in the protection against H. pylori-induced gastric injury using MT-null mice. Female MT-null and wild-type mice were challenged with H. pylori SS1 strain, and then histological changes were evaluated with the updated Sydney grading system at 17 and 21 wk after challenge. Although the colonization efficiency of H. pylori was essentially the same for MT-null and wild-type mice, the scores of activity of inflammatory cells were significantly higher in MT-null mice than in wild-type mice at 17 wk after challenge. Histopathological examination revealed erosive lesions accompanied by infiltration of inflammatory cells in the infected MT-null mice but not in wild-type mice. Furthermore, activation of NF-kappaB and expression of NF-kappaB-mediated chemokines such as macrophage inflammatory protein-1alpha and monocytes chemoattractant protein-1 in gastric cells were markedly higher in MT-null mice than in wild-type mice. These results suggest that MT in the gastric mucosa might play an important role in the protection against H. pylori-induced gastric ulceration.
Collapse
Affiliation(s)
- Masaharu Mita
- Laboratory Animal Research Center, School of Pharmaceutical Sciences, Kitasato University, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
Nuclear factor kappaB (NF-kappaB) plays a key regulatory role in host cell responses to Helicobacter pylori infection in humans. Although mice are routinely used as a model to study H. pylori pathogenesis, the role of NF-kappaB in murine cell responses to helicobacters has not been studied in detail. We thus investigated the abilities of different Helicobacter isolates to induce NF-kappaB-dependent responses in murine gastric epithelial cells (GECs) and in transgenic mice harboring an NF-kappaB-responsive lacZ reporter gene. H. pylori and Helicobacter felis strains up-regulated the synthesis in mouse GECs of the NF-kappaB-dependent chemokines KC (CXCL1) and MIP-2 (CXCL2). These responses were cag pathogenicity island (cagPAI) independent and could be abolished by pretreatment with a pharmacological inhibitor of NF-kappaB. Consistent with the in vitro data, experimental Helicobacter infection of transgenic mice resulted in increased numbers of GECs with nuclear beta-galactosidase activity, which is indicative of specific NF-kappaB activation. The numbers of beta-galactosidase-positive cells in mice were significantly increased at day 1 postinoculation with wild-type H. pylori strains harboring or not harboring a functional cagPAI, compared to naive animals (P = 0.007 and P = 0.04, respectively). Strikingly, however, no differences were observed in the levels of gastric NF-kappaB activation at day 1 postinoculation with H. felis or at day 30 or 135 postinoculation with H. pylori. This work demonstrates for the first time the induction of NF-kappaB activation within gastric mucosal cells during acute H. pylori infection. Furthermore, the data suggest that helicobacters may be able to regulate NF-kappaB signaling during chronic infection.
Collapse
|
25
|
Maróstica M, Arçari DP, Bartchewsky W, Trevisan M, Ribeiro ML, Pedrazzoli J, Hoehr NF, Gambero A. Effects of a one-week treatment with acid gastric inhibitors on Helicobacter pylori-infected mice. Scand J Gastroenterol 2007; 42:1404-12. [PMID: 17994467 DOI: 10.1080/00365520701514396] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Antiacid drugs, including omeprazole and ranitidine, were prescribed to Helicobacter pylori-infected subjects in combination with antibiotics during eradication treatment. Several reports suggest that these drugs have additional pharmacological properties, such as antineutrophil, antiapoptotic and antioxidant characteristics. The aim of this work was to study the effects of acid suppressive medication treatment in the H. pylori infection experimental model, focusing on possible additional pharmacological properties. MATERIAL AND METHODS The ability of gastric acid suppression was assessed in pylorus-ligated animals. Gastric H. pylori colonization levels, myeloperoxidase (MPO) acitivity, macroscopic damage, Bax and Bcl-2 expression and DNA damage levels were assessed in C57BL/6-infected mice after treatment for one week with omeprazole (100 mg kg(-1)) or ranitidine (100 mg kg(-1)). RESULTS Omeprazole treatment increased bacteria colonization and MPO activity in mice stomachs. Both antiacid drugs efficiently improved macroscopic damage, although only omeprazole restored the expression of the antiapoptotic Bcl-2 protein in gastric mucosa of infected animals. CONCLUSIONS Some additional omeprazole-related properties, such as antineutrophil properties, were not observed in H. pylori-infected mice after one week of treatment, suggesting that this property is restricted to in vitro approaches. However, the antiapoptotic activity of omeprazole could be attributed to an ability to modify the protein expression of Bcl-2, decreased by H. pylori infection.
Collapse
Affiliation(s)
- Marta Maróstica
- Clinical Pharmacology and Gastroenterology Unit, São Francisco University Medical School, Bragança Paulista, SP, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Algood HMS, Gallo-Romero J, Wilson KT, Peek RM, Cover TL. Host response to Helicobacter pylori infection before initiation of the adaptive immune response. ACTA ACUST UNITED AC 2007; 51:577-86. [PMID: 17919297 DOI: 10.1111/j.1574-695x.2007.00338.x] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Helicobacter pylori persistently colonizes the human stomach. In this study, immune responses to H. pylori that occur in the early stages of infection were investigated. Within the first 2 days after orogastric infection of mice with H. pylori, there was a transient infiltration of macrophages and neutrophils into the glandular stomach. By day 10 postinfection, the numbers of macrophages and neutrophils decreased to baseline levels. By 3 weeks postinfection, an adaptive immune response was detected, marked by gastric infiltration of T lymphocytes, macrophages, and neutrophils, as well as increased numbers of H. pylori-specific T cells, macrophages, and dendritic cells in paragastric lymph nodes. Neutrophil-attracting and macrophage-attracting chemokines were expressed at higher levels in the stomachs of H. pylori-infected mice than in the stomachs of uninfected mice. Increased expression of TNFalpha and IFNgamma (Th1-type inflammatory cytokines) and IL-17 (a Th17-type cytokine) was detected in the stomachs of H. pylori-infected mice, but increased expression of IL-4 (a Th2-type cytokine) was not detected. These data indicate that a transient gastric inflammatory response to H. pylori occurs within the first few days after infection, before the priming of T cells and initiation of an adaptive immune response. It is speculated that inappropriate waning of the innate immune response during early stages of infection may be a factor that contributes to H. pylori persistence.
