1
|
Naumann M, Ferino L, Sharafutdinov I, Backert S. Gastric Epithelial Barrier Disruption, Inflammation and Oncogenic Signal Transduction by Helicobacter pylori. Curr Top Microbiol Immunol 2023; 444:207-238. [PMID: 38231220 DOI: 10.1007/978-3-031-47331-9_8] [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: 01/18/2024]
Abstract
Helicobacter pylori exemplifies one of the most favourable bacterial pathogens worldwide. The bacterium colonizes the gastric mucosa in about half of the human population and constitutes a major risk factor for triggering gastric diseases such as stomach cancer. H. pylori infection represents a prime example of chronic inflammation and cancer-inducing bacterial pathogens. The microbe utilizes a remarkable set of virulence factors and strategies to control cellular checkpoints of inflammation and oncogenic signal transduction. This chapter emphasizes on the pathogenicity determinants of H. pylori such as the cytotoxin-associated genes pathogenicity island (cagPAI)-encoded type-IV secretion system (T4SS), effector protein CagA, lipopolysaccharide (LPS) metabolite ADP-glycero-β-D-manno-heptose (ADP-heptose), cytotoxin VacA, serine protease HtrA, and urease, and how they manipulate various key host cell signaling networks in the gastric epithelium. In particular, we highlight the H. pylori-induced disruption of cell-to-cell junctions, pro-inflammatory activities, as well as proliferative, pro-apoptotic and anti-apoptotic responses. Here we review these hijacked signal transduction events and their impact on gastric disease development.
Collapse
Affiliation(s)
- Michael Naumann
- Institute of Experimental Internal Medicine, Medical Faculty, Otto Von Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany.
| | - Lorena Ferino
- Institute of Experimental Internal Medicine, Medical Faculty, Otto Von Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Irshad Sharafutdinov
- Dept. Biology, Division of Microbiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Staudtstr. 5, 91058, Erlangen, Germany
| | - Steffen Backert
- Dept. Biology, Division of Microbiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Staudtstr. 5, 91058, Erlangen, Germany.
| |
Collapse
|
2
|
Aguilar-Rojas A, Olivo-Marin JC, Guillen N. Human intestinal models to study interactions between intestine and microbes. Open Biol 2020; 10:200199. [PMID: 33081633 PMCID: PMC7653360 DOI: 10.1098/rsob.200199] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 09/24/2020] [Indexed: 12/12/2022] Open
Abstract
Implementations of suitable in vitro cell culture systems of the human intestine have been essential tools in the study of the interaction among organs, commensal microbiota, pathogens and parasites. Due to the great complexity exhibited by the intestinal tissue, researchers have been developing in vitro/ex vivo systems to diminish the gap between conventional cell culture models and the human intestine. These models are able to reproduce different structures and functional aspects of the tissue. In the present review, information is recapitulated on the most used models, such as cell culture, intestinal organoids, scaffold-based three-dimensional models, and organ-on-a-chip and their use in studying the interaction between human intestine and microbes, and their advantages and limitations are also discussed.
Collapse
Affiliation(s)
- Arturo Aguilar-Rojas
- Instituto Mexicano del Seguro Social, Unidad de Investigación Médica en Medicina Reproductiva, Unidad Médica de Alta Especialidad en Ginecología y Obstetricia No. 4 ‘Dr. Luis Castelazo Ayala’, Av. Río Magdalena No. 289, Col. Tizapán San Ángel, C.P. 01090 Ciudad de México, México
- Institut Pasteur, Unité d'Analyse d'Images Biologiques, 25 Rue du Dr Roux, 75015 Paris, France
| | - Jean-Christophe Olivo-Marin
- Institut Pasteur, Unité d'Analyse d'Images Biologiques, 25 Rue du Dr Roux, 75015 Paris, France
- Centre National de la Recherche Scientifique, UMR3691, 25 Rue du Dr Roux, 75015 Paris, France
| | - Nancy Guillen
- Institut Pasteur, Unité d'Analyse d'Images Biologiques, 25 Rue du Dr Roux, 75015 Paris, France
- Centre National de la Recherche Scientifique, ERL9195, 25 Rue du Dr Roux, 75015 Paris, France
| |
Collapse
|
3
|
Natarajan V, Moar P, Kaur US, Venkatesh V, Kumar A, Chaturvedi R, Himanshu D, Tandon R. Helicobacter pylori Reactivates Human Immunodeficiency Virus-1 in Latently Infected Monocytes with Increased Expression of IL-1β and CXCL8. Curr Genomics 2020; 20:556-568. [PMID: 32581644 PMCID: PMC7290055 DOI: 10.2174/1389202921666191226091138] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 12/08/2019] [Accepted: 12/08/2019] [Indexed: 12/13/2022] Open
Abstract
Background Helicobacter pylori are gram-negative bacteria, which colonize the human stomach. More than 50% of the world's population is infected by H. pylori. Based on the high prevalence of H. pylori, it is very likely that HIV and H. pylori infection may coexist. However, the molecular events that occur during HIV-H. pylori co-infection remain unclear. Latent HIV reservoirs are the major obstacle in HIV cure despite effective therapy. Here, we explored the effect of H. pylori stimulation on latently HIV-infected monocytic cell line U1. Methods High throughput RNA-Seq using Illumina platform was performed to analyse the change in transcriptome between unstimulated and H. pylori-stimulated latently HIV-infected U1 cells. Transcriptome analysis identified potential genes and pathways involved in the reversal of HIV latency using bioinformatic tools that were validated by real-time PCR. Results H. pylori stimulation increased the expression of HIV-1 Gag, both at transcription (p<0.001) and protein level. H. pylori stimulation also increased the expression of proinflammatory cytokines IL-1β, CXCL8 and CXCL10 (p<0.0001). Heat-killed H. pylori retained their ability to induce HIV transcription. RNA-Seq analysis revealed 197 significantly upregulated and 101 significantly downregulated genes in H. pylori-stimulated U1 cells. IL-1β and CXCL8 were found to be significantly upregulated using transcriptome analysis, which was consistent with real-time PCR data. Conclusion H. pylori reactivate HIV-1 in latently infected monocytes with the upregulation of IL-1β and CXCL8, which are prominent cytokines involved in the majority of inflammatory pathways. Our results warrant future in vivo studies elucidating the effect of H. pylori in HIV latency and pathogenesis.
Collapse
Affiliation(s)
- Vidhya Natarajan
- 1Laboratory of AIDS Research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India; 2Department of Microbiology, King Georges Medical University, Lucknow, India; 3Institute of Bioinformatics, International Technology Park, Bangaluru, 560066, India; 4Manipal Academy of Higher Education (MAHE), Manipal576104, Karnataka, India; 5Host Pathogen Interaction Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India; 6Department of Medicine, King Georges Medical University, Lucknow, India
| | - Preeti Moar
- 1Laboratory of AIDS Research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India; 2Department of Microbiology, King Georges Medical University, Lucknow, India; 3Institute of Bioinformatics, International Technology Park, Bangaluru, 560066, India; 4Manipal Academy of Higher Education (MAHE), Manipal576104, Karnataka, India; 5Host Pathogen Interaction Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India; 6Department of Medicine, King Georges Medical University, Lucknow, India
| | - Urvinder S Kaur
- 1Laboratory of AIDS Research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India; 2Department of Microbiology, King Georges Medical University, Lucknow, India; 3Institute of Bioinformatics, International Technology Park, Bangaluru, 560066, India; 4Manipal Academy of Higher Education (MAHE), Manipal576104, Karnataka, India; 5Host Pathogen Interaction Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India; 6Department of Medicine, King Georges Medical University, Lucknow, India
| | - Vimala Venkatesh
- 1Laboratory of AIDS Research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India; 2Department of Microbiology, King Georges Medical University, Lucknow, India; 3Institute of Bioinformatics, International Technology Park, Bangaluru, 560066, India; 4Manipal Academy of Higher Education (MAHE), Manipal576104, Karnataka, India; 5Host Pathogen Interaction Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India; 6Department of Medicine, King Georges Medical University, Lucknow, India
| | - Abhishek Kumar
- 1Laboratory of AIDS Research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India; 2Department of Microbiology, King Georges Medical University, Lucknow, India; 3Institute of Bioinformatics, International Technology Park, Bangaluru, 560066, India; 4Manipal Academy of Higher Education (MAHE), Manipal576104, Karnataka, India; 5Host Pathogen Interaction Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India; 6Department of Medicine, King Georges Medical University, Lucknow, India
| | - Rupesh Chaturvedi
- 1Laboratory of AIDS Research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India; 2Department of Microbiology, King Georges Medical University, Lucknow, India; 3Institute of Bioinformatics, International Technology Park, Bangaluru, 560066, India; 4Manipal Academy of Higher Education (MAHE), Manipal576104, Karnataka, India; 5Host Pathogen Interaction Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India; 6Department of Medicine, King Georges Medical University, Lucknow, India
| | - D Himanshu
- 1Laboratory of AIDS Research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India; 2Department of Microbiology, King Georges Medical University, Lucknow, India; 3Institute of Bioinformatics, International Technology Park, Bangaluru, 560066, India; 4Manipal Academy of Higher Education (MAHE), Manipal576104, Karnataka, India; 5Host Pathogen Interaction Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India; 6Department of Medicine, King Georges Medical University, Lucknow, India
| | - Ravi Tandon
- 1Laboratory of AIDS Research and Immunology, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India; 2Department of Microbiology, King Georges Medical University, Lucknow, India; 3Institute of Bioinformatics, International Technology Park, Bangaluru, 560066, India; 4Manipal Academy of Higher Education (MAHE), Manipal576104, Karnataka, India; 5Host Pathogen Interaction Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi, India; 6Department of Medicine, King Georges Medical University, Lucknow, India
| |
Collapse
|
4
|
Atnip A, Giusti MM, Sigurdson GT, Failla ML, Chitchumroonchokchai C, Bomser JA. The NCI-N87 Cell Line as a Gastric Epithelial Model to Study Cellular Uptake, Trans-Epithelial Transport, and Gastric Anti-Inflammatory Properties of Anthocyanins. Nutr Cancer 2019; 72:686-695. [PMID: 31353956 DOI: 10.1080/01635581.2019.1644354] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Anthocyanins are ubiquitous plant pigments with reported antioxidant, anti-inflammatory, and anti-cancer activities. To better understand these benefits, metabolism of anthocyanins requires further evaluation, especially in the stomach. Mammalian cell cultures provide useful models for investigating compound metabolism and absorption, but they are generally maintained at physiological pH. The NCI-N87 cell line is an acid-stable model of the gastric epithelium used to study gastric drug metabolism. The objective of this work was to investigate the uptake, trans-epithelial transport, and anti-inflammatory activity of anthocyanins by the NCI-N87 cell line. The cells formed a coherent monolayer, stable ≤32 days post confluency. Minimal effects on monolayer integrity were observed when the pH of the apical chamber was adjusted to pH 3.0, 5.0, or 7.4. Anthocyanins were transported across the NCI-N87 cell monolayer at 37 °C, but not at 0 °C, suggesting a facilitated process. Chokeberry anthocyanins (0-1500 μM) were not cytotoxic. At apical pH 3.0, they had anti-inflammatory properties by significantly attenuating IL-8 secretion when added to medium before, during, and after incubation with IL-1β. These results suggest that the NCI-N87 cell line is a physiologically relevant model for in vitro studies of the transport, anti-inflammatory and potential anti-carcinogenic activities of anthocyanins in gastric tissue.
