1
|
Patel NM, Patel PH, Bhogal RH, Harrington KJ, Singanayagam A, Kumar S. Altered Microbiome Promotes Pro-Inflammatory Pathways in Oesophago-Gastric Tumourigenesis. Cancers (Basel) 2024; 16:3426. [PMID: 39410045 PMCID: PMC11476036 DOI: 10.3390/cancers16193426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/27/2024] [Accepted: 10/04/2024] [Indexed: 10/20/2024] Open
Abstract
INTRODUCTION The upper gastrointestinal microbiome is a dynamic entity that is involved in numerous processes including digestion, production of vitamins and protection against pathogens. Many external and intrinsic factors may cause changes in the proportions of bacteria within the microbial community, termed 'dysbiosis'. A number of these have been identified as risk factors for a range of diseases, including oesophago-gastric carcinoma. MATERIALS AND METHODS A narrative review was conducted to elucidate the current evidence on the role of the microbiome in promoting oesophago-gastric tumourigenesis. Significant causes of dysbiosis including age, medications and GORD were examined and key pro-inflammatory pathways implicated in tumourigenesis and their interaction with the microbiome were described. RESULTS AND DISCUSSION An association between microbial dysbiosis and development of oesophago-gastric cancer may be mediated via activation of pro-inflammatory pathways, the inflammasome and the innate immune system. Advances in sequencing technology allow microbial communities to be fingerprinted by sequencing the 16S rRNA gene, enabling a deeper understanding of the genera that may be implicated in driving tumourigenesis. CONCLUSIONS Developing a greater understanding of the influence of the microbiota on oesophago-gastric tumourigenesis may enable advances to be made in the early detection of malignancy and in the development of novel systemic therapies, leading to improved rates of survival.
Collapse
Affiliation(s)
- Nikhil Manish Patel
- Department of Upper GI Surgery, The Royal Marsden NHS Foundation Trust, London SW3 6JJ, UK; (N.M.P.)
- The Upper Gastrointestinal Surgical Oncology Research Group, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London SW7 3RP, UK
| | - Pranav Harshad Patel
- Department of Upper GI Surgery, The Royal Marsden NHS Foundation Trust, London SW3 6JJ, UK; (N.M.P.)
- The Upper Gastrointestinal Surgical Oncology Research Group, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London SW7 3RP, UK
| | - Ricky Harminder Bhogal
- Department of Upper GI Surgery, The Royal Marsden NHS Foundation Trust, London SW3 6JJ, UK; (N.M.P.)
- The Upper Gastrointestinal Surgical Oncology Research Group, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London SW7 3RP, UK
| | - Kevin Joseph Harrington
- Targeted Therapy Group, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London SW7 3RP, UK
| | - Aran Singanayagam
- Centre for Bacterial Resistance Biology, Department of Infectious Disease, Imperial College London, London SW7 2AZ, UK
| | - Sacheen Kumar
- Department of Upper GI Surgery, The Royal Marsden NHS Foundation Trust, London SW3 6JJ, UK; (N.M.P.)
- The Upper Gastrointestinal Surgical Oncology Research Group, Division of Radiotherapy and Imaging, The Institute of Cancer Research, London SW7 3RP, UK
- Department of Upper Gastrointestinal Surgery, Digestive Disease and Surgery Institute, Cleveland Clinic London Hospital, London SW1X 7HY, UK
| |
Collapse
|
2
|
Drnovsek J, Homan M, Zidar N, Smid LM. Pathogenesis and potential reversibility of intestinal metaplasia - a milestone in gastric carcinogenesis. Radiol Oncol 2024; 58:186-195. [PMID: 38643513 PMCID: PMC11165985 DOI: 10.2478/raon-2024-0028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 03/19/2024] [Indexed: 04/23/2024] Open
Abstract
BACKGROUND Non-cardia gastric cancer remains a major cause of cancer-related mortality worldwide, despite declining incidence rates in many industrialized countries. The development of intestinal-type gastric cancer occurs through a multistep process in which normal mucosa is sequentially transformed into hyperproliferative epithelium, followed by metaplastic processes leading to carcinogenesis. Chronic infection with Helicobacter pylori is the primary etiological agent that causes chronic inflammation of the gastric mucosa, induces atrophic gastritis, and can lead to intestinal metaplasia and dysplasia. Both intestinal metaplasia and dysplasia are precancerous lesions, in which gastric cancer is more likely to occur. Atrophic gastritis often improves after eradication of Helicobacter pylori; however, the occurrence of intestinal metaplasia has been traditionally regarded as "the point of no return" in the carcinogenesis sequence. Helicobacter pylori eradication heals non-atrophic chronic gastritis, may lead to regression of atrophic gastritis, and reduces the risk of gastric cancer in patients with these conditions. In this article, we discuss the pathogenesis, epigenomics, and reversibility of intestinal metaplasia and briefly touch upon potential treatment strategy. CONCLUSIONS Gastric intestinal metaplasia no longer appears to be an irreversible precancerous lesion. However, there are still many controversies regarding the improvement of intestinal metaplasia after Helicobacter pylori eradication.