Collapse
Affiliation(s)
- Holly M Scott Algood
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | | | | | | | | |
Collapse
|
27
|
Hoffelner H, Rieder G, Haas R. Helicobacter pylori vaccine development: optimisation of strategies and importance of challenging strain and animal model. Int J Med Microbiol 2007; 298:151-9. [PMID: 17714988 DOI: 10.1016/j.ijmm.2007.07.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Gastric infection with the gram-negative bacterial pathogen Helicobacter pylori is widespread (approximately 50% of the human population is affected) and is associated with the induction of specific gastroduodenal disease. Although extensive studies in the H. pylori mouse model have demonstrated the feasibility of both therapeutic and prophylactic immunisations, the mechanism of vaccine-induced protection is still poorly understood. We report here on novel strategies to optimise the generation of H. pylori ghosts as vaccine candidates and highlight the need to concentrate on alternative animal models and the use of fully virulent H. pylori type I strains for vaccination. An effective vaccine strategy against H. pylori has the potential to significantly improve population health worldwide.
Collapse
Affiliation(s)
- Herbert Hoffelner
- Max von Pettenkofer Institut für Hygiene und Medizinische Mikrobiologie, LMU München, Pettenkoferstrasse 9a, D-80336 München, Germany
| | | | | |
Collapse
|
28
|
Williams SM, Chen YT, Andermann TM, Carter JE, McGee DJ, Ottemann KM. Helicobacter pylori chemotaxis modulates inflammation and bacterium-gastric epithelium interactions in infected mice. Infect Immun 2007; 75:3747-57. [PMID: 17517875 PMCID: PMC1952010 DOI: 10.1128/iai.00082-07] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The ulcer-causing pathogen Helicobacter pylori uses directed motility, or chemotaxis, to both colonize the stomach and promote disease development. Previous work showed that mutants lacking the TlpB chemoreceptor, one of the receptors predicted to drive chemotaxis, led to less inflammation in the gerbil stomach than did the wild type. Here we expanded these findings and examined the effects on inflammation of completely nonchemotactic mutants and mutants lacking each chemoreceptor. Of note, all mutants colonized mice to the same levels as did wild-type H. pylori. Infection by completely nonchemotactic mutants (cheW or cheY) resulted in significantly less inflammation after both 3 and 6 months of infection. Mutants lacking either the TlpA or TlpB H. pylori chemotaxis receptors also had alterations in inflammation severity, while mutants lacking either of the other two chemoreceptors (TlpC and HylB) behaved like the wild type. Fully nonchemotactic and chemoreceptor mutants adhered to cultured gastric epithelial cells and caused cellular release of the chemokine interleukin-8 in vitro similar to the release caused by the wild type. The situation appeared to be different in the stomach. Using silver-stained histological sections, we found that nonchemotactic cheY or cheW mutants were less likely than the wild type to be intimately associated with the cells of the gastric mucosa, although there was not a strict correlation between intimate association and inflammation. Because others have shown that in vivo adherence promotes inflammation, we propose a model in which H. pylori uses chemotaxis to guide it to a productive interaction with the stomach epithelium.
Collapse
Affiliation(s)
- Susan M Williams
- Department of Environmental Toxicology (ETOX), University of California at Santa Cruz, 1156 High Street, Santa Cruz, CA 95064, USA
| | | | | | | | | | | |
Collapse
|
29
|
Ruggiero P, Rossi G, Tombola F, Pancotto L, Lauretti L, Del Giudice G, Zoratti M. Red wine and green tea reduce H pylori- or VacA-induced gastritis in a mouse model. World J Gastroenterol 2007; 13:349-54. [PMID: 17230601 PMCID: PMC4065887 DOI: 10.3748/wjg.v13.i3.349] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate whether red wine and green tea could exert anti-H pylori or anti-VacA activity in vivo in a mouse model of experimental infection.
METHODS: Ethanol-free red wine and green tea concentrates were administered orally as a mixture of the two beverages to H pylori infected mice, or separately to VacA-treated mice. Gastric colonization and gastric inflammation were quantified by microbiological, histopathological, and immunohistochemical analyses.
RESULTS: In H pylori-infected mice, the red wine and green tea mixture significantly prevented gastritis and limited the localization of bacteria and VacA to the surface of the gastric epithelium. Similarly, both beverages significantly prevented gastric epithelium damage in VacA-treated mice; green tea, but not red wine, also altered the VacA localization in the gastric epithelium.
CONCLUSION: Red wine and green tea are able to prevent H pylori-induced gastric epithelium damage, possibly involving VacA inhibition. This observation supports the possible relevance of diet on the pathological outcome of H pylori infection.
Collapse
Affiliation(s)
- Paolo Ruggiero
- Novartis Vaccines & Diagnostics s.r.l., Research Center, Via Fiorentina 1, Siena I-53100, Italy.
| | | | | | | | | | | | | |
Collapse
|
30
|
Baldwin DN, Shepherd B, Kraemer P, Hall MK, Sycuro LK, Pinto-Santini DM, Salama NR. Identification of Helicobacter pylori genes that contribute to stomach colonization. Infect Immun 2006; 75:1005-16. [PMID: 17101654 PMCID: PMC1828534 DOI: 10.1128/iai.01176-06] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Chronic infection of the human stomach by Helicobacter pylori leads to a variety of pathological sequelae, including peptic ulcer and gastric cancer, resulting in significant human morbidity and mortality. Several genes have been implicated in disease related to H. pylori infection, including the vacuolating cytotoxin and the cag pathogenicity island. Other factors important for the establishment and maintenance of infection include urease enzyme production, motility, iron uptake, and stress response. We utilized a C57BL/6 mouse infection model to query a collection of 2,400 transposon mutants in two different bacterial strain backgrounds for H. pylori genetic loci contributing to colonization of the stomach. Microarray-based tracking of transposon mutants allowed us to monitor the behavior of transposon insertions in 758 different gene loci. Of the loci measured, 223 (29%) had a predicted colonization defect. These included previously described H. pylori virulence genes, genes implicated in virulence in other pathogenic bacteria, and 81 hypothetical proteins. We have retested 10 previously uncharacterized candidate colonization gene loci by making independent null alleles and have confirmed their colonization phenotypes by using competition experiments and by determining the dose required for 50% infection. Of the genetic loci retested, 60% have strain-specific colonization defects, while 40% have phenotypes in both strain backgrounds for infection, highlighting the profound effect of H. pylori strain variation on the pathogenic potential of this organism.