Collapse
Affiliation(s)
- Allison Atnip
- Department of Food Science and Technology, The Ohio State University, Columbus, OH, USA
| | - M Mónica Giusti
- Department of Food Science and Technology, The Ohio State University, Columbus, OH, USA
| | - Gregory T Sigurdson
- Department of Food Science and Technology, The Ohio State University, Columbus, OH, USA
| | - Mark L Failla
- Department of Human Sciences, The Ohio State University, Columbus, OH, USA
| | | | - Joshua A Bomser
- Department of Human Sciences, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
5
|
Zhang Y, Li D, Dai Y, Li R, Gao Y, Hu L. The Role of E-cadherin in Helicobacter pylori-Related Gastric Diseases. Curr Drug Metab 2018; 20:23-28. [PMID: 29938616 DOI: 10.2174/1389200219666180625113010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 04/09/2018] [Accepted: 04/17/2018] [Indexed: 12/29/2022]
Abstract
BACKGROUND Helicobacter pylori (H. pylori)-related gastric diseases are a series of gastric mucosal disorders associated with H. pylori infection. Gastric cancer (GC) is widely believed to evolve from gastritis and gastric ulcer. As an important adhesion molecule of epithelial cells, E-cadherin plays a key role in the development of gastric diseases. In this review, we aim to seek the characteristic of E-cadherin expression at different stages of gastric diseases. METHODS We searched plenty of databases for research literature about E-cadherin expression in H. pylori-related gastric diseases, and reviewed the relationship of E-cadherin and H. pylori, and the role of E-cadherin at different stages of gastric diseases. RESULTS H. pylori was shown to decrease E-cadherin expression by various ways in vitro, while most of clinical studies have not found the relationship between H. pylori and E-cadherin expression. It is defined that poor outcome of GC is related to loss expression of E-cadherin, but it is still unclear when qualitative change of E-cadherin expression in gastric mucosa emerges. CONCLUSION Expression level of E-cadherin in gastric cells may be a consequence of injury factors and body's selfrepairing ability. More studies on E-cadherin expression in gastric mucosa with precancerous lesions need to be performed, which may be potential and useful for early detection, prevention and treatment of GC.
Collapse
Affiliation(s)
- Yunzhan Zhang
- Piwei Institute, Guangzhou University of Chinese Medicine, Guangdong, China
| | - Danyan Li
- Piwei Institute, Guangzhou University of Chinese Medicine, Guangdong, China
| | - Yunkai Dai
- Piwei Institute, Guangzhou University of Chinese Medicine, Guangdong, China
| | - Ruliu Li
- Piwei Institute, Guangzhou University of Chinese Medicine, Guangdong, China
| | - Yong Gao
- Piwei Institute, Guangzhou University of Chinese Medicine, Guangdong, China
| | - Ling Hu
- Piwei Institute, Guangzhou University of Chinese Medicine, Guangdong, China
| |
Collapse
|
6
|
Pandey S, Jha HC, Shukla SK, Shirley MK, Robertson ES. Epigenetic Regulation of Tumor Suppressors by Helicobacter pylori Enhances EBV-Induced Proliferation of Gastric Epithelial Cells. mBio 2018; 9:e00649-18. [PMID: 29691341 PMCID: PMC5915740 DOI: 10.1128/mbio.00649-18] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 03/27/2018] [Indexed: 12/11/2022] Open
Abstract
Helicobacter pylori and Epstein-Barr virus (EBV) are two well-known contributors to cancer and can establish lifelong persistent infection in the host. This leads to chronic inflammation, which also contributes to development of cancer. Association with H. pylori increases the risk of gastric carcinoma, and coexistence with EBV enhances proliferation of infected cells. Further, H. pylori-EBV coinfection causes chronic inflammation in pediatric patients. We have established an H. pylori-EBV coinfection model system using human gastric epithelial cells. We showed that H. pylori infection can increase the oncogenic phenotype of EBV-infected cells and that the cytotoxin-associated gene (CagA) protein encoded by H. pylori stimulated EBV-mediated cell proliferation in this coinfection model system. This led to increased expression of DNA methyl transferases (DNMTs), which reprogrammed cellular transcriptional profiles, including those of tumor suppressor genes (TSGs), through hypermethylation. These findings provide new insights into a molecular mechanism whereby cooperativity between two oncogenic agents leads to enhanced oncogenic activity of gastric cancer cells.IMPORTANCE We have studied the cooperativity between H. pylori and EBV, two known oncogenic agents. This led to an enhanced oncogenic phenotype in gastric epithelial cells. We now demonstrate that EBV-driven epigenetic modifications are enhanced in the presence of H. pylori, more specifically, in the presence of its CagA secretory antigen. This results in increased proliferation of the infected gastric cells. Our findings now elucidate a molecular mechanism whereby expression of cellular DNA methyl transferases is induced influencing infection by EBV. Hypermethylation of the regulatory genomic regions of tumor suppressor genes results in their silencing. This drastically affects the expression of cell cycle, apoptosis, and DNA repair genes, which dysregulates their associated processes, and promotion of the oncogenic phenotype.
Collapse
Affiliation(s)
- Saurabh Pandey
- Departments of Otorhinolaryngology-Head and Neck Surgery, and Microbiology, the Tumor Virology Program, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Hem Chandra Jha
- Departments of Otorhinolaryngology-Head and Neck Surgery, and Microbiology, the Tumor Virology Program, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sanket Kumar Shukla
- Departments of Otorhinolaryngology-Head and Neck Surgery, and Microbiology, the Tumor Virology Program, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Meghan K Shirley
- Departments of Otorhinolaryngology-Head and Neck Surgery, and Microbiology, the Tumor Virology Program, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Erle S Robertson
- Departments of Otorhinolaryngology-Head and Neck Surgery, and Microbiology, the Tumor Virology Program, Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
7
|
Zhao K, Xie Q, Xu D, Guo Y, Tao X, Wei H, Wan C. Antagonistics of Lactobacillus plantarum ZDY2013 against Helicobacter pylori SS1 and its infection in vitro in human gastric epithelial AGS cells. J Biosci Bioeng 2018; 126:458-463. [PMID: 29699944 DOI: 10.1016/j.jbiosc.2018.04.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Revised: 03/28/2018] [Accepted: 04/03/2018] [Indexed: 12/12/2022]
Abstract
In this study, the anti-Helicobacterpylori activity of Lactobacillusplantarum ZDY2013 was investigated and Lactobacillusrhamnosus GG was used as a positive control. The anti-H. pylori mechanism in vitro was also examined. Results revealed that either the viable cells or supernatant of L. plantarum ZDY2013 could suppress the growth or urease activity of H. pylori. The inhibitory effects of L. plantarum ZDY2013 were relatively higher than those of L. rhamnosus GG (P < 0.05), and such effects might be a result of their lactic acid production (e.g., 51.105 ± 0.097 mmol/L for L. plantarum ZDY2013 and 33.113 ± 0.063 mmol/L for L. rhamnosus GG). The anti-adhesion capacity of L. plantarum ZDY2013 against H. pylori was also stronger than that of L. rhamnosus GG in terms of inhibition, competition, and displacement. Among these inhibitory strategies, competition exhibited the best performance, with an inhibition ratio of 92.65%. Upon inhibition and anti-adhesion, the cells and supernatant of L. plantarum ZDY2013 significantly strengthened the expression of the anti-inflammatory cytokine IL-10, but attenuated the expression of the pro-inflammatory cytokine TNF-α in AGS cells induced by H. pylori SS1. Remarkably, the supernatant of ZDY2013 achieved a relatively higher anti-inflammatory effect than that exerted by its cells. With excellent lactic acid yield and antagonistic and anti-inflammatory effects against H. pylori SS1 infection, L. plantarum ZDY2013 shows potential to be used as a probiotics candidate.
Collapse
Affiliation(s)
- Kui Zhao
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, PR China
| | - Qiong Xie
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, PR China
| | - Di Xu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, PR China
| | - Yilin Guo
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, PR China
| | - Xueying Tao
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, PR China
| | - Hua Wei
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang 330047, PR China
| | - Cuixiang Wan
- Sino-German Joint Research Institute, Nanchang University, Nanchang 330047, PR China.
| |
Collapse
|
8
|
The Middle Fragment of Helicobacter pylori CagA Induces Actin Rearrangement and Triggers Its Own Uptake into Gastric Epithelial Cells. Toxins (Basel) 2017; 9:toxins9080237. [PMID: 28788072 PMCID: PMC5577571 DOI: 10.3390/toxins9080237] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 07/07/2017] [Accepted: 07/26/2017] [Indexed: 01/14/2023] Open
Abstract
Cytotoxin-associated gene product A (CagA) is a major virulence factor secreted by Helicobacter pylori. CagA activity in the gastric epithelium is associated with higher risk of gastric cancer development. Bacterial type IV secretion system (T4SS)-mediated translocation of CagA into the cytosol of human epithelial cells occurs via a poorly understood mechanism that requires CagA interaction with the host membrane lipid phosphatidylserine (PS) and host cell receptor integrin α5β1. Here we have characterized the isolated recombinant middle fragment of CagA (CagA-M) that contains the positively-charged PS-binding region (aa 613–636) and a putative β1 integrin binding site, but lacks the EPIYA region, secretion signal peptide and the CagA multimerization motif. We show that CagA-M, when immobilized on latex beads, is capable of binding to, and triggering its own uptake into, gastric epithelial cells in the absence of infection with cagA-positive H. pylori. Using site-directed mutagenesis, fluorescent and electron microscopy, and highly-specific inhibitors, we demonstrate that the cell-binding and endocytosis-like internalization of CagA-M are dependent on (1) binding to PS; (2) β1 integrin activity; and (3) actin dynamics. Interaction of CagA-M with the host cells is accompanied by the development of long filopodia-like protrusions (macrospikes). This novel morphology is different from the hummingbird phenotype induced by the translocation of full-length CagA. The determinants within CagA-M and within the host that are important for endocytosis-like internalization into host cells are very similar to those observed for T4SS-mediated internalization of full-length CagA, suggesting that the latter may involve an endocytic pathway.
Collapse
|
9
|
Backert S, Schmidt TP, Harrer A, Wessler S. Exploiting the Gastric Epithelial Barrier: Helicobacter pylori's Attack on Tight and Adherens Junctions. Curr Top Microbiol Immunol 2017; 400:195-226. [PMID: 28124155 DOI: 10.1007/978-3-319-50520-6_9] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Highly organized intercellular tight and adherens junctions are crucial structural components for establishing and maintenance of epithelial barrier functions, which control the microbiota and protect against intruding pathogens in humans. Alterations in these complexes represent key events in the development and progression of multiple infectious diseases as well as various cancers. The gastric pathogen Helicobacter pylori exerts an amazing set of strategies to manipulate these epithelial cell-to-cell junctions, which are implicated in changing cell polarity, migration and invasive growth as well as pro-inflammatory and proliferative responses. This chapter focuses on the H. pylori pathogenicity factors VacA, CagA, HtrA and urease, and how they can induce host cell signaling involved in altering cell-to-cell permeability. We propose a stepwise model for how H. pylori targets components of tight and adherens junctions in order to disrupt the gastric epithelial cell layer, giving fresh insights into the pathogenesis of this important bacterium.
