1
|
Duan Y, Xu Y, Dou Y, Xu D. Helicobacter pylori and gastric cancer: mechanisms and new perspectives. J Hematol Oncol 2025; 18:10. [PMID: 39849657 PMCID: PMC11756206 DOI: 10.1186/s13045-024-01654-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 12/23/2024] [Indexed: 01/25/2025] Open
Abstract
Gastric cancer remains a significant global health challenge, with Helicobacter pylori (H. pylori) recognized as a major etiological agent, affecting an estimated 50% of the world's population. There has been a rapidly expanding knowledge of the molecular and pathogenetic mechanisms of H. pylori over the decades. This review summarizes the latest research advances to elucidate the molecular mechanisms underlying the H. pylori infection in gastric carcinogenesis. Our investigation of the molecular mechanisms reveals a complex network involving STAT3, NF-κB, Hippo, and Wnt/β-catenin pathways, which are dysregulated in gastric cancer caused by H. pylori. Furthermore, we highlight the role of H. pylori in inducing oxidative stress, DNA damage, chronic inflammation, and cell apoptosis-key cellular events that pave the way for carcinogenesis. Emerging evidence also suggests the effect of H. pylori on the tumor microenvironment and its possible implications for cancer immunotherapy. This review synthesizes the current knowledge and identifies gaps that warrant further investigation. Despite the progress in our previous knowledge of the development in H. pylori-induced gastric cancer, a comprehensive investigation of H. pylori's role in gastric cancer is crucial for the advancement of prevention and treatment strategies. By elucidating these mechanisms, we aim to provide a more in-depth insights for the study and prevention of H. pylori-related gastric cancer.
Collapse
Affiliation(s)
- Yantao Duan
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yonghu Xu
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi Dou
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Dazhi Xu
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
2
|
Zahir SO, El Khadir M, Boukhris SA, Benajah DA, Ibrahimi SA, Chbani L, El Abkari M, Bennani B. Helicobacter pylori vacA Allelic Combination, dupA, cagE and cagA Genotypes and Their Associations with Gastric Diseases in the Moroccan Population. Jpn J Infect Dis 2024; 77:325-333. [PMID: 38945860 DOI: 10.7883/yoken.jjid.2024.061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
This study aimed to investigate the combination of the four regions of Helicobacter pylori vacA with cagA, cagE, dupA genes and cagA-EPIYA motifs to identify the most likely combination that could be used as a disease determinant marker in the Moroccan population. A total of 838 H. pylori-positive samples were obtained from consenting patients, that were previously analyzed by PCR to characterize vacA-s, -m, and -i regions; cagE status; and cagA 3' region polymorphism, were used to characterize vacA-d region and to determine dupA gene status. The analysis showed the predominance of the less virulent combination {vacA(s2m2i2d2)dupA(-)cagE(-)cagA(-)}, and showed that the risk of gastric cancer is 13.33 fold higher (95% confidence interval [CI] = 1.06-166.37) in patients infected with strains harboring vacA(s1m1i1d1)dupA(-)cagE(+)cagA(2EPIYA-C) compared to patients with gastritis without lesions and infected by H. pylori strains harboring vacA(s2m2i2d2)dupA(-) cagE(-)cagA(-). Infection with strains harboring the vacA(s1m1i1d1)dupA(+)cagE(+)cagA(1EPIYA-C) genotype combination represented a risk factor for both gastric ulcer and duodenal ulcer than gastritis without lesions; odds ratio (OR) =16 (95% CI = 1.09-234.24) and OR = 12.39 (95% CI = 1.09-140.81), respectively. These results suggest that the combination of the active form of vacA genotypes, dupA gene status, and the number of EPIYA-C motifs may be helpful markers for discriminating between several gastric diseases.
Collapse
Affiliation(s)
- Souad Oirdi Zahir
- URL-CNRST No. 15, Laboratory of Human Pathology Biomedicine and Environment, Pharmacy and Dentistry of Fez (FMPDF), Sidi Mohammed Ben Abdellah University (USMBA), Morocco
- Laboratory of Microbiology and Molecular Biology, Faculty of Medicine, Pharmacy and Dentistry of Fez (FMPDF), Sidi Mohammed Ben Abdellah University (USMBA), Morocco
| | - Mounia El Khadir
- URL-CNRST No. 15, Laboratory of Human Pathology Biomedicine and Environment, Pharmacy and Dentistry of Fez (FMPDF), Sidi Mohammed Ben Abdellah University (USMBA), Morocco
- Laboratory of Microbiology and Molecular Biology, Faculty of Medicine, Pharmacy and Dentistry of Fez (FMPDF), Sidi Mohammed Ben Abdellah University (USMBA), Morocco
- The Higher Institute of Nursing Professions and Health Techniques (ISPITS), Morocco
| | - Samia Alaoui Boukhris
- URL-CNRST No. 15, Laboratory of Human Pathology Biomedicine and Environment, Pharmacy and Dentistry of Fez (FMPDF), Sidi Mohammed Ben Abdellah University (USMBA), Morocco
- Laboratory of Microbiology and Molecular Biology, Faculty of Medicine, Pharmacy and Dentistry of Fez (FMPDF), Sidi Mohammed Ben Abdellah University (USMBA), Morocco
| | - Dafr-Allah Benajah
- URL-CNRST No. 15, Laboratory of Human Pathology Biomedicine and Environment, Pharmacy and Dentistry of Fez (FMPDF), Sidi Mohammed Ben Abdellah University (USMBA), Morocco
- Department of Hepato-Gastroenterology, University Hospital Center Hassan II, Morocco
| | - Sidi Adil Ibrahimi
- URL-CNRST No. 15, Laboratory of Human Pathology Biomedicine and Environment, Pharmacy and Dentistry of Fez (FMPDF), Sidi Mohammed Ben Abdellah University (USMBA), Morocco
- Department of Hepato-Gastroenterology, University Hospital Center Hassan II, Morocco