Collapse
Affiliation(s)
- Jan Drnovsek
- Department of Gastroenterology, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Matjaz Homan
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Department of Gastroenterology, Hepatology and Nutrition, University Children’s Hospital, Ljubljana, Slovenia
| | - Nina Zidar
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
- Institute of Pathology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Lojze M Smid
- Department of Gastroenterology, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
3
|
Ong LL, Jan HM, Le HHT, Yang TC, Kuo CY, Feng AF, Mong KKT, Lin CH. Membrane lipid remodeling eradicates Helicobacter pylori by manipulating the cholesteryl 6'-acylglucoside biosynthesis. J Biomed Sci 2024; 31:44. [PMID: 38685037 PMCID: PMC11057186 DOI: 10.1186/s12929-024-01031-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 04/14/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND Helicobacter pylori, the main cause of various gastric diseases, infects approximately half of the human population. This pathogen is auxotrophic for cholesterol which it converts to various cholesteryl α-glucoside derivatives, including cholesteryl 6'-acyl α-glucoside (CAG). Since the related biosynthetic enzymes can be translocated to the host cells, the acyl chain of CAG likely comes from its precursor phosphatidylethanolamine (PE) in the host membranes. This work aims at examining how the acyl chain of CAG and PE inhibits the membrane functions, especially bacterial adhesion. METHODS Eleven CAGs that differ in acyl chains were used to study the membrane properties of human gastric adenocarcinoma cells (AGS cells), including lipid rafts clustering (monitored by immunofluorescence with confocal microscopy) and lateral membrane fluidity (by the fluorescence recovery after photobleaching). Cell-based and mouse models were employed to study the degree of bacterial adhesion, the analyses of which were conducted by using flow cytometry and immunofluorescence staining, respectively. The lipidomes of H. pylori, AGS cells and H. pylori-AGS co-cultures were analyzed by Ultraperformance Liquid Chromatography-Tandem Mass Spectroscopy (UPLC-MS/MS) to examine the effect of PE(10:0)2, PE(18:0)2, PE(18:3)2, or PE(22:6)2 treatments. RESULTS CAG10:0, CAG18:3 and CAG22:6 were found to cause the most adverse effect on the bacterial adhesion. Further LC-MS analysis indicated that the treatment of PE(10:0)2 resulted in dual effects to inhibit the bacterial adhesion, including the generation of CAG10:0 and significant changes in the membrane compositions. The initial (1 h) lipidome changes involved in the incorporation of 10:0 acyl chains into dihydro- and phytosphingosine derivatives and ceramides. In contrast, after 16 h, glycerophospholipids displayed obvious increase in their very long chain fatty acids, monounsaturated and polyunsaturated fatty acids that are considered to enhance membrane fluidity. CONCLUSIONS The PE(10:0)2 treatment significantly reduced bacterial adhesion in both AGS cells and mouse models. Our approach of membrane remodeling has thus shown great promise as a new anti-H. pylori therapy.