Collapse
Affiliation(s)
- Steffen Backert
- Division of Microbiology, Department of Biology, Friedrich Alexander University Erlangen-Nuremberg, Staudtstr. 5, 91058, Erlangen, Germany.
| | - Thomas P Schmidt
- Division of Microbiology, Department of Molecular Biology, Paris-Lodron University of Salzburg, Billroth Str. 11, 5020, Salzburg, Austria
| | - Aileen Harrer
- Division of Microbiology, Department of Biology, Friedrich Alexander University Erlangen-Nuremberg, Staudtstr. 5, 91058, Erlangen, Germany
| | - Silja Wessler
- Division of Microbiology, Department of Molecular Biology, Paris-Lodron University of Salzburg, Billroth Str. 11, 5020, Salzburg, Austria.
| |
Collapse
|
10
|
Bessède E, Molina S, Amador LA, Dubus P, Staedel C, Chambonnier L, Buissonnière A, Sifré E, Giese A, Bénéjat L, Rousseau B, Costet P, Sacks DB, Mégraud F, Varon C. Deletion of IQGAP1 promotes Helicobacter pylori-induced gastric dysplasia in mice and acquisition of cancer stem cell properties in vitro. Oncotarget 2016; 7:80688-80699. [PMID: 27729612 PMCID: PMC5340252 DOI: 10.18632/oncotarget.12486] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 07/18/2016] [Indexed: 02/06/2023] Open
Abstract
Helicobacter pylori infection is responsible for gastric carcinogenesis but host factors are also implicated. IQGAP1, a scaffolding protein of the adherens junctions interacting with E-cadherin, regulates cellular plasticity and proliferation. In mice, IQGAP1 deficiency leads to gastric hyperplasia. The aim of this study was to elucidate the consequences of IQGAP1 deletion on H. pylori-induced gastric carcinogenesis.Transgenic mice deleted for iqgap1 and WT littermates were infected with Helicobacter sp., and histopathological analyses of the gastric mucosa were performed. IQGAP1 and E-cadherin expression was evaluated in gastric tissues and in gastric epithelial cell lines in response to H. pylori infection. The consequences of IQGAP1 deletion on gastric epithelial cell behaviour and on the acquisition of cancer stem cell (CSC)-like properties were evaluated. After one year of infection, iqgap1+/- mice developed more preneoplastic lesions and up to 8 times more gastro-intestinal neoplasia (GIN) than WT littermates. H. pylori infection induced IQGAP1 and E-cadherin delocalization from cell-cell junctions. In vitro, knock-down of IQGAP1 favoured the acquisition of a mesenchymal phenotype and CSC-like properties induced by H. pylori infection.Our results indicate that alterations in IQGAP1 signalling promote the emergence of CSCs and gastric adenocarcinoma development in the context of an H. pylori infection.
Collapse
Affiliation(s)
- Emilie Bessède
- Bacteriology Laboratory, University of Bordeaux, Bordeaux, France
- INSERM, U853, Bordeaux, France
| | - Silvia Molina
- Bacteriology Laboratory, University of Bordeaux, Bordeaux, France
- INSERM, U853, Bordeaux, France
| | - Luis Acuña Amador
- Bacteriology Laboratory, University of Bordeaux, Bordeaux, France
- INSERM, U853, Bordeaux, France
| | - Pierre Dubus
- EA2406 Histologie et pathologie moléculaire des tumeurs, University of Bordeaux, Bordeaux, France
| | - Cathy Staedel
- ‘RNA: Natural and Artificial Regulation’ (ARNA) Laboratory, University of Bordeaux, Bordeaux, France
- INSERM, U869, Bordeaux, France
| | - Lucie Chambonnier
- Bacteriology Laboratory, University of Bordeaux, Bordeaux, France
- INSERM, U853, Bordeaux, France
| | - Alice Buissonnière
- Bacteriology Laboratory, University of Bordeaux, Bordeaux, France
- INSERM, U853, Bordeaux, France
| | - Elodie Sifré
- Bacteriology Laboratory, University of Bordeaux, Bordeaux, France
- INSERM, U853, Bordeaux, France
| | - Alban Giese
- EA2406 Histologie et pathologie moléculaire des tumeurs, University of Bordeaux, Bordeaux, France
- Experimental Pathology Platform, SIRIC BRIO, University of Bordeaux, Bordeaux, France
| | - Lucie Bénéjat
- Bacteriology Laboratory, University of Bordeaux, Bordeaux, France
- INSERM, U853, Bordeaux, France
| | - Benoît Rousseau
- Service Commun des Animaleries, Animalerie A2, University of Bordeaux, Bordeaux, France
| | - Pierre Costet
- Service Commun des Animaleries, Animalerie Transgénique, University of Bordeaux, Bordeaux, France
| | - David B. Sacks
- Department of Laboratory Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Francis Mégraud
- Bacteriology Laboratory, University of Bordeaux, Bordeaux, France
- INSERM, U853, Bordeaux, France
| | - Christine Varon
- Bacteriology Laboratory, University of Bordeaux, Bordeaux, France
- INSERM, U853, Bordeaux, France
| |
Collapse
|
11
|
Brücher BLDM, Li Y, Schnabel P, Daumer M, Wallace TJ, Kube R, Zilberstein B, Steele S, Voskuil JLA, Jamall IS. Genomics, microRNA, epigenetics, and proteomics for future diagnosis, treatment and monitoring response in upper GI cancers. Clin Transl Med 2016; 5:13. [PMID: 27053248 PMCID: PMC4823224 DOI: 10.1186/s40169-016-0093-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2016] [Accepted: 03/29/2016] [Indexed: 12/15/2022] Open
Abstract
One major objective for our evolving understanding in the treatment of cancers will be to address how a combination of diagnosis and treatment strategies can be used to integrate patient and tumor variables with an outcome-oriented approach. Such an approach, in a multimodal therapy setting, could identify those patients (1) who should undergo a defined treatment (personalized therapy) (2) in whom modifications of the multimodal therapy due to observed responses might lead to an improvement of the response and/or prognosis (individualized therapy), (3) who might not benefit from a particular toxic treatment regimen, and (4) who could be identified early on and thereby be spared the morbidity associated with such treatments. These strategies could lead in the direction of precision medicine and there is hope of integrating translational molecular data to improve cancer classifications. In order to achieve these goals, it is necessary to understand the key issues in different aspects of biotechnology to anticipate future directions of personalized and individualized diagnosis and multimodal treatment strategies. Providing an overview of translational data in cancers proved to be a challenge as different methods and techniques used to obtain molecular data are used and studies are based on different tumor entities with different tumor biology and prognoses as well as vastly different therapeutic approaches. The pros and cons of the available methodologies and the potential response data in genomics, microRNA, epigenetics and proteomics with a focus on upper gastrointestinal cancers are considered herein to allow for an understanding of where these technologies stand with respect to cancer diagnosis, prognosis and treatment.
Collapse
Affiliation(s)
- Björn L. D. M. Brücher
- />Theodor-Billroth-Academy®, Munich, Germany
- />Theodor-Billroth-Academy®, Sacramento, CA USA
- />INCORE, International Consortium of Research Excellence of the Theodor-Billroth-Academy®, Munich, Germany
- />INCORE, International Consortium of Research Excellence of the Theodor-Billroth-Academy®, Sacramento, CA USA
- />Bon Secours Cancer Institute, Richmond, VA USA
- />Department of Surgery, Carl-Thiem-Klinikum, Cottbus, Germany
| | - Yan Li
- />Proteogenomics Research Institute for Systems Medicine, San Diego, CA USA
| | - Philipp Schnabel
- />Institute of Pathology, University of Homburg Saar, Homburg, Germany
| | - Martin Daumer
- />Theodor-Billroth-Academy®, Munich, Germany
- />Theodor-Billroth-Academy®, Sacramento, CA USA
- />INCORE, International Consortium of Research Excellence of the Theodor-Billroth-Academy®, Munich, Germany
- />INCORE, International Consortium of Research Excellence of the Theodor-Billroth-Academy®, Sacramento, CA USA
- />Sylvia Lawry Center for MS Research, Munich, Germany
| | | | - Rainer Kube
- />Department of Surgery, Carl-Thiem-Klinikum, Cottbus, Germany
| | | | - Scott Steele
- />Case Western Reserve University, Cleveland, OH USA
- />Department of Surgery, Madigan Army Medical Center, Tacoma, WA USA
| | | | - Ijaz S. Jamall
- />Theodor-Billroth-Academy®, Munich, Germany
- />Theodor-Billroth-Academy®, Sacramento, CA USA
- />INCORE, International Consortium of Research Excellence of the Theodor-Billroth-Academy®, Munich, Germany
- />INCORE, International Consortium of Research Excellence of the Theodor-Billroth-Academy®, Sacramento, CA USA
- />Risk-Based Decisions, Inc., Sacramento, CA USA
| |
Collapse
|
12
|
Garay J, Piazuelo MB, Majumdar S, Li L, Trillo-Tinoco J, Del Valle L, Schneider BG, Delgado AG, Wilson KT, Correa P, Zabaleta J. The homing receptor CD44 is involved in the progression of precancerous gastric lesions in patients infected with Helicobacter pylori and in development of mucous metaplasia in mice. Cancer Lett 2016; 371:90-8. [PMID: 26639196 PMCID: PMC4714604 DOI: 10.1016/j.canlet.2015.10.037] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 09/09/2015] [Accepted: 10/05/2015] [Indexed: 01/06/2023]
Abstract
Infection with Helicobacter pylori (H. pylori) leads to inflammatory events that can promote gastric cancer development. Immune cells transition from the circulation into the infected mucosa through the interaction of their receptors and ligands in the endothelial compartment. CD44 expression is increased in advanced gastric lesions. However, the association of this molecule with the progression of these lesions over time has not been investigated. In addition, there is a lack of understanding of the CD44-dependent cellular processes that lead to gastritis, and possibly to gastric cancer. Here we studied H. pylori-positive subjects with gastric lesions that ranged from multifocal atrophic gastritis to dysplasia to determine gene expression changes associated with disease progression over a period of 6 years. We report that CD44 expression is significantly increased in individuals whose gastric lesions progressed along the gastric precancerous cascade. We also show that CD44-/- mice develop less severe and less extensive H. pylori-induced metaplasia, and show fewer infiltrating Gr1+ cells compared to wild type mice. We present data suggesting that CD44 is associated with disease progression. Mechanisms associated with these effects include induction of interferon gamma responses.
Collapse
MESH Headings
- Animals
- Antigens, Ly/metabolism
- Cells, Cultured
- Chemotaxis, Leukocyte
- Disease Models, Animal
- Disease Progression
- Female
- Gastric Mucosa/immunology
- Gastric Mucosa/metabolism
- Gastric Mucosa/microbiology
- Gastric Mucosa/pathology
- Gastritis, Atrophic/diagnosis
- Gastritis, Atrophic/genetics
- Gastritis, Atrophic/immunology
- Gastritis, Atrophic/metabolism
- Gastritis, Atrophic/microbiology
- Helicobacter Infections/diagnosis
- Helicobacter Infections/genetics
- Helicobacter Infections/immunology
- Helicobacter Infections/metabolism
- Helicobacter Infections/microbiology
- Helicobacter pylori/immunology
- Helicobacter pylori/pathogenicity
- Humans
- Hyaluronan Receptors/genetics
- Hyaluronan Receptors/immunology
- Hyaluronan Receptors/metabolism
- Interferon-gamma/metabolism
- Macrophages, Peritoneal/immunology
- Macrophages, Peritoneal/metabolism
- Mice, Knockout
- Neutrophil Infiltration
- Neutrophils/immunology
- Neutrophils/metabolism
- Precancerous Conditions/diagnosis
- Precancerous Conditions/genetics
- Precancerous Conditions/immunology
- Precancerous Conditions/metabolism
- Precancerous Conditions/microbiology
- Signal Transduction
- Stomach Neoplasms/diagnosis
- Stomach Neoplasms/genetics
- Stomach Neoplasms/immunology
- Stomach Neoplasms/metabolism
- Stomach Neoplasms/microbiology
- Time Factors
Collapse
Affiliation(s)
- Jone Garay
- Stanley S. Scott Cancer Center, LSUHSC, New Orleans, LA, USA
| | - M Blanca Piazuelo
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sumana Majumdar
- Stanley S. Scott Cancer Center, LSUHSC, New Orleans, LA, USA
| | - Li Li
- Stanley S. Scott Cancer Center, LSUHSC, New Orleans, LA, USA
| | | | - Luis Del Valle
- Stanley S. Scott Cancer Center, LSUHSC, New Orleans, LA, USA; Department of Pathology, LSUHSC, New Orleans, LA, USA
| | - Barbara G Schneider
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alberto G Delgado
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Keith T Wilson
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Pelayo Correa
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jovanny Zabaleta
- Stanley S. Scott Cancer Center, LSUHSC, New Orleans, LA, USA; Department of Pediatrics, LSUHSC New Orleans, LA, USA.
| |
Collapse
|
13
|
De Falco M, Lucariello A, Iaquinto S, Esposito V, Guerra G, De Luca A. Molecular Mechanisms of Helicobacter pylori Pathogenesis. J Cell Physiol 2015; 230:1702-7. [PMID: 25639461 DOI: 10.1002/jcp.24933] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 01/16/2015] [Indexed: 12/12/2022]
Abstract
Helicobacter pylori infects 50% of mankind. The vast majority of H. pylori infection occurs in the developing countries where up to 80% of the middle-aged adults may be infected. Bacterial infection causes an inflammatory response that proceeds through a series of intermediated stages of precancerous lesions (gastritis, atrophy, intestinal metaplasia, and dysplasia). Among infected individuals, approximately 10% develops severe gastric lesions such as peptic ulcer disease, 1-3% progresses to gastric cancer (GC) with a low 5-year survival rate, and 0.1% develops mucosa-associated lymphoid tissue (MALT). GC is one of the most common cancer and the third leading cause of cancer-related deaths worldwide. In this review, we have summarized the most recent papers about molecular mechanisms of H. pylori pathogenesis. The main important steps of H. pylori infection such as adhesion, entry in epithelial gastric cells, activation of intracellular pathways until epigenetic modifications have been described.