| | - Laila Chbani
- Department of Pathological Anatomy, University Hospital Center Hassan II, Morocco
| | - Mohamed El Abkari
- URL-CNRST No. 15, Laboratory of Human Pathology Biomedicine and Environment, Pharmacy and Dentistry of Fez (FMPDF), Sidi Mohammed Ben Abdellah University (USMBA), Morocco
- Department of Hepato-Gastroenterology, University Hospital Center Hassan II, Morocco
| | - Bahia Bennani
- URL-CNRST No. 15, Laboratory of Human Pathology Biomedicine and Environment, Pharmacy and Dentistry of Fez (FMPDF), Sidi Mohammed Ben Abdellah University (USMBA), Morocco
- Laboratory of Microbiology and Molecular Biology, Faculty of Medicine, Pharmacy and Dentistry of Fez (FMPDF), Sidi Mohammed Ben Abdellah University (USMBA), Morocco
| |
Collapse
|
3
|
JAK-STAT1 Signaling Pathway Is an Early Response to Helicobacter pylori Infection and Contributes to Immune Escape and Gastric Carcinogenesis. Int J Mol Sci 2022; 23:ijms23084147. [PMID: 35456965 PMCID: PMC9031264 DOI: 10.3390/ijms23084147] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 03/22/2022] [Indexed: 01/05/2023] Open
Abstract
Helicobacter pylori infection induces a number of pro-inflammatory signaling pathways contributing to gastric inflammation and carcinogenesis and has been identified as a major risk factor for the development of gastric cancer (GC). Janus kinase-signal transducer and activator of transcription (JAK-STAT) signaling mediates immune regulatory processes, including tumor-driven immune escape. Programmed death ligand 1 (PD-L1) expressed on gastric epithelium can suppress the immune system by shutting down T cell effector function. In a human cohort of subjects with gastric lesions and GC analyzed by proteomics, STAT1 increased along the cascade of progression of precancerous gastric lesions to GC and was further associated with a poor prognosis of GC (Hazard Ratio (95% confidence interval): 2.34 (1.04-5.30)). We observed that STAT1 was activated in human H. pylori-positive gastritis, while in GC, STAT1, and its target gene, PD-L1, were significantly elevated. To confirm the dependency of H. pylori, we infected gastric epithelial cells in vitro and observed strong activation of STAT1 and upregulation of PD-L1, which depended on cytokines produced by immune cells. To investigate the correlation of immune infiltration with STAT1 activation and PD-L1 expression, we employed a mouse model of H. pylori-induced gastric lesions in an Rnf43-deficient background. Here, phosphorylated STAT1 and PD-L1 were correlated with immune infiltration and proliferation. STAT1 and PD-L1 were upregulated in gastric tumor tissues compared with normal tissues and were associated with immune infiltration and poor prognosis based on the TCGA-STAD database. H. pylori-induced activation of STAT1 and PD-L1 expression may prevent immune surveillance in the gastric mucosa, allowing premalignant lesions to progress to gastric cancer.
Collapse
|
4
|
Helicobacter pylori outer membrane vesicles induce expression and secretion of oncostatin M in AGS gastric cancer cells. Braz J Microbiol 2021; 52:1057-1066. [PMID: 33851342 DOI: 10.1007/s42770-021-00490-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 04/06/2021] [Indexed: 01/01/2023] Open
Abstract
Helicobacter pylori, a human pathogen that colonizes the stomach of 50% of the world's population, is associated with gastritis, gastric adenocarcinoma, and mucosa-associated lymphoid tissue (MALT) lymphoma. Diseases are characterized by severe inflammatory responses in the stomach that are induced by various chemokines and cytokines. Recently, oncostatin M (OSM), an IL-6 family cytokine, was detected in early gastric cancer biopsies. In this study, we showed that Helicobacter pylori induced secretion of OSM and overexpression of its type II receptor OSMRβ (OSM/OSMRβ) in a human gastric adenocarcinoma cell line (AGS) over 24 h of infection. Furthermore, we showed that the induction of OSM and OSMRβ was carried out by heat-sensitive Helicobacter pylori outer membrane vesicle (OMV) protein. Collectively, our results established, for the first time, a direct relation between Helicobacter pylori OMVs and the OSM/OSMRβ signaling axis.
Collapse
|
5
|
El Khadir M, Boukhris SA, Zahir SO, Benajah DA, Ibrahimi SA, Chbani L, El Abkari M, Bennani B. CagE, cagA and cagA 3' region polymorphism of Helicobacter pylori and their association with the intra-gastric diseases in Moroccan population. Diagn Microbiol Infect Dis 2021; 100:115372. [PMID: 33813354 DOI: 10.1016/j.diagmicrobio.2021.115372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 03/02/2021] [Accepted: 03/06/2021] [Indexed: 10/21/2022]
Abstract
Helicobacter pylori infection is the most important etiological factor in gastroduodenal diseases development. Its evolution is influenced by several factors, including bacterial virulence genes such as cagA and cagE. This work aimed to evaluate the predictive value of cagE alone and in combination with cagA and CagA-EPIYA-C motifs number as a marker of the infection evolution. A total of 823 H. pylori DNA extracted from biopsies of consenting patients suffering from gastritis, peptic ulcer, or gastric cancer. The cagE, cagA status and cagA 3' region polymorphism were determined by PCR. The analysis shows that the risk of duodenal ulcer is 1.97-fold higher (CI = 1.18-3.30) in patients infected by strains cagA+/cagE+. And the risk of gastric cancer is 5.19-fold higher (CI = 1.18-22.70) in patients harboring strains cagE+/2EPIYA-C. The results suggest that cagE in combination with cagA-EPIYA-C motifs number can be used as predictive biomarker of H. pylori infection evolution.