Collapse
Affiliation(s)
- Lih-Lih Ong
- Institute of Biological Chemistry, Academia Sinica, No. 128, Academia Road Section 2, Nan-Kang, Taipei, 11529, Taiwan
- Department of Applied Chemistry, National Yang Ming Chiao Tung University, 1001, University Road, Eastern District, Hsinchu, 300093, Taiwan
- Institute of Chemistry, Academia Sinica, No. 128, Academia Road Section 2, Nan-Kang, Taipei, 11529, Taiwan
- Sustainable Chemical Science and Technology, Taiwan International Graduate Program, Academia Sinica, No. 128, Academia Road Section 2, Nan-Kang, Taipei, 11529, Taiwan
| | - Hau-Ming Jan
- Institute of Biological Chemistry, Academia Sinica, No. 128, Academia Road Section 2, Nan-Kang, Taipei, 11529, Taiwan
- Joint Program in Transfusion Medicine, Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, USA
| | - Hong-Hanh Thi Le
- Institute of Biological Chemistry, Academia Sinica, No. 128, Academia Road Section 2, Nan-Kang, Taipei, 11529, Taiwan
| | - Tsai-Chen Yang
- Institute of Biological Chemistry, Academia Sinica, No. 128, Academia Road Section 2, Nan-Kang, Taipei, 11529, Taiwan
| | - Chou-Yu Kuo
- Institute of Biological Chemistry, Academia Sinica, No. 128, Academia Road Section 2, Nan-Kang, Taipei, 11529, Taiwan
| | - Ai-Feng Feng
- Department of Applied Chemistry, National Yang Ming Chiao Tung University, 1001, University Road, Eastern District, Hsinchu, 300093, Taiwan
| | - Kwok-Kong Tony Mong
- Department of Applied Chemistry, National Yang Ming Chiao Tung University, 1001, University Road, Eastern District, Hsinchu, 300093, Taiwan.
| | - Chun-Hung Lin
- Institute of Biological Chemistry, Academia Sinica, No. 128, Academia Road Section 2, Nan-Kang, Taipei, 11529, Taiwan.
- Department of Chemistry and Institute of Biochemical Sciences, National Taiwan University, Taipei, 10617, Taiwan.
| |
Collapse
|
4
|
Bhattacharjee A, Sahoo OS, Sarkar A, Bhattacharya S, Chowdhury R, Kar S, Mukherjee O. Infiltration to infection: key virulence players of Helicobacter pylori pathogenicity. Infection 2024; 52:345-384. [PMID: 38270780 DOI: 10.1007/s15010-023-02159-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 12/13/2023] [Indexed: 01/26/2024]
Abstract
PURPOSE This study aims to comprehensively review the multifaceted factors underlying the successful colonization and infection process of Helicobacter pylori (H. pylori), a prominent Gram-negative pathogen in humans. The focus is on elucidating the functions, mechanisms, genetic regulation, and potential cross-interactions of these elements. METHODS Employing a literature review approach, this study examines the intricate interactions between H. pylori and its host. It delves into virulence factors like VacA, CagA, DupA, Urease, along with phase variable genes, such as babA, babC, hopZ, etc., giving insights about the bacterial perspective of the infection The association of these factors with the infection has also been added in the form of statistical data via Funnel and Forest plots, citing the potential of the virulence and also adding an aspect of geographical biasness to the virulence factors. The biochemical characteristics and clinical relevance of these factors and their effects on host cells are individually examined, both comprehensively and statistically. RESULTS H. pylori is a Gram-negative, spiral bacterium that successfully colonises the stomach of more than half of the world's population, causing peptic ulcers, gastric cancer, MALT lymphoma, and other gastro-duodenal disorders. The clinical outcomes of H. pylori infection are influenced by a complex interplay between virulence factors and phase variable genes produced by the infecting strain and the host genetic background. A meta-analysis of the prevalence of all the major virulence factors has also been appended. CONCLUSION This study illuminates the diverse elements contributing to H. pylori's colonization and infection. The interplay between virulence factors, phase variable genes, and host genetics determines the outcome of the infection. Despite biochemical insights into many factors, their comprehensive regulation remains an understudied area. By offering a panoramic view of these factors and their functions, this study enhances understanding of the bacterium's perspective, i.e. H. pylori's journey from infiltration to successful establishment within the host's stomach.