Collapse
Affiliation(s)
- Maria De Falco
- Department of Biology, University Federico II of Naples, Naples, Italy; National Institute of Biostructures and Biosystems (INBB), Rome, Italy
| | | | | | | | | | | |
Collapse
|
14
|
Saraiva-Pava K, Navabi N, Skoog EC, Lindén SK, Oleastro M, Roxo-Rosa M. New NCI-N87-derived human gastric epithelial line after human telomerase catalytic subunit over-expression. World J Gastroenterol 2015; 21:6526-6542. [PMID: 26074691 PMCID: PMC4458763 DOI: 10.3748/wjg.v21.i21.6526] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 02/07/2015] [Accepted: 03/31/2015] [Indexed: 02/06/2023] Open
Abstract
AIM: To establish a cellular model correctly mimicking the gastric epithelium to overcome the limitation in the study of Helicobacter pylori (H. pylori) infection.
METHODS: Aiming to overcome this limitation, clones of the heterogenic cancer-derived NCI-N87 cell line were isolated, by stably-transducing it with the human telomerase reverse-transcriptase (hTERT) catalytic subunit gene. The clones were first characterized regarding their cell growth pattern and phenotype. For that we measured the clones’ adherence properties, expression of cell-cell junctions’ markers (ZO-1 and E-cadherin) and ability to generate a sustained transepithelial electrical resistance. The gastric properties of the clones, concerning expression of mucins, zymogens and glycan contents, were then evaluated by haematoxylin and eosin staining, Periodic acid Schiff (PAS) and PAS/Alcian Blue-staining, immunocytochemistry and Western blot. In addition, we assessed the usefulness of the hTERT-expressing gastric cell line for H. pylori research, by performing co-culture assays and measuring the IL-8 secretion, by ELISA, upon infection with two H. pylori strains differing in virulence.
RESULTS: Compared with the parental cell line, the most promising NCI-hTERT-derived clones (CL5 and CL6) were composed of cells with homogenous phenotype, presented higher relative telomerase activities, better adhesion properties, ability to be maintained in culture for longer periods after confluency, and were more efficient in PAS-reactive mucins secretion. Both clones were shown to produce high amounts of MUC1, MUC2 and MUC13. NCI-hTERT-CL5 mucins were shown to be decorated with blood group H type 2 (BG-H), Lewis-x (Lex), Ley and Lea and, in a less extent, with BG-A antigens, but the former two antigens were not detected in the NCI-hTERT-CL6. None of the clones exhibited detectable levels of MUC6 nor sialylated Lex and Lea glycans. Entailing good gastric properties, both NCI-hTERT-clones were found to produce pepsinogen-5 and human gastric lipase. The progenitor-like phenotype of NCI-hTERT-CL6 cells was highlighted by large nuclei and by the apical vesicular-like distribution of mucin 5AC and Pg5, supporting the accumulation of mucus-secreting and zymogens-chief mature cells functions.
CONCLUSION: These traits, in addition to resistance to microaerobic conditions and good responsiveness to H. pylori co-culture, in a strain virulence-dependent manner, make the NCI-hTERT-CL6 a promising model for future in vitro studies.
Collapse
|
15
|
Turkina MV, Olofsson A, Magnusson KE, Arnqvist A, Vikström E. Helicobacter pylori vesicles carrying CagA localize in the vicinity of cell-cell contacts and induce histone H1 binding to ATP in epithelial cells. FEMS Microbiol Lett 2015; 362:fnv076. [PMID: 25956174 DOI: 10.1093/femsle/fnv076] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/05/2015] [Indexed: 12/14/2022] Open
Abstract
Helicobacter pylori produces outer membrane vesicles (OMV), delivering bacterial substances including the oncogenic cytotoxin-associated CagA protein to their surroundings. We investigated the effects of H. pylori OMV carrying CagA (OMV-CagA) on cell junctions and ATP-binding proteome of epithelial monolayers, using proteomics, mass spectrometry and imaging. OMV-CagA localized in close vicinity of ZO-1 tight junction protein and induced histone H1 binding to ATP. We suggest the expression of novel events in the interactions between H. pylori OMV and epithelia, which may have an influence on host gene transcription and lead to different outcomes of an infection and development of cancer.
Collapse
Affiliation(s)
- Maria V Turkina
- Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, SE-58185 Linköping, Sweden
| | - Annelie Olofsson
- Department of Medical Biochemistry and Biophysics, Umeå University, SE-90187 Umeå, Sweden
| | - Karl-Eric Magnusson
- Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, SE-58185 Linköping, Sweden
| | - Anna Arnqvist
- Department of Medical Biochemistry and Biophysics, Umeå University, SE-90187 Umeå, Sweden
| | - Elena Vikström
- Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University, SE-58185 Linköping, Sweden
| |
Collapse
|
16
|
Gao C, Liang C, Nie Z, Liu Y, Wang J, Zhang D. Alkannin inhibits growth and invasion of glioma cells C6 through IQGAP/mTOR signal pathway. Int J Clin Exp Med 2015; 8:5287-5294. [PMID: 26131103 PMCID: PMC4483937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 03/27/2015] [Indexed: 06/04/2023]
Abstract
OBJECTIVE This study aims to explore the effect of alkannin on the growth and invasion of glioma cells and its mechanism. METHODS The effects of alkannin on the growth and invasion of glioma cells were detected with MTT assay, clone forming test and transwell assay. The effects of alkannin on the cell cycle were detected with flow cytometry assay. The changes of cyclin, MMPs and IQGAP/mTOR signal pathway related proteins were detected with western blotting methods. RESULTS Alkannin (1 μM, 3 μM and 10 μM) can significantly inhibit the growth, proliferation, migration and invasion of glioma cells C6 with dose dependent. Alkannin can block cell cycle in G1 phase with the increased concentration, which was related with the down-regulation of cyclinA1, cyclinA2 and cyclinD1 expression. Alkannin can also down-regulate the expression of MMP 2, MMP 9 and IQGAP. Alkannin has no effect on mTOR but can inhibit the phosphorylation of mTOR. CONCLUSIONS Alkannin can inhibit the growth and invasion of glioma cells C6 through IQGAP/mTOR signal pathway.
Collapse
Affiliation(s)
- Chunyan Gao
- Yanjing Medical School, Capital University of Medical Sciences Beijing 101300, China
| | - Cunyin Liang
- Yanjing Medical School, Capital University of Medical Sciences Beijing 101300, China
| | - Zhengui Nie
- Yanjing Medical School, Capital University of Medical Sciences Beijing 101300, China
| | - Ying Liu
- Yanjing Medical School, Capital University of Medical Sciences Beijing 101300, China
| | - Junya Wang
- Yanjing Medical School, Capital University of Medical Sciences Beijing 101300, China
| | - Dongmei Zhang
- Yanjing Medical School, Capital University of Medical Sciences Beijing 101300, China
| |
Collapse
|
17
|
Brücher BL, Jamall IS. Epistemology of the origin of cancer: a new paradigm. BMC Cancer 2014; 14:331. [PMID: 24885752 PMCID: PMC4026115 DOI: 10.1186/1471-2407-14-331] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Accepted: 05/06/2014] [Indexed: 02/06/2023] Open
Abstract
Background Carcinogenesis is widely thought to originate from somatic mutations and an inhibition of growth suppressors, followed by cell proliferation, tissue invasion, and risk of metastasis. Fewer than 10% of all cancers are hereditary; the ratio in gastric (1%), colorectal (3-5%) and breast (8%) cancers is even less. Cancers caused by infection are thought to constitute some 15% of the non-hereditary cancers. Those remaining, 70 to 80%, are called “sporadic,” because they are essentially of unknown etiology. We propose a new paradigm for the origin of the majority of cancers. Presentation of hypothesis Our paradigm postulates that cancer originates following a sequence of events that include (1) a pathogenic stimulus (biological or chemical) followed by (2) chronic inflammation, from which develops (3) fibrosis with associated changes in the cellular microenvironment. From these changes a (4) pre-cancerous niche develops, which triggers the deployment of (5) a chronic stress escape strategy, and when this fails to resolve, (6) a transition of a normal cell to a cancer cell occurs. If we are correct, this paradigm would suggest that the majority of the findings in cancer genetics so far reported are either late events or are epiphenomena that occur after the appearance of the pre-cancerous niche. Testing the hypothesis If, based on experimental and clinical findings presented here, this hypothesis is plausible, then the majority of findings in the genetics of cancer so far reported in the literature are late events or epiphenomena that could have occurred after the development of a PCN. Our model would make clear the need to establish preventive measures long before a cancer becomes clinically apparent. Future research should focus on the intermediate steps of our proposed sequence of events, which will enhance our understanding of the nature of carcinogenesis. Findings on inflammation and fibrosis would be given their warranted importance, with research in anticancer therapies focusing on suppressing the PCN state with very early intervention to detect and quantify any subclinical inflammatory change and to treat all levels of chronic inflammation and prevent fibrotic changes, and so avoid the transition from a normal cell to a cancer cell. Implication of the hypothesis The paradigm proposed here, if proven, spells out a sequence of steps, one or more of which could be interdicted or modulated early in carcinogenesis to prevent or, at a minimum, slow down the progression of many cancers.
Collapse
|
18
|
Marcus EA, Vagin O, Tokhtaeva E, Sachs G, Scott DR. Helicobacter pylori impedes acid-induced tightening of gastric epithelial junctions. Am J Physiol Gastrointest Liver Physiol 2013; 305:G731-9. [PMID: 23989011 PMCID: PMC3840231 DOI: 10.1152/ajpgi.00209.2013] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Gastric infection by Helicobacter pylori is the most common cause of ulcer disease and gastric cancer. The mechanism of progression from gastritis and inflammation to ulcers and cancer in a fraction of those infected is not definitively known. Significant acidity is unique to the gastric environment and is required for ulcer development. The interplay between gastric acidity and H. pylori pathogenesis is important in progression to advanced disease. The aim of this study was to characterize the impact of acid on gastric epithelial integrity and cytokine release and how H. pylori infection alters these responses. Human gastric epithelial (HGE-20) cells were grown on porous inserts, and survival, barrier function, and cytokine release were studied at various apical pH levels in the presence and absence of H. pylori. With apical acidity, gastric epithelial cells demonstrate increased barrier function, as evidenced by increased transepithelial electrical resistance (TEER) and decreased paracellular permeability. This effect is reduced in the presence of wild-type, but not urease knockout, H. pylori. The epithelial inflammatory response is also modulated by acidity and H. pylori infection. Without H. pylori, epithelial IL-8 release decreases in acid, while IL-6 release increases. In the presence of H. pylori, acidic pH diminishes the magnitude of the previously reported increase in IL-8 and IL-6 release. H. pylori interferes with the gastric epithelial response to acid, contributing to altered barrier function and inflammatory response. H. pylori diminishes acid-induced tightening of cell junctions in a urease-dependent manner, suggesting that local pH elevation promotes barrier compromise and progression to mucosal damage.