Collapse
Affiliation(s)
- Mounia El Khadir
- Laboratoire de Pathologie Humaine Biomédecine et Environnement, Equipe micro-organismes génomique et facteurs oncogènes, Faculté de médecine et de Pharmacie de Fès (FMPF), Université Sidi Mohammed Ben Abdellah (USMBA), Fès, Maroc.; Laboratoire de microbiologie et de Biologie Moléculaire, FMPF, USMBA
| | - Samia Alaoui Boukhris
- Laboratoire de Pathologie Humaine Biomédecine et Environnement, Equipe micro-organismes génomique et facteurs oncogènes, Faculté de médecine et de Pharmacie de Fès (FMPF), Université Sidi Mohammed Ben Abdellah (USMBA), Fès, Maroc.; Laboratoire de microbiologie et de Biologie Moléculaire, FMPF, USMBA
| | - Souad Oirdi Zahir
- Laboratoire de Pathologie Humaine Biomédecine et Environnement, Equipe micro-organismes génomique et facteurs oncogènes, Faculté de médecine et de Pharmacie de Fès (FMPF), Université Sidi Mohammed Ben Abdellah (USMBA), Fès, Maroc.; Laboratoire de microbiologie et de Biologie Moléculaire, FMPF, USMBA
| | - Dafr-Allah Benajah
- Laboratoire de Pathologie Humaine Biomédecine et Environnement, Equipe micro-organismes génomique et facteurs oncogènes, Faculté de médecine et de Pharmacie de Fès (FMPF), Université Sidi Mohammed Ben Abdellah (USMBA), Fès, Maroc.; Service d'Hépato gastro-entérologie CHU Hassan II, Fès, Maroc
| | - Sidi Adil Ibrahimi
- Laboratoire de Pathologie Humaine Biomédecine et Environnement, Equipe micro-organismes génomique et facteurs oncogènes, Faculté de médecine et de Pharmacie de Fès (FMPF), Université Sidi Mohammed Ben Abdellah (USMBA), Fès, Maroc.; Service d'Hépato gastro-entérologie CHU Hassan II, Fès, Maroc
| | - Laila Chbani
- Service d'Anatomie pathologique CHU Hassan II, Fès, Maroc
| | - Mohamed El Abkari
- Laboratoire de Pathologie Humaine Biomédecine et Environnement, Equipe micro-organismes génomique et facteurs oncogènes, Faculté de médecine et de Pharmacie de Fès (FMPF), Université Sidi Mohammed Ben Abdellah (USMBA), Fès, Maroc.; Service d'Hépato gastro-entérologie CHU Hassan II, Fès, Maroc
| | - Bahia Bennani
- Laboratoire de Pathologie Humaine Biomédecine et Environnement, Equipe micro-organismes génomique et facteurs oncogènes, Faculté de médecine et de Pharmacie de Fès (FMPF), Université Sidi Mohammed Ben Abdellah (USMBA), Fès, Maroc.; Laboratoire de microbiologie et de Biologie Moléculaire, FMPF, USMBA.
| |
Collapse
|
6
|
Algood HMS. T Cell Cytokines Impact Epithelial Cell Responses during Helicobacter pylori Infection. THE JOURNAL OF IMMUNOLOGY 2020; 204:1421-1428. [PMID: 32152211 DOI: 10.4049/jimmunol.1901307] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 12/08/2019] [Indexed: 12/24/2022]
Abstract
The goal of this Brief Review is to highlight literature that demonstrates how cytokines made by T lymphocytes impact the gastric epithelium, especially during Helicobacter pylori infection. These cytokines effect many of the diverse functions of the epithelium and the epithelium's interactions with H. pylori The focal point of this Brief Review will be on how T cell cytokines impact antimicrobial function and barrier function and how T cell cytokines influence the development and progression of cancer. Furthermore, the modulation of epithelial-derived chemokines by H. pylori infection will be discussed.
Collapse
Affiliation(s)
- Holly M Scott Algood
- Veterans Affairs Tennessee Valley Healthcare Services, Nashville, TN 37212; Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37212; and Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37212
| |
Collapse
|
7
|
Alarcón-Millán J, Martínez-Carrillo DN, Peralta-Zaragoza O, Fernández-Tilapa G. Regulation of GKN1 expression in gastric carcinogenesis: A problem to resolve (Review). Int J Oncol 2019; 55:555-569. [PMID: 31322194 DOI: 10.3892/ijo.2019.4843] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 07/04/2019] [Indexed: 11/05/2022] Open
Abstract
Gastrokine 1 (GKN1) is a protein expressed on the surface mucosa cells of the gastric antrum and fundus, which contributes to maintaining gastric homeostasis, inhibits inflammation and is a tumor suppressor. The expression of GKN1 decreases in mucosa that are either inflamed or infected by Helicobacter pylori, and is absent in gastric cancer. The measurement of circulating GKN1 concentration, the protein itself, or the mRNA in gastric tissue may be of use for the early diagnosis of cancer. The mechanisms that modulate the deregulation or silencing of GKN1 expression have not been completely described. The modification of histones, methylation of the GKN1 promoter, or proteasomal degradation of the protein have been detected in some patients; however, these mechanisms do not completely explain the absence of GKN1 or the reduction in GKN1 levels. Only NKX6.3 transcription factor has been shown to be a positive modulator of GKN1 transcription, although others also have an affinity with sequences in the promoter of this gene. While microRNAs (miRNAs) are able to directly or indirectly regulate the expression of genes at the post‑transcriptional level, the involvement of miRNAs in the regulation of GKN1 has not been reported. The present review analyzes the information reported on the determination of GKN1 expression and the regulation of its expression at the transcriptional, post‑transcriptional and post‑translational levels; it proposes an integrated model that incorporates the regulation of GKN1 expression via transcription factors and miRNAs in H. pylori infection.
Collapse
Affiliation(s)
- Judit Alarcón-Millán
- Clinical Research Laboratory, Faculty of Biological Chemical Sciences, Guerrero Autonomous University, Chilpancingo, Guerrero 39070, México
| | - Dinorah Nashely Martínez-Carrillo
- Clinical Research Laboratory, Faculty of Biological Chemical Sciences, Guerrero Autonomous University, Chilpancingo, Guerrero 39070, México
| | - Oscar Peralta-Zaragoza
- Direction of Chronic Infections and Cancer, Research Center in Infection Diseases, National Institute of Public Health, Cuernavaca, Morelos 62100, México
| | - Gloria Fernández-Tilapa
- Clinical Research Laboratory, Faculty of Biological Chemical Sciences, Guerrero Autonomous University, Chilpancingo, Guerrero 39070, México
| |
Collapse
|
8
|
Asl SF, Pourvahedi M, Mojtahedi A, Shenagari M. Analysis of babA, cagE and cagA Genes in Helicobacter pylori from Upper Gastric Patients in the North of Iran. Infect Disord Drug Targets 2019; 19:274-278. [PMID: 29766826 DOI: 10.2174/1871526518666180515113218] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 03/20/2018] [Accepted: 04/10/2018] [Indexed: 06/08/2023]
Abstract
OBJECTIVE Helicobacter pylori is a Gram-negative bacterium which has a serious effect on up to half of the world's population and has been related to different gastric diseases. The goal of this study was to assess the frequency of babA, cagE and cagA genotypes among H. pylori strains isolated from gastric biopsies of endoscopic patients in the north of Iran. METHODS The present study was performed on 90 strains of H. pylori isolated from patients with gastric diseases (Gastric ulcer (GU), Duodenal ulcer (DU), Gastritis (G), Non-ulcer dyspepsia (NUD) and Gastric adenocarcinoma (GC)). DNA was extracted from all isolated strains and PCR method was performed to detect the prevalence of babA2, cagE and cagA genes using specific primers. RESULTS Among 90 samples of H. pylori, babA2, cagE, and cagA genes were detected in 42.2%, 30% and 82.2% of strains respectively. The statistical analysis showed that the prevalence of cagA gene in GU, G, DU, and NUD was significantly higher than other genes. Moreover, cagA, and babA2 genes were significantly more prevalent in GC patients compared to cagE gene. Our isolates exhibited 8 distinct arrangements of virulence patterns. The occurrence of cagA (35.6%) was the most prevalent pattern followed by cagA/babA2 (20%) and cagA/babA2/cagE (14.4%). CONCLUSION In summary, as first report from Guilan province in the north of Iran, we showed significant association between the presence of babA2, cagE, and cagA genes in different types of gastric disorders.