Collapse
Affiliation(s)
- Arghyadeep Bhattacharjee
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur, West Bengal, 713209, India
- Department of Microbiology, Kingston College of Science, Beruanpukuria, Barasat, West Bengal, 700219, India
| | - Om Saswat Sahoo
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur, West Bengal, 713209, India
| | - Ahana Sarkar
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur, West Bengal, 713209, India
| | - Saurabh Bhattacharya
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, The Hebrew University of Jerusalem, P.O.B. 12272, 9112001, Jerusalem, Israel
| | - Rukhsana Chowdhury
- School of Biological Sciences, RKM Vivekananda Educational and Research Institute Narendrapur, Kolkata, India
| | - Samarjit Kar
- Department of Mathematics, National Institute of Technology Durgapur, Durgapur, West Bengal, 713209, India
| | - Oindrilla Mukherjee
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur, West Bengal, 713209, India.
| |
Collapse
|
5
|
Zeng J, Xie C, Huang Z, Cho CH, Chan H, Li Q, Ashktorab H, Smoot DT, Wong SH, Yu J, Gong W, Liang C, Xu H, Chen H, Liu X, Wu JCY, Ip M, Gin T, Zhang L, Chan MTV, Hu W, Wu WKK. LOX-1 acts as an N 6-methyladenosine-regulated receptor for Helicobacter pylori by binding to the bacterial catalase. Nat Commun 2024; 15:669. [PMID: 38253620 PMCID: PMC10803311 DOI: 10.1038/s41467-024-44860-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
The role of N6-methyladenosine (m6A) modification of host mRNA during bacterial infection is unclear. Here, we show that Helicobacter pylori infection upregulates host m6A methylases and increases m6A levels in gastric epithelial cells. Reducing m6A methylase activity via hemizygotic deletion of methylase-encoding gene Mettl3 in mice, or via small interfering RNAs targeting m6A methylases, enhances H. pylori colonization. We identify LOX-1 mRNA as a key m6A-regulated target during H. pylori infection. m6A modification destabilizes LOX-1 mRNA and reduces LOX-1 protein levels. LOX-1 acts as a membrane receptor for H. pylori catalase and contributes to bacterial adhesion. Pharmacological inhibition of LOX-1, or genetic ablation of Lox-1, reduces H. pylori colonization. Moreover, deletion of the bacterial catalase gene decreases adhesion of H. pylori to human gastric sections. Our results indicate that m6A modification of host LOX-1 mRNA contributes to protection against H. pylori infection by downregulating LOX-1 and thus reducing H. pylori adhesion.
Collapse
Affiliation(s)
- Judeng Zeng
- State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China
- CUHK Shenzhen Research Institute, Shenzhen, China
| | - Chuan Xie
- State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Jiangxi Province, China
| | - Ziheng Huang
- State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China
- CUHK Shenzhen Research Institute, Shenzhen, China
| | - Chi H Cho
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Hung Chan
- State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China
- CUHK Shenzhen Research Institute, Shenzhen, China
| | - Qing Li
- State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China
- CUHK Shenzhen Research Institute, Shenzhen, China
| | - Hassan Ashktorab
- Department of Medicine, Howard University, Washington, DC, USA
- Cancer Center, Howard University, Washington, DC, USA
- Howard University Hospital, Howard University, Washington, DC, USA
| | - Duane T Smoot
- Department of Internal Medicine, Meharry Medical College, Nashville, TN, USA
| | - Sunny H Wong
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Jun Yu
- State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China
- CUHK Shenzhen Research Institute, Shenzhen, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China
| | - Wei Gong
- Department of Gastroenterology, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
- The Third School of Clinical Medicine, Southern Medical University, Shenzhen, Guangdong, China
| | - Cong Liang
- State Key Laboratory of Cellular Stress Biology and School of Life Sciences, Xiamen University, Xiamen, China
| | - Hongzhi Xu
- Institute for Microbial Ecology, School of Medicine, Department of Gastroenterology, Zhongshan Hospital, Xiamen University, Xiamen, China
| | - Huarong Chen
- State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China
- CUHK Shenzhen Research Institute, Shenzhen, China
| | - Xiaodong Liu
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China
- CUHK Shenzhen Research Institute, Shenzhen, China
| | - Justin C Y Wu
- State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China
| | - Margaret Ip
- CUHK Shenzhen Research Institute, Shenzhen, China
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China
| | - Tony Gin
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China
| | - Lin Zhang
- State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China.
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China.
- CUHK Shenzhen Research Institute, Shenzhen, China.
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China.
| | - Matthew T V Chan
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China.
- CUHK Shenzhen Research Institute, Shenzhen, China.
| | - Wei Hu
- Department of Gastroenterology, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China.