Collapse
Affiliation(s)
- Elizabeth A. Marcus
- 1Department of Pediatrics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California; ,4Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, California
| | - Olga Vagin
- 2Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California; ,4Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, California
| | - Elmira Tokhtaeva
- 2Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California; ,4Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, California
| | - George Sachs
- 2Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California; ,3Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California; and ,4Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, California
| | - David R. Scott
- 2Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California; ,4Veterans Administration Greater Los Angeles Healthcare System, Los Angeles, California
| |
Collapse
|
19
|
Osman MA, Bloom GS, Tagoe EA. Helicobacter pylori-induced alteration of epithelial cell signaling and polarity: a possible mechanism of gastric carcinoma etiology and disparity. Cytoskeleton (Hoboken) 2013; 70:349-59. [PMID: 23629919 DOI: 10.1002/cm.21114] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2013] [Revised: 03/04/2013] [Accepted: 04/24/2013] [Indexed: 12/11/2022]
Abstract
Gastric cancer, a disease of disparity associated with Helicobacter pylori (H. pylori) infection, is the world's second leading cause of cancer deaths. The pathogen H. pylori target the epithelial adhesion receptors, E-cadherin, and β1-integrin, to modulate the host cytoskeleton via disruption of the epithelial cell polarity necessary for maintaining the infection, but how this leads to the development of the carcinoma is widely unclear. While Rho family GTPases' signaling to the cytoskeleton and these receptors is required for initiating and maintaining the infection, the responsible effectors, and how they might influence the etiology of the carcinomas are currently unknown. Here we discuss the potential role of the Cdc42-IQGAP1 axis, a negative regulator of the tumor suppressors E-cadherin and β1-integrin, as a potential driver of H. pylori-induced gastric carcinoma and propose avenues for addressing its disparity. Chronic dysfunction of the IQGAP1-signaling pathway, resulting from H. pylori-induced disruption of cell polarity, can explain the pathogenesis of the carcinoma, at least, in subsets of infected population, and thus could provide a potential means for personalized medicine.
Collapse
Affiliation(s)
- Mahasin A Osman
- Department of Molecular Pharmacology, Physiology and Biotechnology, Warren Alpert Medical School, Brown University, Providence, RI 02912, USA.
| | | | | |
Collapse
|
20
|
Wroblewski LE, Peek RM. Helicobacter pylori in gastric carcinogenesis: mechanisms. Gastroenterol Clin North Am 2013; 42:285-98. [PMID: 23639641 PMCID: PMC3648881 DOI: 10.1016/j.gtc.2013.01.006] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Helicobacter pylori infection induces chronic inflammation and is the strongest known risk factor for gastric cancer. The genomes of H pylori are highly diverse and therefore bacterial virulence factors play an important role in determining the outcome of H pylori infection, in combination with host responses that are augmented by environmental and dietary risk factors. It is important to gain further understanding of the pathogenesis of H pylori infection to develop more effective treatments for this common but deadly malignancy. This review focuses on the specific mechanisms used by H pylori to drive gastric carcinogenesis.
Collapse
Affiliation(s)
| | - Richard M. Peek
- Department of Medicine, Vanderbilt University, Nashville, TN.
,Department of Cancer Biology, Vanderbilt University, Nashville, TN
| |
Collapse
|
21
|
Blair V, Kahokehr A, Sammour T. Cancer in Māori: lessons from prostate, colorectal and gastric cancer and progress in hereditary stomach cancer in New Zealand. ANZ J Surg 2013; 83:42-8. [PMID: 23279256 DOI: 10.1111/ans.12042] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/06/2012] [Indexed: 01/26/2023]
Abstract
Persisting ethnic disparities in cancer incidence and outcomes exist between Māori and non-Māori in Aotearoa/New Zealand. It is difficult to disentangle the complex interplay of environmental and genetic factors that contribute to the variation in cancer statistics between these two groups. In Māori, the sites of highest cancer incidence are the prostate in men, breast in women and lung in both - the next most common cancers in Māori are colorectal and stomach cancer. This paper discusses colorectal, prostate and stomach cancer in Māori to illustrate selected issues that impact on cancer care. Colorectal cancer is discussed to illustrate the importance of accurate cancer statistics to focus management strategies. Prostate cancer in Māori is reviewed - an area where cultural factors impact on care delivery. Sporadic stomach cancer in New Zealand is used to show how sub-classification of different types of cancer can be important and illustrate the breadth of putative causal factors. Then follows an overview of developments in hereditary gastric cancer in New Zealand in the last 15 years, showing how successful clinical and research partnerships can improve patient outcomes. One example is the Kimi Hauora Clinic, which provides support to cancer patients, mutation carriers and their families, helping them navigate the interface with the many health-care professionals involved in the multidisciplinary care of cancer patients in the 21st century.
Collapse
Affiliation(s)
- Vanessa Blair
- Department of Surgery, University of Auckland, Auckland, New Zealand.
| | | | | |
Collapse
|
22
|
Costa AM, Leite M, Seruca R, Figueiredo C. Adherens junctions as targets of microorganisms: a focus on Helicobacter pylori. FEBS Lett 2012; 587:259-65. [PMID: 23262219 DOI: 10.1016/j.febslet.2012.12.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Revised: 12/07/2012] [Accepted: 12/10/2012] [Indexed: 12/14/2022]
Abstract
Mucosal epithelia are targeted by several microorganisms as a way of adhesion, internalization, and/or exploitation of the host properties to induce disease. Helicobacter pylori are worldwide prevalent bacteria that colonize the human stomach. Persistent infection of the gastric mucosa with H. pylori and concurrent chronic gastritis are risk factors for ulcer disease and gastric carcinoma. Therefore, interactions at the H. pylori-epithelial interface are important to understand the pathogenesis of these bacteria and the host responses that contribute to disease development. Here, we provide an overview of the interactions between microorganisms and the adherens junctions with an emphasis on H. pylori.
Collapse
Affiliation(s)
- Angela Margarida Costa
- IPATIMUP - Institute of Molecular Pathology and Immunology of the University of Porto,Portugal
| | | | | | | |
Collapse
|
23
|
Carneiro P, Fernandes MS, Figueiredo J, Caldeira J, Carvalho J, Pinheiro H, Leite M, Melo S, Oliveira P, Simões-Correia J, Oliveira MJ, Carneiro F, Figueiredo C, Paredes J, Oliveira C, Seruca R. E-cadherin dysfunction in gastric cancer--cellular consequences, clinical applications and open questions. FEBS Lett 2012; 586:2981-9. [PMID: 22841718 DOI: 10.1016/j.febslet.2012.07.045] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Revised: 07/16/2012] [Accepted: 07/17/2012] [Indexed: 02/06/2023]
Abstract
E-cadherin plays a major role in cell-cell adhesion and inactivating germline mutations in its encoding gene predispose to hereditary diffuse gastric cancer. Evidence indicates that aside from its recognized role in early tumourigenesis, E-cadherin is also pivotal for tumour progression, including invasion and metastization. Herein, we discuss E-cadherin alterations found in a cancer context, associated cellular effects and signalling pathways, and we raise new key questions that will impact in the management of GC patients and families.
Collapse
Affiliation(s)
- Patrícia Carneiro
- IPATIMUP, Institute of Molecular Pathology and Immunology of the University of Porto, Rua Dr. Roberto Frias s/n, Porto, Portugal
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Paredes J, Figueiredo J, Albergaria A, Oliveira P, Carvalho J, Ribeiro AS, Caldeira J, Costa AM, Simões-Correia J, Oliveira MJ, Pinheiro H, Pinho SS, Mateus R, Reis CA, Leite M, Fernandes MS, Schmitt F, Carneiro F, Figueiredo C, Oliveira C, Seruca R. Epithelial E- and P-cadherins: role and clinical significance in cancer. Biochim Biophys Acta Rev Cancer 2012; 1826:297-311. [PMID: 22613680 DOI: 10.1016/j.bbcan.2012.05.002] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Revised: 05/09/2012] [Accepted: 05/11/2012] [Indexed: 01/26/2023]
Abstract
E-cadherin and P-cadherin are major contributors to cell-cell adhesion in epithelial tissues, playing pivotal roles in important morphogenetic and differentiation processes during development, and in maintaining integrity and homeostasis in adult tissues. It is now generally accepted that alterations in these two molecules are observed during tumour progression of most carcinomas. Genetic or epigenetic alterations in E- and P-cadherin-encoding genes (CDH1 and CDH3, respectively), or alterations in their proteins expression, often result in tissue disorder, cellular de-differentiation, increased invasiveness of tumour cells and ultimately in metastasis. In this review, we will discuss the major properties of E- and P-cadherin molecules, its regulation in normal tissue, and their alterations and role in cancer, with a specific focus on gastric and breast cancer models.
Collapse
|
25
|
Wroblewski LE, Peek RM. "Targeted disruption of the epithelial-barrier by Helicobacter pylori". Cell Commun Signal 2011; 9:29. [PMID: 22044698 PMCID: PMC3225297 DOI: 10.1186/1478-811x-9-29] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2011] [Accepted: 11/01/2011] [Indexed: 02/08/2023] Open
Abstract
Helicobacter pylori colonizes the human gastric epithelium and induces chronic gastritis, which can lead to gastric cancer. Through cell-cell contacts the gastric epithelium forms a barrier to protect underlying tissue from pathogenic bacteria; however, H. pylori have evolved numerous strategies to perturb the integrity of the gastric barrier. In this review, we summarize recent research into the mechanisms through which H. pylori disrupts intercellular junctions and disrupts the gastric epithelial barrier.
Collapse
Affiliation(s)
- Lydia E Wroblewski
- Division of Gastroenterology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
| | | |
Collapse
|
26
|
Wessler S, Gimona M, Rieder G. Regulation of the actin cytoskeleton in Helicobacter pylori-induced migration and invasive growth of gastric epithelial cells. Cell Commun Signal 2011; 9:27. [PMID: 22044652 PMCID: PMC3214149 DOI: 10.1186/1478-811x-9-27] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2011] [Accepted: 11/01/2011] [Indexed: 02/08/2023] Open
Abstract
Dynamic rearrangement of the actin cytoskeleton is a significant hallmark of Helicobacter pylori (H. pylori) infected gastric epithelial cells leading to cell migration and invasive growth. Considering the cellular mechanisms, the type IV secretion system (T4SS) and the effector protein cytotoxin-associated gene A (CagA) of H. pylori are well-studied initiators of distinct signal transduction pathways in host cells targeting kinases, adaptor proteins, GTPases, actin binding and other proteins involved in the regulation of the actin lattice. In this review, we summarize recent findings of how H. pylori functionally interacts with the complex signaling network that controls the actin cytoskeleton of motile and invasive gastric epithelial cells.