Collapse
Affiliation(s)
- Saba Fakhrieh Asl
- Internal Medicine Department, Poursina Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Mehrnaz Pourvahedi
- Genetics Department, Faculty of Basic Sciences, Guilan University, Rasht, Iran
| | - Ali Mojtahedi
- Cellular and Molecular Research Center, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Microbiology Department, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammad Shenagari
- Microbiology Department, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
9
|
Ibana JA, Sherchand SP, Fontanilla FL, Nagamatsu T, Schust DJ, Quayle AJ, Aiyar A. Chlamydia trachomatis-infected cells and uninfected-bystander cells exhibit diametrically opposed responses to interferon gamma. Sci Rep 2018; 8:8476. [PMID: 29855501 PMCID: PMC5981614 DOI: 10.1038/s41598-018-26765-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 05/18/2018] [Indexed: 12/13/2022] Open
Abstract
The intracellular bacterial pathogen, Chlamydia trachomatis, is a tryptophan auxotroph. Therefore, induction of the host tryptophan catabolizing enzyme, indoleamine-2,3-dioxgenase-1 (IDO1), by interferon gamma (IFNγ) is one of the primary protective responses against chlamydial infection. However, despite the presence of a robust IFNγ response, active and replicating C. trachomatis can be detected in cervical secretions of women. We hypothesized that a primary C. trachomatis infection may evade the IFNγ response, and that the protective effect of this cytokine results from its activation of tryptophan catabolism in bystander cells. To test this hypothesis, we developed a novel method to separate a pool of cells exposed to C. trachomatis into pure populations of live infected and bystander cells and applied this technique to distinguish between the effects of IFNγ on infected and bystander cells. Our findings revealed that the protective induction of IDO1 is suppressed specifically within primary infected cells because Chlamydia attenuates the nuclear import of activated STAT1 following IFNγ exposure, without affecting STAT1 levels or phosphorylation. Critically, the IFNγ-mediated induction of IDO1 activity is unhindered in bystander cells. Therefore, the IDO1-mediated tryptophan catabolism is functional in these cells, transforming these bystander cells into inhospitable hosts for a secondary C. trachomatis infection.
Collapse
Affiliation(s)
- Joyce A Ibana
- Immunopharmacology Research Laboratory, Institute of Biology, College of Science, University of the Philippines, Diliman, Quezon City, 1101, Philippines.
| | - Shardulendra P Sherchand
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Francis L Fontanilla
- Immunopharmacology Research Laboratory, Institute of Biology, College of Science, University of the Philippines, Diliman, Quezon City, 1101, Philippines
| | - Takeshi Nagamatsu
- Department of Obstetrics and Gynecology, Faculty Medicine, University of Tokyo, Tokyo, Japan
| | - Danny J Schust
- Department of Obstetrics, Gynecology, and Women's Health, University of Missouri, Columbia, MO, 15276, USA
| | - Alison J Quayle
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Ashok Aiyar
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| |
Collapse
|
10
|
Sica A, Erreni M, Allavena P, Porta C. Macrophage polarization in pathology. Cell Mol Life Sci 2015; 72:4111-26. [PMID: 26210152 PMCID: PMC11113543 DOI: 10.1007/s00018-015-1995-y] [Citation(s) in RCA: 499] [Impact Index Per Article: 49.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 07/07/2015] [Accepted: 07/16/2015] [Indexed: 01/05/2023]
Abstract
Macrophages are cells of the innate immunity constituting the mononuclear phagocyte system and endowed with remarkable different roles essential for defense mechanisms, development of tissues, and homeostasis. They derive from hematopoietic precursors and since the early steps of fetal life populate peripheral tissues, a process continuing throughout adult life. Although present essentially in every organ/tissue, macrophages are more abundant in the gastro-intestinal tract, liver, spleen, upper airways, and brain. They have phagocytic and bactericidal activity and produce inflammatory cytokines that are important to drive adaptive immune responses. Macrophage functions are settled in response to microenvironmental signals, which drive the acquisition of polarized programs, whose extremes are simplified in the M1 and M2 dichotomy. Functional skewing of monocyte/macrophage polarization occurs in physiological conditions (e.g., ontogenesis and pregnancy), as well as in pathology (allergic and chronic inflammation, tissue repair, infection, and cancer) and is now considered a key determinant of disease development and/or regression. Here, we will review evidence supporting a dynamic skewing of macrophage functions in disease, which may provide a basis for macrophage-centered therapeutic strategies.
Collapse
Affiliation(s)
- Antonio Sica
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", via Bovio 6, Novara, Italy.
- Humanitas Clinical and Research Center, Via Manzoni 56, 20089, Milan, Rozzano, Italy.