- The Third School of Clinical Medicine, Southern Medical University, Shenzhen, Guangdong, China.
| | - William K K Wu
- State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China.
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China.
- CUHK Shenzhen Research Institute, Shenzhen, China.
| |
Collapse
|
6
|
López-Luis MA, Soriano-Pérez EE, Parada-Fabián JC, Torres J, Maldonado-Rodríguez R, Méndez-Tenorio A. A Proposal for a Consolidated Structural Model of the CagY Protein of Helicobacter pylori. Int J Mol Sci 2023; 24:16781. [PMID: 38069104 PMCID: PMC10706595 DOI: 10.3390/ijms242316781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 11/17/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
CagY is the largest and most complex protein from Helicobacter pylori's (Hp) type IV secretion system (T4SS), playing a critical role in the modulation of gastric inflammation and risk for gastric cancer. CagY spans from the inner to the outer membrane, forming a channel through which Hp molecules are injected into human gastric cells. Yet, a tridimensional structure has been reported for only short segments of the protein. This intricate protein was modeled using different approaches, including homology modeling, ab initio, and deep learning techniques. The challengingly long middle repeat region (MRR) was modeled using deep learning and optimized using equilibrium molecular dynamics. The previously modeled segments were assembled into a 1595 aa chain and a 14-chain CagY multimer structure was assembled by structural alignment. The final structure correlated with published structures and allowed to show how the multimer may form the T4SS channel through which CagA and other molecules are translocated to gastric cells. The model confirmed that MRR, the most polymorphic and complex region of CagY, presents numerous cysteine residues forming disulfide bonds that stabilize the protein and suggest this domain may function as a contractile region playing an essential role in the modulating activity of CagY on tissue inflammation.
Collapse
Affiliation(s)
- Mario Angel López-Luis
- Laboratorio de Biotecnología y Bioinformática Genómica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Campus Lázaro Cárdenas, Mexico City 11340, Mexico; (M.A.L.-L.); (E.E.S.-P.); (J.C.P.-F.); (R.M.-R.)
| | - Eva Elda Soriano-Pérez
- Laboratorio de Biotecnología y Bioinformática Genómica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Campus Lázaro Cárdenas, Mexico City 11340, Mexico; (M.A.L.-L.); (E.E.S.-P.); (J.C.P.-F.); (R.M.-R.)
| | - José Carlos Parada-Fabián
- Laboratorio de Biotecnología y Bioinformática Genómica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Campus Lázaro Cárdenas, Mexico City 11340, Mexico; (M.A.L.-L.); (E.E.S.-P.); (J.C.P.-F.); (R.M.-R.)
| | - Javier Torres
- Unidad de Investigación en Enfermedades Infecciosas, UMAE Pediatría, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico;
| | - Rogelio Maldonado-Rodríguez
- Laboratorio de Biotecnología y Bioinformática Genómica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Campus Lázaro Cárdenas, Mexico City 11340, Mexico; (M.A.L.-L.); (E.E.S.-P.); (J.C.P.-F.); (R.M.-R.)
| | - Alfonso Méndez-Tenorio
- Laboratorio de Biotecnología y Bioinformática Genómica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Campus Lázaro Cárdenas, Mexico City 11340, Mexico; (M.A.L.-L.); (E.E.S.-P.); (J.C.P.-F.); (R.M.-R.)
| |
Collapse
|
7
|
Jamal Eddin TM, Nasr SM, Gupta I, Zayed H, Al Moustafa AE. Helicobacter pylori and epithelial mesenchymal transition in human gastric cancers: An update of the literature. Heliyon 2023; 9:e18945. [PMID: 37609398 PMCID: PMC10440535 DOI: 10.1016/j.heliyon.2023.e18945] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/25/2023] [Accepted: 08/03/2023] [Indexed: 08/24/2023] Open
Abstract
Gastric cancer, a multifactorial disease, is considered one of the most common malignancies worldwide. In addition to genetic and environmental risk factors, infectious agents, such as Epstein-Barr virus (EBV) and Helicobacter pylori (H.pylori) contribute to the onset and development of gastric cancer. H. pylori is a type I carcinogen that colonizes the gastric epithelium of approximately 50% of the world's population, thus increasing the risk of gastric cancer development. On the other hand, epithelial mesenchymal transition (EMT) is a fundamental process crucial to embryogenic growth, wound healing, organ fibrosis and cancer progression. Several studies associate gastric pathogen infection of the epithelium with EMT initiation, provoking cancer metastasis in the gastric mucosa through various molecular signaling pathways. Additionally, EMT is implicated in the progression and development of H. pylori-associated gastric cancer. In this review, we recapitulate recent findings elucidating the association between H. pylori infection in EMT promotion leading to gastric cancer progression and metastasis.