Collapse
Affiliation(s)
- Silja Wessler
- Division of Molecular Biology, Department of Microbiology, University of Salzburg, Salzburg, Austria.
| | | | | |
Collapse
|
27
|
Helicobacter pylori activates calpain via toll-like receptor 2 to disrupt adherens junctions in human gastric epithelial cells. Infect Immun 2011; 79:3887-94. [PMID: 21825064 DOI: 10.1128/iai.05109-11] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Helicobacter pylori is a risk factor for the development of gastritis, gastroduodenal ulcers, and gastric adenocarcinoma. H. pylori-induced disruption of epithelial adherens junctions (AJs) is thought to promote the development of severe disease; however, the mechanisms whereby H. pylori alters AJ structure remain incompletely understood. The present study demonstrates that H. pylori infection in human patients is associated with elevated serum levels of an 80-kDa E-cadherin ectodomain, whose presence is independent of the presence of serum antibodies against CagA. In vitro, a heat-labile H. pylori surface component activates the host protease calpain in human gastric MKN45 cells independently of the virulence factors CagA and VacA. H. pylori-induced calpain activation results in cleavage of E-cadherin to produce a 100-kDa truncated form and induce relocalization of E-cadherin and β-catenin. Stimulation of MKN45 cells with the toll-like receptor 2 (TLR2) ligand P3C activated calpain and disrupted E-cadherin and β-catenin in a pattern similar to that induced by H. pylori. Inhibition of TLR2 prevented H. pylori-induced calpain activation and AJ disassembly. Together, these findings identify a novel pathway whereby H. pylori activates calpain via TLR2 to disrupt gastric epithelial AJ structure.
Collapse
|
28
|
Lin WH, Wu CR, Fang TJ, Guo JT, Huang SY, Lee MS, Yang HL. Anti-Helicobacter pylori activity of fermented milk with lactic acid bacteria. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2011; 91:1424-1431. [PMID: 21445876 DOI: 10.1002/jsfa.4327] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2010] [Revised: 12/29/2010] [Accepted: 01/10/2011] [Indexed: 05/30/2023]
Abstract
BACKGROUND Ten strains of lactic acid bacteria (LAB) were investigated for their anti-Helicobacter pylori effects. The bactericidal activity and organic acid content in spent culture supernatants (SCS) from fermented milk were measured. In addition, the exclusion effect of SCS against H. pylori infection of human gastric epithelial AGS cells was assayed. RESULTS Three LAB strains, LY1, LY5 and IF22, showed better anti-Helicobacter effects than the other strains. There were no significant differences in the bactericidal activity of LAB strains between original SCS, artificial SCS and SCS treated by heating or protease digestion. However, neutralised SCS lost this activity. These results suggest that the anti-H. pylori activity of SCS may be related to the concentration of organic acids and the pH value but not to protein components. In the AGS cell culture test, both fermented LY5-SCS and artificial LY5-SCS significantly reduced H. pylori infection and urease activity (P < 0.05). CONCLUSION In this study, in vitro methods were used to screen potential probiotics with anti-H. pylori activity. This may provide an excellent and rapid system for studying probiotics in the functional food and dairy industries.
Collapse
Affiliation(s)
- Wen-Hsin Lin
- School of Pharmacy, China Medical University, No. 91, Hsueh Shih Road, Taichung City 404, Taiwan
| | | | | | | | | | | | | |
Collapse
|
29
|
Jacobs G, Hellmig S, Huse K, Titz A, Franke A, Kwiatkowski R, Ott S, Kosmahl M, Fischbach W, Lucius R, Klapper W, Fölsch UR, Hampe J, Schreiber S, Rosenstiel P. Polymorphisms in the 3'-untranslated region of the CDH1 gene are a risk factor for primary gastric diffuse large B-cell lymphoma. Haematologica 2011; 96:987-95. [PMID: 21459793 DOI: 10.3324/haematol.2010.033126] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Primary gastric B-cell lymphomas arise from mucosa-associated lymphatic tissue (MALT) in patients with chronic Helicobacter pylori infection. We investigated whether germline variants in the CDH1 gene, coding for E-cadherin, genetically predispose patients to primary gastric B-cell lymphoma. DESIGN AND METHODS Single marker analyses of the CDH1 gene were conducted in patients with primary gastric B-cell lymphoma (n=144), in patients with primary gastric high-grade lymphoma (n=61), and in healthy blood donors (n=361). Twelve single nucleotide polymorphisms were genotyped by TaqMan(®) technology. Allelic imbalance was tested by pyrosequencing and clone direct sequencing of heterozygote genomic and cDNA. Mutation detection was conducted around the poly-A signal of the CDH1 3'-untranslated region. The influence of the 3'-untranslated region on protein translation was determined by a luciferase reporter assay. RESULTS Single marker analyses identified two single nucleotide polymorphisms in strong linkage disequilibrium located in the CDH1 3'-untranslated region. One of them was significantly associated with primary gastric diffuse large B-cell lymphomas after correction for multiple testing and this association was confirmed in an independent sample set. Patients homozygous for the rare T allele (rs1801026) had a 4.9-fold increased risk (95% CI: 1.5-15.9) of developing primary gastric diffuse large B-cell lymphoma. Allelic imbalance and reporter gene assays indicated a putative influence on mRNA stability and/or translational efficacy. CONCLUSIONS We identified variants in CDH1 as the first potential genetic risk factors for the development of primary gastric diffuse large B-cell lymphomas. One of the potentially causative variants affects allelic CDH1 expression. These findings support the hypothesis that besides somatic alterations of B-cells, germline variants in the CDH1 gene contribute to a predisposition to the development of primary gastric diffuse large B-cell lymphomas.
Collapse
Affiliation(s)
- Gunnar Jacobs
- Institute of Clinical Molecular Biology, Christian-Albrechts University, Kiel, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Kim H, White CD, Sacks DB. IQGAP1 in microbial pathogenesis: Targeting the actin cytoskeleton. FEBS Lett 2011; 585:723-9. [PMID: 21295032 PMCID: PMC3085995 DOI: 10.1016/j.febslet.2011.01.041] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2010] [Revised: 01/25/2011] [Accepted: 01/26/2011] [Indexed: 11/18/2022]
Abstract
Microbial pathogens cause widespread morbidity and mortality. Central to the pathogens' virulence is manipulation of the host cell's cytoskeleton, which facilitates microbial invasion, multiplication, and avoidance of the innate immune response. IQGAP1 is a ubiquitously expressed scaffold protein that integrates diverse signaling cascades. Research has shown that IQGAP1 binds to and modulates the activity of multiple proteins that participate in bacterial invasion. Here, we review data that support a role for IQGAP1 in infectious disease via its ability to regulate the actin cytoskeleton. In addition, we explore other mechanisms by which IQGAP1 may be exploited by microbial pathogens.
Collapse
Affiliation(s)
- Hugh Kim
- Department of Translational Medicine, Brigham and Women's Hospital and Harvard Medical School, 1 Blackfan Circle, Boston, MA 02115, USA
| | | | | |
Collapse
|
31
|
Helicobacter pylori and gastric cancer: factors that modulate disease risk. Clin Microbiol Rev 2010; 23:713-39. [PMID: 20930071 DOI: 10.1128/cmr.00011-10] [Citation(s) in RCA: 993] [Impact Index Per Article: 66.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Helicobacter pylori is a gastric pathogen that colonizes approximately 50% of the world's population. Infection with H. pylori causes chronic inflammation and significantly increases the risk of developing duodenal and gastric ulcer disease and gastric cancer. Infection with H. pylori is the strongest known risk factor for gastric cancer, which is the second leading cause of cancer-related deaths worldwide. Once H. pylori colonizes the gastric environment, it persists for the lifetime of the host, suggesting that the host immune response is ineffective in clearing this bacterium. In this review, we discuss the host immune response and examine other host factors that increase the pathogenic potential of this bacterium, including host polymorphisms, alterations to the apical-junctional complex, and the effects of environmental factors. In addition to host effects and responses, H. pylori strains are genetically diverse. We discuss the main virulence determinants in H. pylori strains and the correlation between these and the diverse clinical outcomes following H. pylori infection. Since H. pylori inhibits the gastric epithelium of half of the world, it is crucial that we continue to gain understanding of host and microbial factors that increase the risk of developing more severe clinical outcomes.
Collapse
|
32
|
You YH, Song YY, Meng FL, He LH, Zhang MJ, Yan XM, Zhang JZ. Time-series gene expression profiles in AGS cells stimulated with Helicobacter pylori. World J Gastroenterol 2010; 16:1385-96. [PMID: 20238406 PMCID: PMC2842531 DOI: 10.3748/wjg.v16.i11.1385] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To extend the knowledge of the dynamic interaction between Helicobacter pylori (H. pylori) and host mucosa.
METHODS: A time-series cDNA microarray was performed in order to detect the temporal gene expression profiles of human gastric epithelial adenocarcinoma cells infected with H. pylori. Six time points were selected to observe the changes in the model. A differential expression profile at each time point was obtained by comparing the microarray signal value with that of 0 h. Real-time polymerase chain reaction was subsequently performed to evaluate the data quality.
RESULTS: We found a diversity of gene expression patterns at different time points and identified a group of genes whose expression levels were significantly correlated with several important immune response and tumor related pathways.
CONCLUSION: Early infection may trigger some important pathways and may impact the outcome of the infection.
Collapse
|
33
|
Lin WH, Lin CK, Sheu SJ, Hwang CF, Ye WT, Hwang WZ, Tsen HY. Antagonistic Activity of Spent Culture Supernatants of Lactic Acid Bacteria against Helicobacter Pylori Growth and Infection in Human Gastric Epithelial AGS Cells. J Food Sci 2009; 74:M225-30. [DOI: 10.1111/j.1750-3841.2009.01194.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
34
|
Krueger S, Kuester D, Bernhardt A, Wex T, Roessner A. Regulation of cathepsin X overexpression in H. pylori-infected gastric epithelial cells and macrophages. J Pathol 2009; 217:581-8. [PMID: 19090485 DOI: 10.1002/path.2485] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Cathepsin X (CTSX) is strongly up-regulated in Helicobacter pylori-infected gastric mucosa and intestinal-type gastric cancer. The overexpression of CTSX is mediated predominantly by associated macrophages; depends on a functional type IV-secretion system; and leads to increased migration of gastric epithelial cells. In the present study, we analysed the role of CagA in CTSX overexpression and identified H. pylori-induced inflammatory factors and signalling pathways required for stimulating CTSX expression by H. pylori. Gastric epithelial cells were co-cultured with macrophages in Transwell chambers of 0.4 microm pore size, enabling exchange of fluids but retracting H. pylori. N87 gastric epithelial cells were infected with H. pylori P1 wild-type strain in the presence of inhibitors for p38, JNK, and ERK1/2 signal transduction pathways. Furthermore, cytokines and growth factors were tested for their regulatory function using inhibitory antibodies, and their gene expression was studied by quantitative RT-PCRs and western blots. CTSX is strongly up-regulated at both the mRNA and the protein levels by TNF-alpha, IL-1beta, IL-6, and IL-8, depending on cell type. All these cytokines were found to be increased by five- to ten-fold in macrophages by H. pylori infection of co-cultured N87 gastric epithelial cells. In macrophages, H. pylori up-regulated CTSX via ERK1/2 signalling pathways, and in N87 cells via JNK irrespective of p38 signalling. Our results suggest that H. pylori induced overexpression of CTSX in macrophages and epithelium through specific cytokines that are initiated by CagA-dependent pathways in a cell type-dependent manner.