Collapse
Affiliation(s)
- David N Baldwin
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | | | | | | | | | | | |
Collapse
|
31
|
Ruggiero P, Tombola F, Rossi G, Pancotto L, Lauretti L, Del Giudice G, Zoratti M. Polyphenols reduce gastritis induced by Helicobacter pylori infection or VacA toxin administration in mice. Antimicrob Agents Chemother 2006; 50:2550-2. [PMID: 16801443 PMCID: PMC1489755 DOI: 10.1128/aac.01042-05] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Helicobacter pylori colonizes the human gastric mucosa, causing inflammation that leads to atrophic gastritis, and it can cause peptic ulcer and gastric cancer. We show that polyphenol administration to mice experimentally infected by H. pylori or treated with VacA toxin can limit gastric epithelium damage, an effect that may be linked to VacA inhibition.
Collapse
Affiliation(s)
- P Ruggiero
- Chiron s.r.l., Research Center, Via Fiorentina 1, I-53100 Siena, Italy.
| | | | | | | | | | | | | |
Collapse
|
32
|
McCracken VJ, Martin SM, Lorenz RG. The Helicobacter felis model of adoptive transfer gastritis. Immunol Res 2006; 33:183-94. [PMID: 16234583 DOI: 10.1385/ir:33:2:183] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The bacterium Helicobacter pylori is a major human pathogen and the principal cause of acute and chronic gastritis, gastric and duodenal ulcer disease, and gastric adenocarcinoma. Infection with gastric Helicobacter results in an early infiltration of neutrophils, monocytes, and natural killer cells, followed by an influx of T cells and plasma cells. Although the critical components of this gastric infiltrate that lead to disease are unclear, the Helicobacter felis-infected mouse and other mouse models of Helicobacter-associated gastritis have demonstrated the critical nature of adaptive immunity in the development of gastric epithelial pathology. To further investigate the role of adaptive immunity in this disease, adoptive transfer models of disease have also been utilized. These models clearly demonstrate that it is the host CD4+ T lymphocyte response that is crucial for the development of Helicobacter-associated gastric epithelial changes.
Collapse
Affiliation(s)
- Vance J McCracken
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL 35294-2170, USA
| | | | | |
Collapse
|
33
|
Spinetti N, Zavala-Spinetti L, Hempe J, Correa H, Bégué RE. Effect of long-term Helicobacter felis infection in a mouse model of streptozotocin-induced diabetes. Helicobacter 2005; 10:586-91. [PMID: 16302984 DOI: 10.1111/j.1523-5378.2005.00362.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
BACKGROUND We previously REPORTED that mice with diabetes and short-term Helicobacter felis infection had an increase in glycated hemoglobin (HbA1c). Here we report the effect of long-term infection. MATERIALS AND METHODS Six-week-old C57BL/6 mice were injected with streptozotocin to induce diabetes and started on daily insulin. Following streptozotocin injection, animals were paired according to their HbA1c values and randomized to orally receive either H. felis or culture medium alone. Weight and HbA1c were monitored monthly for 6 months. RESULTS Thirty animals corresponding to 15 pairs were included in the study. H. felis-infected diabetic mice developed significantly more gastritis than uninfected animals. Sixteen mice died during the observation period. As compared to uninfected animals, infected mice died more frequently (40% versus 67%, p = .14) and earlier (160 versus 61 days, p = .20); both variables combined showed that H. felis infection significantly decreased the chances of survival during the study period (p = .045). In addition, infected mice showed a trend for higher increase in their HbA1c (0.97 +/- 2.5% versus - 0.22 +/- 3.0%; p = .21) and lower weight gain (2.0 +/- 3.4 g versus 2.9 +/- 2.0 g; p = .15) than uninfected mice. CONCLUSION Long-term H. felis infection had a deleterious effect in mice with streptozotocin-induced diabetes resulting in increased mortality. If the same phenomenon occurs in humans this could lead to interventions to improve the long-term outcome of patients with diabetes.
Collapse
Affiliation(s)
- Nelson Spinetti
- Department of Pediatrics, Lousiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | | | | | | | | |
Collapse
|
34
|
De Bock M, Decostere A, Van den Bulck K, Baele M, Duchateau L, Haesebrouck F, Ducatelle R. The inflammatory response in the mouse stomach to Helicobacter bizzozeronii, Helicobacter salomonis and two Helicobacter felis Strains. J Comp Pathol 2005; 133:83-91. [PMID: 15949811 DOI: 10.1016/j.jcpa.2005.01.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2004] [Accepted: 01/18/2005] [Indexed: 12/21/2022]
Abstract
The inflammatory response in the mouse stomach was evaluated as a means of distinguishing different non-pylori Helicobacter (H.) strains in terms of virulence. Mice of four strains (BALB/c, SJL, C57BL/6 and CFW) were infected intragastrically with four bacterial strains (H. felis ATCC 49179 and CCUG 37471, H. bizzozeronii and H. salomonis). The animals were killed for gastric examination at 3, 9 or 16 weeks post-inoculation. H. salomonis could not be detected by the polymerase chain reaction, but the other three organisms were detected in all stomach samples at all timepoints. SJL mice consistently showed particularly severe gastric inflammation regardless of bacterial strain. Lymphocytes and occasionally neutrophils were seen in submucosa and lamina propria mucosae. BALB/c mice showed the least severe inflammatory changes. H. bizzozeronii differed from the two H. felis strains in producing less striking pathological changes in mice. Of the two H. felis strains, ATCC 49179 produced the more severe inflammatory changes in SJL mice.