| | - Marco Erreni
- Humanitas Clinical and Research Center, Via Manzoni 56, 20089, Milan, Rozzano, Italy
| | - Paola Allavena
- Humanitas Clinical and Research Center, Via Manzoni 56, 20089, Milan, Rozzano, Italy
| | - Chiara Porta
- Department of Pharmaceutical Sciences, Università del Piemonte Orientale "Amedeo Avogadro", via Bovio 6, Novara, Italy
| |
Collapse
|
11
|
Helicobacter pylori Outer Membrane Protein 18 (Hp1125) Is Involved in Persistent Colonization by Evading Interferon- γ Signaling. BIOMED RESEARCH INTERNATIONAL 2015; 2015:571280. [PMID: 25945338 PMCID: PMC4402576 DOI: 10.1155/2015/571280] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 10/09/2014] [Accepted: 10/23/2014] [Indexed: 12/30/2022]
Abstract
Outer membrane proteins (OMPs) can induce an immune response. Omp18 (HP1125) of H. pylori is a powerful antigen that can induce significant interferon-γ (IFN-γ) levels. Previous studies have suggested that IFN-γ plays an important role in H. pylori clearance. However, H. pylori has multiple mechanisms to avoid host immune surveillance for persistent colonization. We generated an omp18 mutant (H. pylori 26695 and H. pylori SS1) strain to examine whether Omp18 interacts with IFN-γ and is involved in H. pylori colonization. qRT-PCR revealed that IFN-γ induced Omp18 expression. qRT-PCR and western blot analysis revealed reduced expressions of virulence factors CagA and NapA in H. pylori 26695 with IFN-γ treatment, but they were induced in the Δomp18 strain. In C57BL/6 mice infected with H. pylori SS1 and the Δomp18 strain, the Δomp18 strain conferred defective colonization and activated a stronger inflammatory response. Signal transducer phosphorylation and transcription 1 (STAT1) activator was downregulated by the wild-type strain but not the Δomp18 strain in IFN-γ-treated macrophages. Furthermore, Δomp18 strain survival rates were poor in macrophages compared to the wild-type strain. We concluded that H. pylori Omp18 has an important function influencing IFN-γ-mediated immune response to participate in persistent colonization.
Collapse
|
12
|
Khakzad MR, Saffari A, Mohamadpour N, Sankian M, Varasteh A, Salari F, Meshkat M. TLR4 and TLR2 expression in biopsy specimens from antral and corporal stomach zones in Helicobacter pylori infections. Rep Biochem Mol Biol 2014; 3:29-37. [PMID: 26989734 PMCID: PMC4757086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 07/16/2014] [Indexed: 06/05/2023]
Abstract
BACKGROUND It is not yet known which types of Toll-like receptors (TLRs) are most effective in Helicobacter pylori (H. pylori) recognition. It is also not known which gastric zones have the most prominent roles in TLR-mediated bacterial recognition. The aim of this work was to analyze the expression of TLR2 and TLR4 in biopsy specimens from H. pylori-infected patients. METHODS Thirty-eight patients with gastrointestinal disorders were divided into four groups in this study. The groups were: (A) H. pylori infection and peptic ulcer (n=15), (B) peptic ulcer only (n=5), (C) H. pylori infection only (n=10) and (D) control, with neither H. pylori infection nor peptic ulcer (n=8). Biopsy specimens from sites of redness or atrophic mucosa from gastric antrum and body in patients with gastritis were collected. RNAs from the antrum and body specimens were isolated. TLR2 and TLR4 mRNA expression was assessed by RT-PCR and quantified as densitometric ratios of TLR2 and TLR4/β-actin mRNA. RESULTS In the antral zones of H. pylori-infected patients (Groups A and C) TLR2 and TLR4 expression was significantly greater than in uninfected patients (Groups B and D) regardless of peptic ulcers (p < 0.05). In the gastric body samples TLR2 expression was significantly greater in Group C (H. pylori infection only) than in Group B (peptic ulcer only) and TLR4 expression was significantly greater in group A (H. pylori infection and peptic ulcer) than in Group B (peptic ulcer only) (p < 0.05). No significant differences in expression of TLR4 and TLR2 were observed between samples from the antrum and body in same groups. CONCLUSIONS We conclude that H. pylori infection leads to significant increase in TLR2 and TLR4 molecules expression in antral region related to the control group. Considering the stimulatory effect of H. pylori on TLRs expression in the gastric tissue, we assume that colonization of H. pylori infection might occurs more in the gastric antral region than in the gastric body.
Collapse
Affiliation(s)
- Mohammad Reza Khakzad
- Department of Immunology, Zakariya Research Center, Mashhad Branch, Islamic Azad University, Mashhad, Iran
- Allergy Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ahmad Saffari
- Internal Medicine Department, Mashhad Branch, Aria Hospital, Islamic Azad University, Mashhad, Iran
| | - Niloofar Mohamadpour
- Department of Microbiology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Mojtaba Sankian
- Immunobiochemistry lab, Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Abdolreza Varasteh
- Allergy Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Farhad Salari
- Immunobiochemistry lab, Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mojtaba Meshkat
- Department of Immunology, Zakariya Research Center, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| |
Collapse
|
13
|
Wang YC, Chen CL, Sheu BS, Yang YJ, Tseng PC, Hsieh CY, Lin CF. Helicobacter pylori infection activates Src homology-2 domain-containing phosphatase 2 to suppress IFN-γ signaling. THE JOURNAL OF IMMUNOLOGY 2014; 193:4149-58. [PMID: 25225672 DOI: 10.4049/jimmunol.1400594] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Helicobacter pylori infection not only induces gastric inflammation but also increases the risk of gastric tumorigenesis. IFN-γ has antimicrobial effects; however, H. pylori infection elevates IFN-γ-mediated gastric inflammation and may suppress IFN-γ signaling as a strategy to avoid immune destruction through an as-yet-unknown mechanism. This study was aimed at investigating the mechanism of H. pylori-induced IFN-γ resistance. Postinfection of viable H. pylori decreased IFN-γ-activated signal transducers and activators of transcription 1 and IFN regulatory factor 1 not only in human gastric epithelial MKN45 and AZ-521 but also in human monocytic U937 cells. H. pylori caused an increase in the C-terminal tyrosine phosphorylation of Src homology-2 domain-containing phosphatase (SHP) 2. Pharmacologically and genetically inhibiting SHP2 reversed H. pylori-induced IFN-γ resistance. In contrast to a clinically isolated H. pylori strain HP238, the cytotoxin-associated gene A (CagA) isogenic mutant strain HP238(CagAm) failed to induce IFN-γ resistance, indicating that CagA regulates this effect. Notably, HP238 and HP238(CagAm) differently caused SHP2 phosphorylation; however, imaging and biochemical analyses demonstrated CagA-mediated membrane-associated binding with phosphorylated SHP2. CagA-independent generation of reactive oxygen species (ROS) contributed to H. pylori-induced SHP2 phosphorylation; however, ROS/SHP2 mediated IFN-γ resistance in a CagA-regulated manner. This finding not only provides an alternative mechanism for how CagA and ROS coregulate SHP2 activation but may also explain their roles in H. pylori-induced IFN-γ resistance.