Collapse
Affiliation(s)
- Sabine Krueger
- Institute of Pathology, Otto-von-Guericke University, Leipziger Strasse 44, D-39120 Magdeburg, Germany.
| | | | | | | | | |
Collapse
|
35
|
Wroblewski LE, Shen L, Ogden S, Romero-Gallo J, Lapierre LA, Israel DA, Turner JR, Peek RM. Helicobacter pylori dysregulation of gastric epithelial tight junctions by urease-mediated myosin II activation. Gastroenterology 2009; 136:236-46. [PMID: 18996125 PMCID: PMC2678540 DOI: 10.1053/j.gastro.2008.10.011] [Citation(s) in RCA: 152] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2008] [Revised: 09/26/2008] [Accepted: 10/02/2008] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Helicobacter pylori-induced gastritis predisposes to the development of gastric cancer. Increased epithelial tight junction permeability and alterations in apical-junctional complexes are also associated with an increased risk of carcinogenesis. Phosphorylation of myosin regulatory light chain (MLC) by MLC kinase (MLCK) regulates tight junction function. We determined whether MLCK was activated by H pylori and defined the mechanisms through which such activation dysregulates gastric epithelial barrier function. METHODS MKN28 gastric epithelial cells were cocultured with the H pylori cag(+) strain 60190 or cagA(-), cagE(-), ureB(-), or vacA(-) mutants. MLC phosphorylation and barrier integrity were determined by immunoblot analysis and transepithelial electrical resistance measurements, respectively. Localization of the tight junction protein occludin was determined by immunocytochemistry in MKN28 cells and INS-GAS mice. RESULTS H pylori induced a progressive loss of barrier function that was attenuated by inactivation of ureB, but not cagA, cagE, or vacA. Reductions in transepithelial electrical resistance were also dependent on functional urease activity. H pylori increased MLC phosphorylation in epithelial monolayers; this was significantly decreased by inhibition of MLCK or Rho kinase or by loss of UreB. H pylori infection of either cultured monolayers or hypergastrinemic INS-GAS mice induced occludin endocytosis, reflecting cytoskeletally mediated disruption of tight junctions. CONCLUSIONS H pylori increases MLC phosphorylation, occludin internalization and barrier dysfunction in gastric epithelial cells. This process requires functional urease activity and is independent of the cag pathogenicity island or VacA. These data provide new insights into the mechanisms by which H pylori disrupts gastric barrier function.
Collapse
Affiliation(s)
- Lydia E. Wroblewski
- Division of Gastroenterology, Departments of Medicine and Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232-2279, USA
| | - Le Shen
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - Seth Ogden
- Division of Gastroenterology, Departments of Medicine and Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232-2279, USA
| | - Judith Romero-Gallo
- Division of Gastroenterology, Departments of Medicine and Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232-2279, USA
| | - Lynne A. Lapierre
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Dawn A. Israel
- Division of Gastroenterology, Departments of Medicine and Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232-2279, USA
| | - Jerrold R. Turner
- Department of Pathology, The University of Chicago, Chicago, IL 60637, USA
| | - Richard M. Peek
- Division of Gastroenterology, Departments of Medicine and Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232-2279, USA
| |
Collapse
|
36
|
Freedman ND, Ahn J, Hou L, Lissowska J, Zatonski W, Yeager M, Chanock SJ, Chow WH, Abnet CC. Polymorphisms in estrogen- and androgen-metabolizing genes and the risk of gastric cancer. Carcinogenesis 2008; 30:71-7. [PMID: 19015200 DOI: 10.1093/carcin/bgn258] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Androgens and estrogens may play a role in gastric cancer etiology. To investigate the association of gastric cancer with single-nucleotide polymorphisms (SNPs) in six genes (COMT, CYP1B1, CYP17A1, CYP19A1, HSD17B1 and SHBG) involved in estrogen and androgen synthesis and metabolism, 58 haplotype-tagging SNPs were genotyped in 295 gastric cancer cases and 415 controls from a population-based study in Poland. We assessed differences in haplotype frequency between cases and controls using a global score test and calculated multivariate odds ratios (ORs) and 95% confidence intervals (CIs) for individual haplotypes using logistic regression. We found associations in one linkage disequilibrium (LD) block containing the 3' untranslated region of COMT (rs9332377, rs165728, rs165849 and rs1110478), global score test (df = 4, P = 0.033). Relative to the most frequent GATA haplotype, the GATG haplotype was associated with statistically significant increased gastric cancer risk (OR = 1.50, 95% CI: 1.06-2.12; false discovery rate (FDR) value = 0.459) and the AACA haplotype with borderline increased risk (OR = 1.36, 95% CI = 1.00-1.85; FDR = 0.50). We also found associations for the LD block containing part of the SHBG coding region (rs6258, rs6259, rs2955617, rs1641544 and rs1641537). The CACCC haplotype was associated with statistically significant lower gastric cancer risk relative to the referent CGACC haplotype (OR = 0.55, 95% CI = 0.34-0.90; FDR = 0.459), but the overall score test was statistically non-significant. No other statistically significant associations were observed. In summary, we found possible associations between gastric cancer and polymorphisms in COMT, involved in estrogen inactivation, and SHBG, a modulator of hormone bioavailability. These findings should be interpreted cautiously until replicated in other studies.
Collapse
Affiliation(s)
- Neal D Freedman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD 20852, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Wessler S, Backert S. Molecular mechanisms of epithelial-barrier disruption by Helicobacter pylori. Trends Microbiol 2008; 16:397-405. [PMID: 18619844 DOI: 10.1016/j.tim.2008.05.005] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2008] [Revised: 05/08/2008] [Accepted: 05/28/2008] [Indexed: 12/22/2022]
Abstract
Intact intercellular junctions and cell-matrix contacts are important structures in the formation and maintenance of epithelial-barrier functions against microbes. The human gastric pathogen Helicobacter pylori developed a remarkable network of strategies to alter these epithelial cell-cell and cell-matrix adhesions, which are implicated in inflammation, proliferation, cell migration and invasive growth. This review focuses on recent findings on H. pylori-induced host-cell signaling. We propose a stepwise model for how H. pylori interacts with components of focal adhesions and intercellular tight and adherens junctions to disrupt the epithelial layer, providing novel insights into the pathogenesis of H. pylori.
Collapse
Affiliation(s)
- Silja Wessler
- Junior Research Group, Paul-Ehrlich Institute, D-63225 Langen, Germany.
| | | |
Collapse
|
38
|
Lin LL, Chen CN, Lin WC, Lee PH, Chang KJ, Lai YP, Wang JT, Juan HF. Annexin A4: A novel molecular marker for gastric cancer with Helicobacter pylori infection using proteomics approach. Proteomics Clin Appl 2008; 2:619-34. [PMID: 21136859 DOI: 10.1002/prca.200780088] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2007] [Indexed: 01/29/2023]
Abstract
Helicobacter pylori was reported to be an important risk factor for the carcinogenesis of gastric cancer. Here, we used a proteomic approach to find differentially expressed proteins between the normal and tumor tissue of gastric cancer patients infected with H. pylori. In our results, we found annexin A4 was over-expressed in patients infected with H. pylori and was found in tumor cells, and over-expressed in gastric cancer SCM-1 cells after H. pylori infection. Ca(2+ ) can be induced by H. pylori and interact with annexin A4 Ca(2+) binding site to block the calmodulin-activated chloride conductance activation; therefore, it produces a new environment that benefits the malignant existence of H. pylori and raises the risk for gastric cancer. We also found interleuken-8 (IL-8) expression levels were increased in H. pylori infected SCM-1 cells. Combined with previous reports and our results, we summarize that the over-expression of annexin A4 in SCM-1 cells with H. pylori infection may subsequently induce IL-8 which can further cause tumor angiogenesis. In this paper, we show that annexin A4 is a potential novel molecular marker for gastric cancer with H. pylori infection, and our results may provide a new insight in the development of new anti-cancer drugs.
Collapse
Affiliation(s)
- Li-Ling Lin
- Institute of Molecular and Cellular Biology, National Taiwan University, Taipei, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Mojtahedi A, Salehi R, Navabakbar F, Tamizifar H, Tavakkoli H, Duronio V. Evaluation of apoptosis induction using PARP cleavage on gastric adenocarcinoma and fibroblast cell lines by different strains of Helicobacter pylori. Pak J Biol Sci 2007; 10:4097-4102. [PMID: 19090286 DOI: 10.3923/pjbs.2007.4097.4102] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Helicobacter pylori is one of the most common pathogens affecting humans and is the major environmental factor in the development of gastric cancer increasing from 4 to 6 folds the risk of its development. Variations in cancer risk among H. pylori infected individuals may correlate to difference in H. pylori strains, variable host characteristics and specific interactions between host and microbial determinants. To determine the effect of different strains of H. pylori on cellular apoptosis this study was designed an in vitro model using AGS and HEF cell lines. After specified time intervals total cell proteins was extracted and subjected to SDS-PAGE and immunoblotting using anti poly ADP-ribose polymerase (PARP) antibody. Decrease in densitometric value of PARP was indicative of higher level of apoptosis. The ability of apoptosis induction in AGS and HEF cell lines by wild type (cagA+/vacA+), cagA-/vacA+, vacA-/cagA+ and double negative (cagA-/vacA-) strains were significantly different. The assessed apoptosis in AGS cell line co-cultured with wild type strain was 3.22 +/- 0.2 in 24 h, 2.8 +/- 0.1 in 48 and 2.1 +/- 0.09 in 72 h of incubation time. Similar assessment with cagA-/vacA+ strains in AGS cells was 4.17 +/- 1.49 in 24 h, 3.32 +/- 0.45 in 48 h and 2.32 +/- 0.61 in 72 h incubation. A variation in apoptotic potential between the H. pylori strains on two cells (AGS and HEF) was observed. Based on present results, it is concluded that H. pylori strains as well as target cell types are important in pathogenesis and induction of apoptosis during a specified time interval.
Collapse
Affiliation(s)
- Ali Mojtahedi
- Department of Medical Microbiology, Faculty of Medicine, Guilan University Complex, Rasht-Tehran Road, Rasht 3477, Iran
| | | | | | | | | | | |
Collapse
|
40
|
Singh P. Role of Annexin-II in GI cancers: interaction with gastrins/progastrins. Cancer Lett 2007; 252:19-35. [PMID: 17188424 PMCID: PMC1941619 DOI: 10.1016/j.canlet.2006.11.012] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2006] [Accepted: 11/06/2006] [Indexed: 12/27/2022]
Abstract
The role of the gastrin peptide hormones (G17, G34) and their precursors (progastrins, PG; gly-extended gastrin, G-gly), in gastrointestinal (GI) cancers has been extensively reviewed in recent years [W. Rengifo-Cam, P. Singh, Role of progastrins and gastrins and their receptors in GI and pancreatic cancers: targets for treatment, Curr. Pharm. Des. 10 (19) (2004) 2345-2358; M. Dufresne, C. Seva, D. Fourmy, Cholecystokinin and gastrin receptors, Physiol. Rev. 86 (3) (2006) 805-847; A. Ferrand, T.C. Wang, Gastrin and cancer: a review, Cancer Lett. 238 (1) (2006) 15-29]. A possible important role of progastrin peptides in colon carcinogenesis has become evident from experiments with transgenic mouse models [W. Rengifo-Cam, P. Singh, (2004); A. Ferrand, T.C. Wang, (2006)]. It is now known that growth stimulatory and co-carcinogenic effects of gastrin/PG peptides are mediated by both proliferative and anti-apoptotic effects of the peptides on target cells [H. Wu, G.N. Rao, B. Dai, P. Singh, Autocrine gastrins in colon cancer cells Up-regulate cytochrome c oxidase Vb and down-regulate efflux of cytochrome c and activation of caspase-3, J. Biol. Chem. 275 (42) (2000) 32491-32498; H. Wu, A. Owlia, P. Singh, Precursor peptide progastrin(1-80) reduces apoptosis of intestinal epithelial cells and upregulates cytochrome c oxidase Vb levels and synthesis of ATP, Am. J. Physiol. Gastrointest. Liver Physiol. 285 (6) (2003) G1097-G1110]. Several receptor subtypes have been described that mediate growth effects of gastrin peptides [W. Rengifo-Cam, P. Singh (2004); M. Dufresne, C. Seva, D. Fourmy, (2006)]. Recently, we identified Annexin II as a high affinity binding protein for gastrin/PG peptides [P. Singh, H. Wu, C. Clark, A. Owlia, Annexin II binds progastrin and gastrin-like peptides, and mediates growth factor effects of autocrine and exogenous gastrins on colon cancer and intestinal epithelial cells, Oncogene (2006), doi:10.1038/sj.onc.1209798]. Importantly, the expression of Annexin II was required for mediating growth stimulatory effects of gastrin and PG peptides on intestinal epithelial and colon cancer cells [P. Singh, H. Wu, C. Clark, A. Owlia, Annexin II binds progastrin and gastrin-like peptides, and mediates growth factor effects of autocrine and exogenous gastrins on colon cancer and intestinal epithelial cells, Oncogene (2006), doi:10.1038/sj.onc.1209798], suggesting that Annexin-II may represent the elusive novel receptor for gastrin/PG peptides. The importance of this finding in relation to the structure and function of Annexin-II, especially in GI cancers, is described below. Since this surprising finding represents a new front in our understanding of the mechanisms involved in mediating growth effects of gastrin/PG peptides in GI cancers, our current understanding of the role of Annexin-II in proliferation and metastasis of cancer cells is additionally reviewed.