Collapse
Affiliation(s)
- M De Bock
- Department of Pathology, Bacteriology and Avian Diseases, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| | | | | | | | | | | | | |
Collapse
|
35
|
Jeremy AHT, Du Y, Dixon MF, Robinson PA, Crabtree JE. Protection against Helicobacter pylori infection in the Mongolian gerbil after prophylactic vaccination. Microbes Infect 2005; 8:340-6. [PMID: 16213184 DOI: 10.1016/j.micinf.2005.06.025] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2005] [Revised: 06/21/2005] [Accepted: 06/28/2005] [Indexed: 12/23/2022]
Abstract
Vaccines against Helicobacter pylori could circumvent the problem of increasing antibiotic resistance. They would be particularly useful in developing countries, where re-infection rates are high following standard eradication regimes. The Mongolian gerbil is a good model for H. pylori infection, as the gastric pathology induced by infection is similar to that in humans. The H. pylori-induced inflammatory response in gerbils is considerably greater than in murine models. The aim of this study was to determine if gerbils could be vaccinated against H. pylori. Mongolian gerbils were vaccinated orally with an H. pylori whole cell sonicate preparation and cholera toxin adjuvant. Vaccinated gerbils and controls were challenged with the autologous H. pylori strain 42GX. All infection, and cholera toxin, control gerbils were H. pylori positive 6 weeks post-challenge. By contrast, a significant degree of protection was demonstrated in vaccinated gerbils. Only two of 10 of gerbils were H. pylori positive (P<0.001). Protection was associated with increased serum H. pylori IgG antibodies. Protected gerbils had histologically normal gastric mucosa and, in contrast to mice, no post-immunisation gastritis was evident. In the control groups, the degree of inflammation was variable, with some of the animals having corpus gastritis and corpus mucous metaplasia. The levels of gastric IL-12p40 and IFNgamma transcripts were significantly decreased in vaccinated animals compared to infection and cholera toxin controls (P<0.01). Gastric IL-10 and TGFbeta transcripts were found only at relatively low levels. These results demonstrate that Mongolian gerbils can be successfully vaccinated against H. pylori and protected from H. pylori-induced pathology.
Collapse
Affiliation(s)
- A H T Jeremy
- Molecular Medicine Unit, Level 7 Clinical Sciences Building, St. James's University Hospital, Leeds LS9 7TF, UK
| | | | | | | | | |
Collapse
|
36
|
Padol IT, Hunt RH. Host-specific differences in the physiology of acid secretion related to prostaglandins may play a role in gastric inflammation and injury. Am J Physiol Gastrointest Liver Physiol 2005; 288:G1110-7. [PMID: 15677554 DOI: 10.1152/ajpgi.00364.2004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Immune mediators are involved in strain-specific manifestations of Helicobacter pylori infection, and the type of immune response is associated with production of PGE(2), which in turn influences gastric acid secretion. Acid secretion plays a pivotal role, not only in the pattern of H. pylori-induced gastritis and its consequences, but also in nonsteroidal anti-inflammatory drug (NSAID)-induced gastropathies. Mice and their transgenic modifications are widely used in Helicobacter and eicosanoid research. Using [(14)C]aminopyrine accumulation and pylorus ligation, we aimed to study acid secretion in gastric gland preparations from the commonly used strains of BALB/c and C57BL/6 mice. We found that PGE(2) does not inhibit acid secretion in gastric glands from C57BL/6 mice, in contrast to the expected antisecretory effect of PGE(2) observed in BALB/c mice. In BALB/c mice the effect of histamine and carbachol was reduced by PGE(2), whereas in C57BL/6 mice dose-response curves to these secretagogues were not affected. EP(3) receptors are not involved in acid secretion in C57BL/6 mice, as confirmed by significantly lower expression of mRNA for the EP(3) receptor. These contrary findings are important to the interpretation of the antisecretory role of eicosanoids in BALB/c and C57BL/6 mouse strains and the involvement of prostanoids in the etiology of Helicobacter-induced inflammation and NSAID-induced gastropathies. We propose that the lack of antisecretory effect of PGE(2) observed in C57BL/6 mice could reflect the extent of Helicobacter-induced inflammation and status of acid secretion in response to anti-inflammatory drugs.
Collapse
Affiliation(s)
- Ireneusz T Padol
- Div. of Gastroenterology, McMaster Univ., Health Sciences Centre, Rm. 4W8A, 1200 Main St. West, Hamilton, ON, Canada L8N 3Z5
| | | |
Collapse
|
37
|
Terio KA, Munson L, Marker L, Aldridge BM, Solnick JV. Comparison of Helicobacter spp. in Cheetahs (Acinonyx jubatus) with and without gastritis. J Clin Microbiol 2005; 43:229-34. [PMID: 15634976 PMCID: PMC540127 DOI: 10.1128/jcm.43.1.229-234.2005] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Chronic gastritis causes significant morbidity and mortality in captive cheetahs but is rare in wild cheetahs despite colonization by abundant spiral bacteria. This research aimed to identify the Helicobacter species that were associated with gastritis in captive cheetahs but are apparently commensal in wild cheetahs. Helicobacter species were characterized by PCR amplification and sequencing of the 16S rRNA, urease, and cagA genes and by transmission electron microscopy of frozen or formalin-fixed paraffin-embedded gastric samples from 33 cheetahs infected with Helicobacter organisms (10 wild without gastritis and 23 captive with gastritis). Samples were screened for mixed infections by denaturant gel gradient electrophoresis of the 16S rRNA gene and by transmission electron microscopy. There was no association between Helicobacter infection and the presence or severity of gastritis. Eight cheetahs had 16S rRNA sequences that were most similar (98 to 99%) to H. pylori. Twenty-five cheetahs had sequences that were most similar (97 to 99%) to "H. heilmannii" or H. felis. No cheetahs had mixed infections. The ultrastructural morphology of all bacteria was most consistent with "H. heilmannii," even when 16S rRNA sequences were H. pylori-like. The urease gene from H. pylori-like bacteria could not be amplified with primers for either "H. heilmannii" or H. pylori urease, suggesting that this bacteria is neither H. pylori nor "H. heilmannii." The cagA gene was not identified in any case. These findings question a direct role for Helicobacter infection in the pathogenesis of gastritis and support the premise that host factors account for the differences in disease between captive and wild cheetah populations.