Collapse
Affiliation(s)
- Yu-Chih Wang
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; Department of Microbiology and Immunology, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Chia-Ling Chen
- Center of Translational Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Bor-Shyang Sheu
- Department of Internal Medicine, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Yao-Jong Yang
- Department of Internal Medicine, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Po-Chun Tseng
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Chia-Yuan Hsieh
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Chiou-Feng Lin
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; Department of Microbiology and Immunology, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan 701, Taiwan; and Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
14
|
Luo JJ, Li CY, Liu S, Yu W, Tang SY, Cai HL, Zhang Y. Overexpression of Helicobacter pylori VacA N-terminal fragment induces proinflammatory cytokine expression and apoptosis in human monocytic cell line through activation of NF-κB. Can J Microbiol 2013; 59:523-33. [PMID: 23898995 DOI: 10.1139/cjm-2013-0021] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Vacuolating cytotoxin (VacA) is an important virulence factor in the pathogenesis of Helicobacter pylori-related diseases. The aim of this study was to investigate the function of the amino-terminal 476 residue fragment (p52) of VacA and the possible molecular mechanisms responsible for its induction of proinflammatory cytokines secretion and apoptosis. Human acute monocytic leukemia cell line THP-1 was used as an in vitro model to study proinflammatory cytokines secretion and apoptosis induced by transfection of a recombinant plasmid encoding the amino-terminal 476 residue fragment (p52) of VacA. The results showed that VacA p52 overexpression induced the production of tumor necrosis factor alpha (TNF-α), interleukin-1 beta (IL-1β), nitric oxide, and reactive oxygen species in THP-1 cells in a time-dependent manner. VacA p52 overexpression also promoted THP-1 cells apoptosis. In addition, VacA p52 triggered the activation of nuclear factor kappa B (NF-κB), indicating a possible mechanism for its induction of proinflammatory cytokines secretion and cell apoptosis. Our study demonstrated that the induction of cytokines secretion and apoptosis by VacA p52 in THP-1 cells could be mediated through activation of nuclear factor kappa B.
Collapse
Affiliation(s)
- Jing-Jing Luo
- Institute of Pathogenic Biology, University of South China, Hengyang 421001, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
15
|
Allison CC, Ferrand J, McLeod L, Hassan M, Kaparakis-Liaskos M, Grubman A, Bhathal PS, Dev A, Sievert W, Jenkins BJ, Ferrero RL. Nucleotide oligomerization domain 1 enhances IFN-γ signaling in gastric epithelial cells during Helicobacter pylori infection and exacerbates disease severity. THE JOURNAL OF IMMUNOLOGY 2013; 190:3706-15. [PMID: 23460743 DOI: 10.4049/jimmunol.1200591] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Virulent Helicobacter pylori strains that specifically activate signaling in epithelial cells via the innate immune molecule, nucleotide oligomerization domain 1 (NOD1), are more frequently associated with IFN-γ-dependent inflammation and with severe clinical outcomes (i.e., gastric cancer and peptic ulceration). In cell culture models, we showed that H. pylori activation of the NOD1 pathway caused enhanced proinflammatory signaling in epithelial cells in response to IFN-γ stimulation through the direct effects of H. pylori on two components of the IFN-γ signaling pathway, STAT1 and IFN regulatory factor 1 (IRF1). Specifically, H. pylori activation of the NOD1 pathway was shown to increase the levels of STAT1-Tyr(701)/Ser(727) phosphorylation and IRF1 expression/synthesis in cells, resulting in enhanced production of the NOD1- and IFN-γ-regulated chemokines, IL-8- and IFN-γ-induced protein 10, respectively. Consistent with the notion that heightened proinflammatory signaling in epithelial cells may have an impact on disease severity, we observed significantly increased expression levels of NOD1, CXCL8, IRF1, and CXCL10 in human gastric biopsies displaying severe gastritis, when compared with those without gastritis (p < 0.05, p < 0.001, p < 0.01, and p < 0.05, respectively). Interestingly, NOD1, CXCL8, and IRF1 expression levels were also significantly upregulated in gastric tumor tissues, when compared with paired nontumor samples (p < 0.0001, p < 0.05, and p < 0.05, respectively). Thus, we propose that cross-talk between NOD1 and IFN-γ signaling pathways contribute to H. pylori-induced inflammatory responses, potentially revealing a novel mechanism whereby virulent H. pylori strains promote more severe disease.
Collapse
Affiliation(s)
- Cody C Allison
- Centre for Innate Immunity and Infectious Diseases, Monash Institute of Medical Research, Monash University, Clayton, Victoria 3168, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Regulation of SHP2 by PTEN/AKT/GSK-3β signaling facilitates IFN-γ resistance in hyperproliferating gastric cancer. Immunobiology 2012; 217:926-34. [PMID: 22325465 DOI: 10.1016/j.imbio.2012.01.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2011] [Revised: 12/28/2011] [Accepted: 01/01/2012] [Indexed: 12/16/2022]
Abstract
Oncogenic activation accompanied by escape from immune surveillance, such as IFN-γ resistance, is critical for cancer cell growth and survival. In this study, we investigated the crosstalk signaling between IFN-γ resistance and signaling of hyperproliferation in gastric cancer cells. IFN-γ inhibited the cell growth of MKN45 cells but not hyperproliferating AGS cells. AGS cells did not respond to IFN-γ because of a decrease in STAT1 but not due to dysfunctional IFN-γ receptors. Signaling of PI3K/AKT, as well as MEK/ERK, was required for the hyperproliferation; notably, PI3K/AKT alone mediated the IFN-γ resistance. Aberrant Src homology-2 domain-containing phosphatase (SHP) 2 determined IFN-γ resistance but unexpectedly had no effects on hyperproliferation or ERK activation. In the IFN-γ resistant cells, inactivation of glycogen synthase kinase (GSK)-3β by PI3K/AKT was important for SHP2 activation but not for hyperproliferation. An imbalance of AKT/GSK-3β/SHP2 caused by a reduction of PTEN was important for the crosstalk between IFN-γ resistance and hyperproliferation. PI3K is constitutively expressed in AGS cells and immunohistochemical staining showed a correlation between hyperproliferation and expression of SHP2 and STAT1 in gastric tumors. These results demonstrate the effects of PTEN/AKT/GSK-3β/SHP2 signaling on IFN-γ resistance in hyperproliferating gastric cancer cells.