Collapse
Affiliation(s)
- Pomila Singh
- Department of Neuroscience and Cell Biology, 10.104 Medical Research Building, Route 1043, University of Texas Medical Branch, 301University Blvd., Mail Route 1043, Galveston, TX 77555-1043, USA.
| |
Collapse
|
41
|
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
|
42
|
Krueger S, Hundertmark T, Kuester D, Kalinski T, Peitz U, Roessner A. Helicobacter pylori alters the distribution of ZO-1 and p120ctn in primary human gastric epithelial cells. Pathol Res Pract 2007; 203:433-44. [PMID: 17509776 DOI: 10.1016/j.prp.2007.04.003] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2007] [Accepted: 04/19/2007] [Indexed: 02/08/2023]
Abstract
Helicobacter pylori infection is related to the development of diverse gastric pathologies, possibly by affecting epithelial junctional complexes that define cell polarity and play an essential role in transepithelial transport and cell-cell adhesion. Using primary gastric epithelial cell cultures, effects of H. pylori on the expression and localization of tight/adherence junction proteins and the resulting morphological changes and migratory capabilities were studied under in vivo-like conditions. Gastric epithelial cells were isolated from biopsies or gastrectomies and maintained in Quantum286 on collagen I-coated culture dishes or cover-slips. Cell cultures were characterized and further analyzed by western blot and immunofluorescent staining for ZO-1, p120ctn, and H. pylori CagA. Morphological changes and migratory response were monitored by time-lapse digital image microscopy. ZO-1 and p120ctn protein expression levels remain unaffected by H. pylori infection. Immunocytochemistry on H. pylori-infected primary cell monolayers focally showed disruption of intercellular ZO-1 staining and accumulation of ZO-1 in small vesicles. H. pylori infection recruited non-phosphorylated p120ctn to perinuclear vesicles. The fraction of phosphorylated p120ctn increased and could be detected in the nucleus, at the cell membrane, and at the leading edge of migrating cells. These alterations, triggered by H. pylori infection, are associated with an elongation phenotype and increased migration.
Collapse
Affiliation(s)
- Sabine Krueger
- Department of Pathology, Otto-von-Guericke University, Leipziger Str. 44, D-39120 Magdeburg, Germany.
| | | | | | | | | | | |
Collapse
|
43
|
Humar B, Fukuzawa R, Blair V, Dunbier A, More H, Charlton A, Yang HK, Kim WH, Reeve AE, Martin I, Guilford P. Destabilized adhesion in the gastric proliferative zone and c-Src kinase activation mark the development of early diffuse gastric cancer. Cancer Res 2007; 67:2480-9. [PMID: 17363565 DOI: 10.1158/0008-5472.can-06-3021] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The initial development of diffuse gastric cancer (DGC) is poorly understood. The study of E-cadherin (CDH1) germ line mutation carriers predisposed to DGC provides a rare opportunity to elucidate the genetic and biological events surrounding disease initiation. Samples from various stages of hereditary and sporadic DGC were investigated to determine general mechanisms underlying early DGC development. Paraffin-embedded tissues from 13 CDH1 mutation carriers and from 10 sporadic early DGC cases were analyzed. Immunofluorescence and immunohistochemistry using differentiation, proliferation, and adhesion markers showed that DGC initiation seems to occur at the proliferative zone (the upper neck) of the gastric epithelium and correlates with absent or reduced expression of junctional proteins (beta-actin, p120, Lin-7). Slow proliferation of neoplastic cells at the upper gastric neck leads to the formation of intramucosal signet-ring cell carcinoma (SRCC) displaying differentiated features. As shown by immunolabeling, invasion from SRCC lesions beyond the gastric mucosa is associated with poor differentiation, increased proliferation, activation of the c-Src system, and an epithelial-mesenchymal transition. Our results provide a molecular description of the early development of DGC and explain the relationship between the two main DGC types, poorly differentiated carcinoma and SRCC: both share their origin, but SRCC develops following cancer cell differentiation and seems relatively indolent in its intramucosal stage.
Collapse
Affiliation(s)
- Bostjan Humar
- Cancer Genetics Laboratory, Department of Biochemistry, University of Otago, Dunedin, New Zealand.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Abstract
PURPOSE OF REVIEW Although chronic Helicobacter pylori infection is the strongest known risk factor for development of gastric adenocarcinoma, only a small proportion of infected individuals will ever develop tumours. This article discusses various bacterial, host and environmental factors which may influence an individual's susceptibility. RECENT FINDINGS Recent research on bacterial virulence factors has focussed upon the cag pathogenicity island, particularly its roles in regulating epithelial growth and adhesion. Studies of host genetic factors have included several analyses of polymorphisms in inflammatory cytokines in human cohorts. Animal studies have recently clarified the roles of dysregulated epithelial apoptosis, proliferation and differentiation pathways during gastric carcinogenesis, and novel experiments involving H. felis infection of bone marrow transplanted irradiated mice have suggested that gastric cancer may originate from bone marrow-derived stem cells. Important roles for signalling between epithelial and mesenchymal cells, particularly myofibroblasts, are also emerging. Recent research on the importance of environmental factors has demonstrated how helminth coinfection may protect against atrophic gastritis and T helper type 1 responses. SUMMARY Complex interactions between several bacterial, host genetic and environmental factors determine whether H. pylori infected individuals develop gastric carcinoma. The importance of bone marrow stem cell engraftment during human gastric neoplasia is an area requiring urgent investigation.
Collapse
Affiliation(s)
- D Mark Pritchard
- Division of Gastroenterology, University of Liverpool, Liverpool, UK.
| | | |
Collapse
|
45
|
Suzuki M, Mimuro H, Suzuki T, Park M, Yamamoto T, Sasakawa C. Interaction of CagA with Crk plays an important role in Helicobacter pylori-induced loss of gastric epithelial cell adhesion. ACTA ACUST UNITED AC 2006; 202:1235-47. [PMID: 16275761 PMCID: PMC2213224 DOI: 10.1084/jem.20051027] [Citation(s) in RCA: 185] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
CagA protein is a major virulence factor of Helicobacter pylori, which is delivered into gastric epithelial cells and elicits growth factor-like responses. Once within the cells, CagA is tyrosine phosphorylated by Src family kinases and targets host proteins required to induce the cell responses. We show that the phosphorylated CagA binds Crk adaptor proteins (Crk-II, Crk-I, and Crk-L) and that the interaction is important for the CagA-mediated host responses during H. pylori infection. H. pylori-induced scattering of gastric epithelial cells in culture was blocked by overexpression of dominant-negative Crk and by RNA interference-mediated knockdown of endogenous Crk. H. pylori infection of the gastric epithelium induced disruption of E-cadherin/catenin-containing adherens junctions, which was also dependent on CagA/Crk signaling. Furthermore, inhibition of the SoS1/H-Ras/Raf1, C3G/Rap1/B-Raf, or Dock180/Rac1/Wiskott-Aldrich syndrome protein family verprolin homologous protein pathway, all of which are involved downstream of Crk adaptors, greatly diminished the CagA-associated host responses. Thus, CagA targeting of Crk plays a central role in inducing the pleiotropic cell responses to H. pylori infection that cause several gastric diseases, including gastric cancer.
Collapse
Affiliation(s)
- Masato Suzuki
- Department of Microbiology and Immunology, International Research Center for Infectious Diseases, Institute of Medical Science, University of Tokyo, Minato-ku, Japan
| | | | | | | | | | | |
Collapse
|
46
|
Sousa S, Lecuit M, Cossart P. Microbial strategies to target, cross or disrupt epithelia. Curr Opin Cell Biol 2005; 17:489-98. [PMID: 16102958 DOI: 10.1016/j.ceb.2005.08.013] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2005] [Accepted: 08/03/2005] [Indexed: 12/17/2022]
Abstract
Epithelia are highly organized structures adapted to protect the underlying tissues from external aggressions, including microbial infections. Consequently, pathogens have evolved various strategies to target directly or indirectly intercellular junctions and/or components that maintain the structure of epithelia. Interestingly, some extracellular pathogens secrete enzymes that modify the extracellular part of junction components. Others produce toxins that are endocytosed and act from the inside of the cell to disrupt epithelial junctions. Other pathogens may directly inject into cells factors that are targeted to and destabilize the junctions, or that interact with signaling cascades that affect junction stability. Finally invasive bacteria or viruses may, by entering into cells, destabilize the junctions by targeting junction components directly or by inducing a series of events that lead to chemokine secretion, polymorphonuclear recruitment and inflammation.
Collapse
Affiliation(s)
- Sandra Sousa
- Unité des Interactions Bactéries-Cellules Institut Pasteur, INSERM U604, INRA USC2020, 75724 Paris Cedex 15, France
| | | | | |
Collapse
|
47
|
Stock M, Otto F. Gene deregulation in gastric cancer. Gene 2005; 360:1-19. [PMID: 16154715 DOI: 10.1016/j.gene.2005.06.026] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2005] [Revised: 05/26/2005] [Accepted: 06/13/2005] [Indexed: 12/21/2022]
Abstract
Despite its decreasing frequency in the Western world during recent decades, gastric cancer is still one of the leading causes of cancer-related deaths worldwide. Due to the oligosymptomatic course of early gastric cancer, most cases are diagnosed in the advanced stages of the disease. The curative potential of current standard treatment continues to be unsatisfactory, despite multimodal approaches involving surgery, chemotherapy and radiotherapy. Novel therapeutics including small molecules and monoclonal antibodies are being developed and have been partially introduced into clinical use in connection with neoplastic diseases such as chronic myeloid leukemia, non-Hodgkin's lymphoma and colorectal cancer. Thorough understanding of the changes in gene expression occurring during gastric carcinogenesis may help to develop targeted therapies and improve the treatment of this disease. Novel molecular biology techniques have generated a wealth of data on up- and down-regulation, activation and inhibition of specific pathways in gastric cancer. Here, we provide an overview of the different aspects of aberrant gene expression patterns in gastric cancer.
Collapse
Affiliation(s)
- Michael Stock
- Department of Hematology and Oncology, University Hospital Freiburg, Hugstetter Strasse 55, D-79106 Freiburg, Germany
| | | |
Collapse
|
48
|
Marshall BJ, Windsor HM. The relation of Helicobacter pylori to gastric adenocarcinoma and lymphoma: pathophysiology, epidemiology, screening, clinical presentation, treatment, and prevention. Med Clin North Am 2005; 89:313-44, viii. [PMID: 15656929 DOI: 10.1016/j.mcna.2004.09.001] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Helicobacter pylori infection may be the most common chronic bacterial infection worldwide; however, the prevalence varies between countries and is usually linked to socioeconomic conditions. Gastric cancer is one of the most frequent cancers in developing countries and usually about the seventh most common in developed countries. This article explores the relation of H. pylori to gastric adenocarcinoma and lymphoma. The pathophysiology, epidemiology, screening, clinical presentation, treatment, and prevention are discussed.
Collapse
Affiliation(s)
- Barry J Marshall
- Department of Microbiology, University of Western Australia, 35 Stirling Highway, Crawley, Western Australia 6009, Australia.
| | | |
Collapse
|