Collapse
Affiliation(s)
- K A Terio
- Department of Pathology, Microbiology and Immunology, School of Veterinary Medicine, University of California, Davis, USA.
| | | | | | | | | |
Collapse
|
38
|
Bergman MP, Vandenbroucke-Grauls CMJE, Appelmelk BJ, D'Elios MM, Amedei A, Azzurri A, Benagiano M, Del Prete G. The story so far: Helicobacter pylori and gastric autoimmunity. Int Rev Immunol 2005; 24:63-91. [PMID: 15763990 DOI: 10.1080/08830180590884648] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The gastric mucosal pathogen Helicobacter pylori induces autoantibodies directed against the gastric proton pump H+,K+-ATPase in 20-30% of infected patients. The presence of these autoantibodies is associated with severity of gastritis, increased atrophy, and apoptosis in the corpus mucosa, and patients with these autoantibodies infected with H. pylori display histopathological and clinical features that are similar to those of autoimmune gastritis (AIG). This review will focus on the T helper cell responses, cytokines, and adhesion molecules involved in corpus mucosal atrophy in chronic H. pylori gastritis and in AIG, and the role of H. pylori in the onset of AIG.
Collapse
Affiliation(s)
- Mathijs P Bergman
- Department of Medical Microbiology and Infection Control, VU Medical Center, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Fonseca-Aten M, Ríos AM, Mejías A, Chávez-Bueno S, Katz K, Gómez AM, McCracken GH, Hardy RD. Mycoplasma pneumoniae induces host-dependent pulmonary inflammation and airway obstruction in mice. Am J Respir Cell Mol Biol 2004; 32:201-10. [PMID: 15626776 DOI: 10.1165/rcmb.2004-0197oc] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Respiratory tract infections result in wheezing in a subset of patients. Mycoplasma pneumoniae is a common etiologic agent of acute respiratory infection in children and adults that has been associated with wheezing in 20-40% of individuals. The current study was undertaken to elucidate the host-dependent pulmonary and immunologic response to M. pneumoniae respiratory infection by studying mice with different immunogenetic backgrounds (BALB/c mice versus C57BL/6 mice). After M. pneumoniae infection, only BALB/c mice developed significant airway obstruction (AO) compared with controls. M. pneumoniae-infected BALB/c mice manifested significantly elevated airway hyperresponsiveness (AHR) compared with C57BL/6 mice 4 and 7 d after inoculation as well as BALB/c control mice. Compared with C57BL/6 mice, BALB/c mice developed worse pulmonary inflammation, including greater peribronchial infiltrates. Infected BALB/c mice had significantly higher concentrations of tumor necrosis factor-alpha, interferon-gamma, interleukin (IL)-1beta, IL-6, IL-12, KC (functional IL-8), and macrophage inflammatory protein 1alpha in the bronchoalveolar lavage fluid compared with infected C57BL/6 mice. No differences in IL-2, IL-4, IL-5, IL-10, and granulocyte/macrophage colony-stimulating factor concentrations were found. The mice in this study exhibited host-dependent infection-related AO and AHR associated with chemokine and T-helper type (Th)1 pulmonary host response and not Th2 response after M. pneumoniae infection.
Collapse
Affiliation(s)
- Monica Fonseca-Aten
- Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9063, USA.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Yuan JP, Li T, Chen HB, Li ZH, Yang GZ, Hu BY, Shi XD, Tong SQ, Li YX, Guo XK. Analysis of gene expression profile in gastric cancer cells stimulated with Helicobacter pylori isogenic strains. J Med Microbiol 2004; 53:965-974. [PMID: 15358818 DOI: 10.1099/jmm.0.45634-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
To understand the biological processes within host cells induced by VacA, isogenic strains of Helicobacter pylori (NCTC 11638 or 11638-DeltavacA) were used to stimulate gastric cancer cells SGC7901, and differentially expressed genes in host cells were identified using cDNA microarray technology. More than 300 genes were found to alter their mRNA expression at different time points, among which 68 were related to the cytoskeleton, 87 were associated with cell cycle, cell death and proliferation, IL8 expression was also found to be up-regulated. Cells co-cultured with broth-culture supernatant (BCS) of NCTC 11638 showed more alteration in microtubule cytoskeleton morphology, as observed by laser scanning confocal microscopy, and a lower apoptosis rate, detected by flow cytometry, compared with those co-cultured with BCS of 11638-DeltavacA. The supernatants of cells co-cultured with NCTC 11638 showed significantly higher IL8 expression than those co-cultured with 11638-DeltavacA. It is concluded that VacA disrupts cytoskeletal architecture by influencing the expression of cytoskeleton-associated genes. VacA breaks the balance between cell proliferation and cell death by inducing the maladjustment of genes related to cell cycle. VacA is also able to induce the inflammatory response.
Collapse
Affiliation(s)
- Jian-Ping Yuan
- Department of Medical Microbiology and Parasitology, Shanghai Second Medical University, Shanghai, China 2Institute of Immunology, Second Military Medical University, Shanghai, China 3Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Tao Li
- Department of Medical Microbiology and Parasitology, Shanghai Second Medical University, Shanghai, China 2Institute of Immunology, Second Military Medical University, Shanghai, China 3Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hua-Biao Chen
- Department of Medical Microbiology and Parasitology, Shanghai Second Medical University, Shanghai, China 2Institute of Immunology, Second Military Medical University, Shanghai, China 3Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Zhen-Hong Li
- Department of Medical Microbiology and Parasitology, Shanghai Second Medical University, Shanghai, China 2Institute of Immunology, Second Military Medical University, Shanghai, China 3Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Gui-Zhen Yang
- Department of Medical Microbiology and Parasitology, Shanghai Second Medical University, Shanghai, China 2Institute of Immunology, Second Military Medical University, Shanghai, China 3Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Bao-Yu Hu
- Department of Medical Microbiology and Parasitology, Shanghai Second Medical University, Shanghai, China 2Institute of Immunology, Second Military Medical University, Shanghai, China 3Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xiao-Dong Shi
- Department of Medical Microbiology and Parasitology, Shanghai Second Medical University, Shanghai, China 2Institute of Immunology, Second Military Medical University, Shanghai, China 3Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Shan-Qing Tong
- Department of Medical Microbiology and Parasitology, Shanghai Second Medical University, Shanghai, China 2Institute of Immunology, Second Military Medical University, Shanghai, China 3Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yi-Xue Li
- Department of Medical Microbiology and Parasitology, Shanghai Second Medical University, Shanghai, China 2Institute of Immunology, Second Military Medical University, Shanghai, China 3Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xiao-Kui Guo
- Department of Medical Microbiology and Parasitology, Shanghai Second Medical University, Shanghai, China 2Institute of Immunology, Second Military Medical University, Shanghai, China 3Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
41
|
Mueller A, Merrell DS, Grimm J, Falkow S. Profiling of microdissected gastric epithelial cells reveals a cell type-specific response to Helicobacter pylori infection. Gastroenterology 2004; 127:1446-62. [PMID: 15521014 DOI: 10.1053/j.gastro.2004.08.054] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND AND AIMS Helicobacter pylori colonizes the epithelial lining of the human stomach and is associated with disorders ranging from chronic gastritis to peptic ulcers and gastric cancer. We have explored the transcriptional response of the epithelium globally by applying a whole-genome approach to a murine model of infection. METHODS The 3 major epithelial lineages of the stomach-the parietal, mucus-producing, and chief cells-were harvested from cryosections of infected and uninfected murine stomachs by laser microdissection and subjected to gene expression profiling. The localization and quantity of selected transcripts were verified by in situ hybridization and quantitative real-time reverse-transcription polymerase chain reaction. RESULTS Each cell type is characterized by a transcriptional signature profile. The parietal cell profile is highly enriched for factors involved in mitochondrial energy generation, whereas the chief cell predominantly expresses digestive enzymes and glycosylation-associated proteins. In contrast, the mucus cell signature is distinguished by an abundance of cell-surface receptors, signaling molecules, and factors involved in antigen presentation. All of these indicate a role in sampling, sensing, and responding to environmental stimuli. In line with this biological function, we measured a strong transcriptional response to Helicobacter pylori infection only in this cell type. The genes that are differentially expressed upon infection are implicated in a proinflammatory and mucosal defense response as well as modulation of angiogenesis, iron availability, and tumor suppression. CONCLUSIONS Laser microdissection-assisted transcriptional profiling is a useful tool to explore the biology of specific cell populations and is sensitive enough to measure the transcriptional response to bacterial infection in vivo.