Collapse
|
17
|
Helicobacter pylori induces expression and secretion of oncostatin M in macrophages in vitro. Dig Dis Sci 2011; 56:689-97. [PMID: 20661773 DOI: 10.1007/s10620-010-1341-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2010] [Accepted: 07/01/2010] [Indexed: 12/09/2022]
Abstract
BACKGROUND Helicobacter pylori is pathogenic bacterium that is associated with several gastric diseases in humans. Disease is characterized by severe inflammatory responses is the stomach that are induced by various chemokines and cytokines. Previous reports indicated that some of these responses are mediated through Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling. METHODS We performed JAK/STAT specific microarrays to identify new components of this signaling pathway, which are affected by Helicobacter pylori infection of THP-1 cells. RESULTS We found that expression and secretion of oncostatin M and of its receptor were strongly up-regulated by Helicobacter pylori. OSM secretion was independent of CagA, VacA or Type IV secretion system. Helicobacter pylori culture supernatant induced OSM secretion. CONCLUSION The induction of the pleiotropic cytokine oncostatin M suggests a possible role in Helicobacter pylori-mediated inflammation and diseases.
Collapse
|
18
|
Bronte-Tinkew DM, Terebiznik M, Franco A, Ang M, Ahn D, Mimuro H, Sasakawa C, Ropeleski MJ, Peek RM, Jones NL. Helicobacter pylori cytotoxin-associated gene A activates the signal transducer and activator of transcription 3 pathway in vitro and in vivo. Cancer Res 2009; 69:632-9. [PMID: 19147578 DOI: 10.1158/0008-5472.can-08-1191] [Citation(s) in RCA: 118] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Persistent infection with Helicobacter pylori confers an increased risk for the development of gastric cancer. However, the exact mechanisms whereby this bacterium causes carcinogenesis have not been completely elucidated. Recent evidence indicates that aberrant activation of the signal transducers and activators of transcription 3 (STAT3) signaling pathway may play a role in gastric carcinogenesis. Therefore, we hypothesized that H. pylori infection modulates STAT3 signaling, favoring gastric cancer development. In epithelial cells infected with H. pylori, STAT3 was activated, as assessed by immunoblotting for phosphorylated STAT3, immunofluorescence of translocated STAT3, fluorescence recovery after photobleaching, and luciferase activation in transfected cells. Activation was dependent on translocation but not phosphorylation of cytotoxin-associated gene A (CagA) in host cells. Activation seemed to be receptor-mediated because preincubation of cells with the interleukin-6 (IL-6) receptor superantagonist sant7 or inhibition of gp130 by a monoclonal antibody prevented H. pylori-mediated STAT3 activation. However, activation was not related to autocrine activation by IL-6 or IL-11. CagA+ wild-type H. pylori, but not the noncarcinogenic cagA- mutant, activated STAT3 in gastric epithelial cells in vivo in the gerbil model of H. pylori-mediated gastric carcinogenesis. Collectively, these results indicate that H. pylori CagA activates the STAT3 signaling pathway in vitro and in vivo, providing a potential mechanism by which chronic H. pylori infection promotes the development of gastric cancer.
Collapse
|
19
|
Young DF, Carlos TS, Hagmaier K, Fan L, Randall RE. AGS and other tissue culture cells can unknowingly be persistently infected with PIV5; a virus that blocks interferon signalling by degrading STAT1. Virology 2007; 365:238-40. [PMID: 17509637 DOI: 10.1016/j.virol.2007.03.061] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2007] [Accepted: 03/27/2007] [Indexed: 10/23/2022]
Abstract
Whilst screening various cell lines for their ability to respond to interferon (IFN), we noted that in comparison to other tissue culture cells AGS tumour cells, which are widely used in biomedical research, had very low levels of STAT1. Subsequent analysis showed that the reason for this is that AGS cells are persistently infected with parainfluenza virus type 5 (PIV5; formally known as SV5), a virus that blocks the interferon (IFN) response by targeting STAT1 for proteasome-mediated degradation. Virus protein expression in AGS is altered in comparison to the normal pattern of virus protein synthesis observed in acutely infected cells, suggesting that the AGS virus is defective. We discuss the relevance of these results in terms of the need to screen cell lines for persistent virus infections that can alter cellular functions.
Collapse
Affiliation(s)
- D F Young
- School of Biology, University of St. Andrews, Fife KY16 9ST, Scotland, UK
| | | | | | | | | |
Collapse
|
20
|
Jandu N, Shen S, Wickham ME, Prajapati R, Finlay BB, Karmali MA, Sherman PM. Multiple seropathotypes of verotoxin-producing Escherichia coli (VTEC) disrupt interferon-γ-induced tyrosine phosphorylation of signal transducer and activator of transcription (Stat)-1. Microb Pathog 2007; 42:62-71. [PMID: 17174521 DOI: 10.1016/j.micpath.2006.10.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2006] [Revised: 10/30/2006] [Accepted: 10/30/2006] [Indexed: 12/22/2022]
Abstract
Verotoxin-producing Escherichia coli (VTEC) O157:H7 inhibits interferon-gamma-stimulated tyrosine phosphorylation of signal transducer and activator of transcription (Stat)-1 in epithelial cells, independent of Verotoxins and the locus of enterocyte effacement pathogenicity island. Although E. coli O157:H7 is the major cause of disease in humans, non-O157:H7 VTEC also cause human disease. However, the virulence properties of non-O157:H7 VTEC are less well characterized. The aims of this study were to define the ability of VTEC strains of differing seropathotypes (classified as A-E) to inhibit interferon-gamma stimulated Stat1-phosphorylation and to further characterize the bacterial-derived inhibitory factor. Confluent T84 and HEp-2 cells were infected with VTEC strains (MOI 100:1, 6h, 37 degrees C), and then stimulated with interferon-gamma (50 ng/mL) for 0.5h at 37 degrees C. Whole-cell protein extracts of infected cells were collected and prepared for immunoblotting to detect tyrosine phosphorylation of Stat1. The effects of E. coli O55 strains, the evolutionary precursors of VTEC, on Stat1-tyrosine phosphorylation were also determined. The effects of isogenic mutants of O-islands 47 and 122 were tested to determine the role of genes encoded on these putative pathogenicity islands in mediating VTEC inhibition of the interferon-gamma-Stat1 signaling cascade. To evaluate potential mechanism(s) of inhibition, VTEC O157:H7-infected cells were treated with pharmacological inhibitors, including, wortmannin and LY294002. Relative to uninfected cells, Stat1-tyrosine phosphorylation was significantly reduced after 6h infection of both T84 and HEp-2 cells by VTEC strains of all five seropathotypes. E. coli O55 strains, but not enteropathogenic E. coli (EPEC), also caused inhibition of Stat1-tyrosine phosphorylation, suggesting that this effect was acquired early in the evolution of VTEC. Stat1-activation did not recover in epithelial cells infected with isogenic mutants of O-islands 47 and 122, indicating that the inhibitory factor was not contained in these genomic regions. Stat1-phosphorylation remained intact when VTEC-infected cells were treated with wortmannin (0-100 nM), but not by treatment with the more specific PI3-kinase inhibitor, LY294002. Inhibition of interferon-gamma stimulated Stat1-tyrosine phosphorylation by VTEC of multiple seropathotypes indicates the presence of a common inhibitory factor that is independent of bacterial virulence in humans. The results of treatment with wortmannin suggest that the bacterial-derived inhibitory factor employs host cell signal transduction to mediate inhibition of Stat1-activation.