Collapse
Affiliation(s)
- Anne Mueller
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | | | | | | |
Collapse
|
42
|
Park JH, Kim DJ, Park YH, Seok SH, Cho SA, Baek MW, Lee HY, Park JH. Characteristics of the gastritis induced by Listeria monocytogenes in mice: microbiology, histopathology, and mRNA expression of inflammatory mediators with time course of infection. Microb Pathog 2004; 37:87-94. [PMID: 15312848 DOI: 10.1016/j.micpath.2004.05.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2003] [Revised: 05/18/2004] [Accepted: 05/24/2004] [Indexed: 11/23/2022]
Abstract
Listeria monocytogenes induces the suppurative gastritis in some mice strains. In this study, characteristics of the gastritis caused by L. monocytogenes infection in mice were examined with time course of infection. Mice were administered intragastrically with 1.8 x 10(8) CFU of L. monocytogenes. Each three mice were sacrificed by cervical dislocation at 1, 3, 5, 7, 10, 14, 17, 21, and 28 days postinoculation (pi), respectively. Bacterial colonization in the stomachs reached the peak at 3 days pi, maintained over 4.3 log10 CFU/g tissue until 14 days pi, and was cleared by 28 days pi. However, in the spleens and livers, the bacteria could not be detected after 7 days pi. The gastric lesions were the most prominent at between 3 and 7 days pi. The lesions consisted of marked neutrophilic infiltration, edema, vacuolar degeneration and necrosis of muscle cells and were more severe in the nonglandular region and fundus than in the pylorus, and were in submucosa, lamina muscularis, and serosa than in mucosa. mRNA expression of several cytokines (INF-gamma, IL-1beta, IL-5, IL-6, IL-12, and TNF-alpha) and chemokines (KC, MCP-1) increased in the gastric tissue of infected mice at 1-7 days pi and slightly decreased at 14 days pi. These findings would be useful for studying the pathological mechanism of human febrile gastroenteritis due to L. monocytogenes infection.
Collapse
Affiliation(s)
- Jong-Hwan Park
- Department of Laboratory Animal Medicine, College of Veterinary Medicine and School of Agricultural Biotechnology, Seoul National University, San 56-1, Shinlim-dong, Kwanak-ku, Seoul 151-742, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Walduck A, Schmitt A, Lucas B, Aebischer T, Meyer TF. Transcription profiling analysis of the mechanisms of vaccine‐induced protection against
H. pylori. FASEB J 2004; 18:1955-7. [PMID: 15456742 DOI: 10.1096/fj.04-2321fje] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Development of a vaccine against H. pylori is regarded as desirable alternative to the current antibiotic therapy regimens. Mice immunized with an attenuated recombinant Salmonella typhimurium expressing H. pylori urease subunits A&B have dramatically reduced bacterial loads after a single dose. The mechanism(s) of protection against this largely extra-cellular pathogen are not fully understood. The aim of this study was to identify genes that were regulated specifically in response to immunization, in order to gain a broader picture of the immune response in the immunized gastric epithelium. Gene expression in RNA isolated from the gastric mucosa of immunized and infected Balb/c mice was compared with that in infected only mice at 1, 3, and 14 days after challenge with a mouse-adapted strain of H. pylori. We show that infection with H. pylori causes an immediate reaction in vivo, which was clearly divided into acute and chronic phases, and further that the transcriptional response in the H. pylori infected and immunized gastric mucosa is unique. Analysis of gene expression patterns at day 14 post-infection suggested not only the beginning of a lymphocytic infiltrate, but of an integrated epithelial response characterized by increased expression of genes controlling cell cycle and turnover. This observation was confirmed in independent experiments. The global approach has brought new insights to the effect of immunization on the gastric epithelium and has led us to propose a new multi-factorial model for the mechanisms underlying vaccine-induced protection.
Collapse
Affiliation(s)
- Anna Walduck
- Max-Planck Institute for Infection Biology, Department of Molecular Biology, Berlin, Germany
| | | | | | | | | |
Collapse
|
44
|
Cole SP, Harwood J, Lee R, She R, Guiney DG. Characterization of monospecies biofilm formation by Helicobacter pylori. J Bacteriol 2004; 186:3124-32. [PMID: 15126474 PMCID: PMC400600 DOI: 10.1128/jb.186.10.3124-3132.2004] [Citation(s) in RCA: 158] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2003] [Accepted: 10/24/2003] [Indexed: 12/24/2022] Open
Abstract
As all bacteria studied to date, the gastric pathogen Helicobacter pylori has an alternate lifestyle as a biofilm. H. pylori forms biofilms on glass surfaces at the air-liquid interface in stationary or shaking batch cultures. By light microscopy, we have observed attachment of individual, spiral H. pylori to glass surfaces, followed by division to form microcolonies, merging of individual microcolonies, and growth in the third dimension. Scanning electron micrographs showed H. pylori arranged in a matrix on the glass with channels for nutrient flow, typical of other bacterial biofilms. To understand the importance of biofilms to the H. pylori life cycle, we tested the effect of mucin on biofilm formation. Our results showed that 10% mucin greatly increased the number of planktonic H. pylori while not affecting biofilm bacteria, resulting in a decline in percent adherence to the glass. This suggests that in the mucus-rich stomach, H. pylori planktonic growth is favored over biofilm formation. We also investigated the effect of specific mutations in several genes, including the quorum-sensing gene, luxS, and the cagE type IV secretion gene. Both of these mutants were found to form biofilms approximately twofold more efficiently than the wild type in both assays. These results indicate the relative importance of these genes to the production of biofilms by H. pylori and the selective enhancement of planktonic growth in the presence of gastric mucin.
Collapse
Affiliation(s)
- Sheri P Cole
- Department of Medicine, University of California-San Diego, La Jolla, California 92093-0640, USA
| | | | | | | | | |
Collapse
|
45
|
Jenks PJ, Jeremy AHT, Robinson PA, Walker MM, Crabtree JE. Long-term infection with Helicobacter felis and inactivation of the tumour suppressor gene p53 cumulatively enhance the gastric mutation frequency in Big Blue transgenic mice. J Pathol 2004; 201:596-602. [PMID: 14648663 DOI: 10.1002/path.1488] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The aims of this study were to determine whether colonization with Helicobacter felis resulted in the accumulation of mutations within murine gastric tissue and whether the degree of genetic damage was increased by p53 deficiency. Female C57BL/6 mice carrying either the lambda/lacI transgene (Big Blue transgenic mice) or the lambda/lacI transgene and deficient in one allele of the p53 tumour suppressor gene (TSG-p53/Big Blue) were inoculated with H felis. Seven months after inoculation, mutations in the target lacI gene were assessed using the Big Blue transgenic mutagenesis assay system in these animals and in controls. There was an approximately two-fold increase in lacI mutations in gastric mucosa harvested from mice infected with H felis and also from non-infected mice heterozygous for the p53 allele relative to wild-type mice. The mutation frequency in mice infected with H felis and deficient in one allele of p53 was increased approximately three-fold. Active gastric inflammation was significantly greater in H felis-infected p53 hemizygous mice compared with H felis p53 wild-type mice. Gastric epithelial proliferation was similarly increased with infection in both of these latter groups of mice. In infected mice, there was a significant correlation between the mutation frequency and the degree of active gastric inflammation. These data suggest a synergistic action between infection with H felis and p53 deficiency in the accumulation of mutations within gastric tissue. Active neutrophil infiltration in gastric Helicobacter infection may contribute to the increased levels of mutation observed.
Collapse
Affiliation(s)
- Peter J Jenks
- Institute of Infections, Immunity and Inflammation, University of Nottingham, Nottingham, UK
| | | | | | | | | |
Collapse
|
46
|
Court M, Robinson PA, Dixon MF, Jeremy AHT, Crabtree JE. The effect of gender on Helicobacter felis-mediated gastritis, epithelial cell proliferation, and apoptosis in the mouse model. J Pathol 2003; 201:303-11. [PMID: 14517848 DOI: 10.1002/path.1422] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The murine Helicobacter felis model has been extensively used to investigate the importance of host factors in the development of chronic gastritis. The effect of gender in this murine model is unknown. Male and female C57BL/6J mice were infected with H felis for up to 1 year. At 4, 8, 19, 36, and 52 weeks post-infection, gastric histopathology, epithelial cell proliferation, and apoptosis were examined and compared with age- and gender-matched controls. In female mice, infection with H felis resulted in an earlier onset of chronic gastric inflammation, epithelial hyperplasia, and oxyntic cell loss than males. In females, there was a trend towards increased gastric pathology compared with males, with long-term-infected female mice having significantly greater (p < 0.05) chronic inflammation than male mice. The histopathological differences in male and female mice did not relate to the density of H felis infection. Female mice infected with H felis had significantly increased gastric epithelial cell proliferation in the cardia and corpus at both 8 and 52 weeks post-infection (p < 0.05). Epithelial cell apoptosis in the glandular mucosa of the corpus at 36 and 52 weeks post-infection was significantly increased (p < 0.05) in female mice compared with uninfected gender controls. In contrast, there was no significant increase in epithelial cell proliferation or apoptosis in any area of the stomach at any time point after H felis infection in male mice. These results demonstrate that there are gender differences in the gastric inflammatory and epithelial response to H felis in the murine model. The functional importance of gender should be considered in future murine studies on H felis- and H pylori-induced chronic gastritis.
Collapse
Affiliation(s)
- M Court
- Molecular Medicine Unit, St James's University Hospital, Leeds, UK
| | | | | | | | | |
Collapse
|
47
|
Abstract
Subclinical gastritis was observed in 10 of 10 baboons (Papio spp.) from a toxicity study in a research facility. The lesions were similar in xenobiotic-treated and control animals, suggesting a spontaneous rather than chemical-induced disease. Histologic examination revealed lymphoplasmacytic gastritis in the antral mucosa. The fundic mucosa contained minor, scattered aggregates of lymphocytes and plasma cells. A Warthin-Starry silver stain and ultrastructural examination revealed numerous spiral-shaped bacteria morphologically resembling Helicobacter pylori in antral glands and numerous spiral-shaped bacteria morphologically consistent with H. heilmannii-like organisms in fundic glands. Polymerase chain reaction assay of paraffin-embedded antral and fundic tissue using primers for the urease gene and 16S ribosomal ribonucleic acid gene amplified deoxyribonucleic acid fragments with a high degree of sequence homology for H. pylori and H. heilmannii. This is the first report of gastritis associated with Helicobacter-like organisms in baboons.