Collapse
Affiliation(s)
- Narveen Jandu
- Research Institute, Hospital for Sick Children, Ont., Canada
| | | | | | | | | | | | | |
Collapse
|
21
|
Fedwick JP, Lapointe TK, Meddings JB, Sherman PM, Buret AG. Helicobacter pylori activates myosin light-chain kinase to disrupt claudin-4 and claudin-5 and increase epithelial permeability. Infect Immun 2006; 73:7844-52. [PMID: 16299274 PMCID: PMC1307049 DOI: 10.1128/iai.73.12.7844-7852.2005] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Helicobacter pylori is a spiral, gram-negative bacterium that specifically and persistently infects the human stomach. In some individuals, H. pylori-induced chronic gastritis may progress to gastroduodenal ulcers and gastric cancer. Currently, the host-microbe interactions that determine the clinical outcome of infection are not well defined. H. pylori strains capable of disrupting the gastric epithelial barrier may increase the likelihood of developing serious disease. In this study, H. pylori strain SS1 increased gastric, but not small intestinal, permeability in C57BL/6 mice. H. pylori strain SS1 was able to directly increase paracellular permeability, in the absence of host inflammatory cells, by disrupting the tight-junctional proteins occludin, claudin-4, and claudin-5 in confluent nontransformed epithelial cells. H. pylori SS1 also reduced claudin-4 protein levels in human gastric AGS cells. The ability of H. pylori SS1 to increase permeability appeared to be independent of the well-characterized virulence factors vacuolating cytotoxin and CagA protein. H. pylori activated myosin light-chain kinase in epithelial cells to phosphorylate myosin light chain and increase permeability by disrupting claudin-4 and claudin-5. The bacterial factor responsible for increasing epithelial permeability was heat sensitive, membrane bound, and required apical contact with monolayers. In conclusion, disruptions of the tight junctions observed in this study implicate host cell signaling pathways, including the phosphorylation of myosin light chain and the regulation of tight-junctional proteins claudin-4 and claudin-5, in the pathogenesis of H. pylori infection.
Collapse
Affiliation(s)
- Jason P Fedwick
- Department of Biological Sciences, University of Calgary, 2500 University Drive N.W., Calgary, Alberta, Canada T2N 1N4
| | | | | | | | | |
Collapse
|
22
|
Lingwood C, Mylvaganam M, Minhas F, Binnington B, Branch DR, Pomès R. The Sulfogalactose Moiety of Sulfoglycosphingolipids Serves as a Mimic of Tyrosine Phosphate in Many Recognition Processes. J Biol Chem 2005; 280:12542-7. [PMID: 15634687 DOI: 10.1074/jbc.m413724200] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Multiple ligand co-recognition of 3'-sulfogalactosylceramide (SGC) and sulfotyrosine initiated the comparison of SGC and sulfotyrosine and, subsequently, phosphotyrosine (pY) binding. SGC is a receptor for ligands involved in cell adhesion/microbial pathology. pY forms a Src homology domain 2 recognition motif in intracellular signaling. Using hsp70, anti-SGC, and anti-pY antibodies, ligand binding is retained following phosphate/sulfate and tyrosine/galactose substitution in SGC and sulfate/phosphate exchange in pY. Remarkable lipid-dependent binding to phosphatidylethanolamine-conjugated sulfotyrosine suggests "microenvironmental" modulation of sulfotyrosine-containing receptors, similar to glycosphingolipids. Based on an aryl substrate-bound co-crystal of arylsulfatase A, a sulfogalactose and phosphotyrosine esterase, modeling provides a solvation basis for co-recognition. c-Src/Src homology domain 2:SGC/phosphogalactosylceramide binding confirms our hypothesis, heralding a carbohydrate-based approach to regulation of phosphotyrosine-mediated recognition.
Collapse
Affiliation(s)
- Clifford Lingwood
- Research Institute, The Hospital for Sick Children, Toronto, Ontario M4G 1X8, Canada.
| | | | | | | | | | | |
Collapse
|
23
|
Abstract
Research in the last year has provided new insights into the function of the the cag-associated type IV secretion system and the vacuolating toxin VacA. A quite new aspect was disclosed by the finding that Helicobacter pylori in Mongolian gerbils colonizes a very distinct topology in the gastric mucous layer, obviously providing optimal conditions for long-term survival. Further research activities focused on H. pylori ammonia and metal metabolism as well as on bacterial stress defence mechanisms. Differential expression of approximately 7% of the bacterial genome was found at low pH suggesting that H. pylori has evolved a multitude of acid-adaptive mechanisms. VacA was shown to interrupt phagosome maturation in macrophage cell lines as well as to modulate and interfere with T lymphocyte immunological functions. Gastric mucosa as well as the H. pylori-infected epithelial cell line AGS strongly express IL-8 receptor A and B, which might contribute to the augmentation of the inflammatory response. Accumulating evidence implicates genetic variation in the inflammatory response to H. pylori in the etiology of the increased risk of gastric cancer after H. pylori infection. The chronic imbalance between apoptosis and cell proliferation is the first step of gastric carcinogenesis. In this regard, it was demonstrated that coexpression of two H. pylori proteins, CagA and HspB, in AGS cells, caused an increase in E2F transcription factor, cyclin D3, and phosphorylated retinoblastoma protein. Taken together, we now have a better understanding of the role of different virulence factors of H. pylori. There is still a lot to be learned, but the promising discoveries summarized here, demonstrate that the investigation of the bacterial survival strategies will give novel insights into pathogenesis and disease development.
Collapse
Affiliation(s)
- Paul Hofman
- INSERM 0215 and Laboratory of Clinical and Experimental Pathology, Faculty of Medicine and Pasteur Hospital, University of Nice, 06002 Nice, France.
| | | | | | | | | | | |
Collapse
|