Collapse
Affiliation(s)
- J T Mackie
- IDEXX-Veterinary Pathology Services, PO Box 1119, Coorparoo DC, Qld 4151, Australia.
| | | |
Collapse
|
48
|
Ohana M, Okazaki K, Oshima C, Kawasaki K, Fukui T, Tamaki H, Matsuura M, Asada M, Nishi T, Uchida K, Uose S, Nakase H, Iwano M, Matsushima Y, Hiai H, Chiba T. Inhibitory effects of Helicobacter pylori infection on murine autoimmune gastritis. Gut 2003; 52:1102-10. [PMID: 12865266 PMCID: PMC1773742 DOI: 10.1136/gut.52.8.1102] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND AIM Long term Helicobacter pylori infection leads to atrophic gastritis but the relation between H pylori infection and autoimmune related atrophic gastritis (AIG) remains unclear. We studied the effects of H pylori infection on the pathophysiology of AIG in mice. MATERIALS AND METHODS BALB/c nu/nu mice (n=40) with or without H pylori infection received splenocytes from neonatally thymectomised mice to induce AIG. Half of the mice were orally infected with H pylori prior to AIG induction. Histological findings, and local and systemic immune responses were serially evaluated. RESULTS Two and six months after transfer, parietal cells in uninfected mice were depleted while those in infected mice were well preserved. The degree of gland atrophy (p<0.01), hyperplasia (p<0.01), gastric pH (p<0.05), and serum gastrin levels of infected mice were significantly lower than those of uninfected mice. Serum antiparietal cell antibody levels gradually decreased in infected mice, and were significantly lower than those of uninfected mice at six months (p<0.05). Real time polymerase chain reaction studies revealed significantly higher interleukin 4 (p<0.05) and transforming growth factor beta (p<0.05) gene expression in the gastric mucosa in infected mice than in uninfected mice at both two and six months after AIG induction. CONCLUSIONS H pylori infection inhibited the development of AIG in mice. Th2-type immune responses and transforming growth factor beta in the gastric microenvironment might be involved in the inhibitory effects of H pylori infection on the development of AIG, in which Th1-type responses have an important role.
Collapse
Affiliation(s)
- M Ohana
- Department of Gastroenterology and Endoscopic Medicine, Kyoto University, Sakyo, Kyoto, 606-8507 Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Wang J, Court M, Jeremy AHT, Aboshkiwa MA, Robinson PA, Crabtree JE. Infection of Mongolian gerbils with Chinese Helicobacter pylori strains. FEMS IMMUNOLOGY AND MEDICAL MICROBIOLOGY 2003; 36:207-13. [PMID: 12738393 DOI: 10.1016/s0928-8244(02)00464-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
To date only a few Helicobacter pylori strains have been demonstrated to colonise Mongolian gerbils successfully. The aim of this study was to establish stable colonisation of Chinese strains of H. pylori in gerbils. Fresh clinical isolates from Chinese patients were inoculated into gerbils. At 4-6 weeks post inoculation, infection status was evaluated by culture, biopsy urease test and pathology. Sequencing of glmM and random amplified polymorphic DNA (RAPD) fingerprinting of DNA from cultured H. pylori were used to evaluate the genetic identity of pre-inoculated and post-inoculated strains. The ability of pre- and post-inoculated strains to stimulate interleukin-8 transcription in L5F11 gastric epithelial cells was analysed. Three of five clinical isolates colonised gerbils. The three pre- and post-inoculation strains had identical glmM sequences and RAPD profiles, and stimulated luciferase secretion from L5F11 epithelial cells. The strain that caused severe pathological changes was selected for repeat infection to prove reproducible and stable colonisation. The cagA+ strain 42GX gave stable colonisation in the gerbil and induced severe gastritis.
Collapse
Affiliation(s)
- Jingtong Wang
- Molecular Medicine Unit, Level 7 Clinical Sciences Building, St. James's University Hospital, Leeds LS9 7TF, UK
| | | | | | | | | | | |
Collapse
|
50
|
Straubinger RK, Greiter A, McDonough SP, Gerold A, Scanziani E, Soldati S, Dailidiene D, Dailide G, Berg DE, Simpson KW. Quantitative evaluation of inflammatory and immune responses in the early stages of chronic Helicobacter pylori infection. Infect Immun 2003; 71:2693-703. [PMID: 12704144 PMCID: PMC153233 DOI: 10.1128/iai.71.5.2693-2703.2003] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2002] [Revised: 10/14/2002] [Accepted: 02/10/2003] [Indexed: 01/08/2023] Open
Abstract
The early consequences of Helicobacter pylori infection and the role of bacterial virulence determinants in disease outcome remain to be established. The present study sought to measure the development of host inflammatory and immune responses and their relationship to the putative bacterial virulence factors cag pathogenicity island (cagPAI), vacA allele, and oipA in combination with bacterial colonization density in a feline model of the early stages of H. pylori infection. Gastric tissues obtained from infected and uninfected cats were evaluated for H. pylori ureB, cagPAI, vacA allele, and oipA and colonization density (urease, histology, and real-time PCR). Inflammation was assessed by measuring mRNA upregulation of gamma interferon (IFN-gamma), interleukin (IL)-1 alpha, IL-1 beta, IL-4, IL-6, IL-8, IL-10, and IL-12 p40 and histopathology. The mucosal immune response was characterized by morphometric analysis of lymphoid follicles and by differentiating lymphocyte populations with antibodies against surface markers. Infecting H. pylori strains were positive for vacAs1 but lacked cagPAI and an active oipA gene. Colonization density was uniform throughout the stomach. Upregulation of IFN-gamma, IL-1 alpha, IL-1 beta, and IL-8 and increased severity of inflammatory infiltrates and fibrosis were observed in infected cats. The median number and total area of lymphoid aggregates were 5 and 10 times greater, respectively, in the stomachs of infected than uninfected cats. Secondary lymphoid follicles in uninfected cats were rare and positive for BLA.36 and B220 but negative for CD3 and CD79 alpha, whereas in infected cats they were frequent and positive for BLA.36, CD79 alpha, and CD3 but negative for B220. Upregulation of IFN-gamma, IL-1 alpha, IL-1 beta, and IL-8 and marked hyperplasia of secondary lymphoid follicles are early consequences of H. pylori infection in cats. The response appears to be similar to that of infected people, particularly children, can develop independently of the pathogenicity factors cagPAI and oipA, and is not correlated with the degree of colonization density or urease activity.
Collapse
|