Collapse
Affiliation(s)
- Tala M. Jamal Eddin
- College of Health Sciences, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Shahd M.O. Nasr
- College of Health Sciences, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Ishita Gupta
- College of Medicine, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Hatem Zayed
- College of Health Sciences, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Ala-Eddin Al Moustafa
- College of Medicine, QU Health, Qatar University, PO Box 2713, Doha, Qatar
- Biomedical Research Center, Qatar University, PO Box 2713, Doha, Qatar
- Oncology Department, Faculty of Medicine, McGill University, Montreal, QC, H3G 2M1, Canada
| |
Collapse
|
8
|
Maphosa S, Moleleki LN, Motaung TE. Bacterial secretion system functions: evidence of interactions and downstream implications. MICROBIOLOGY (READING, ENGLAND) 2023; 169. [PMID: 37083586 DOI: 10.1099/mic.0.001326] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/22/2023]
Abstract
Unprecedented insights into the biology and functions of bacteria have been and continue to be gained through studying bacterial secretion systems in isolation. This method, however, results in our understanding of the systems being primarily based on the idea that they operate independently, ignoring the subtleties of downstream interconnections. Gram-negative bacteria are naturally able to adapt to and navigate their frequently varied and dynamic surroundings, mostly because of the covert connections between secretion systems. Therefore, to comprehend some of the linked downstream repercussions for organisms that follow this discourse, it is vital to have mechanistic insights into how the intersecretion system functions in bacterial rivalry, virulence, and survival, among other things. To that purpose, this paper discusses a few key instances of molecular antagonistic and interdependent relationships between bacterial secretion systems and their produced functional products.
Collapse
Affiliation(s)
- Silindile Maphosa
- Division of Microbiology, Department of Biochemistry, Genetics, and Microbiology, University of Pretoria, Hatfield, Pretoria, South Africa
- Department of Plant and Soil Sciences, University of Pretoria, Hatfield, Pretoria, South Africa
- Forestry and Agricultural Biotechnology Institute, University of Pretoria, Hatfield, Pretoria, South Africa
| | - Lucy N Moleleki
- Division of Microbiology, Department of Biochemistry, Genetics, and Microbiology, University of Pretoria, Hatfield, Pretoria, South Africa
- Forestry and Agricultural Biotechnology Institute, University of Pretoria, Hatfield, Pretoria, South Africa
| | - Thabiso E Motaung
- Division of Microbiology, Department of Biochemistry, Genetics, and Microbiology, University of Pretoria, Hatfield, Pretoria, South Africa
- Forestry and Agricultural Biotechnology Institute, University of Pretoria, Hatfield, Pretoria, South Africa
| |
Collapse
|
9
|
Lopes C, Almeida TC, Pimentel-Nunes P, Dinis-Ribeiro M, Pereira C. Linking dysbiosis to precancerous stomach through inflammation: Deeper than and beyond imaging. Front Immunol 2023; 14:1134785. [PMID: 37063848 PMCID: PMC10102473 DOI: 10.3389/fimmu.2023.1134785] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 03/17/2023] [Indexed: 04/03/2023] Open
Abstract
Upper gastrointestinal endoscopy is considered the gold standard for gastric lesions detection and surveillance, but it is still associated with a non-negligible rate of missing conditions. In the Era of Personalized Medicine, biomarkers could be the key to overcome missed lesions or to better predict recurrence, pushing the frontier of endoscopy to functional endoscopy. In the last decade, microbiota in gastric cancer has been extensively explored, with gastric carcinogenesis being associated with progressive dysbiosis. Helicobacter pylori infection has been considered the main causative agent of gastritis due to its interference in disrupting the acidic environment of the stomach through inflammatory mediators. Thus, does inflammation bridge the gap between gastric dysbiosis and the gastric carcinogenesis cascade and could the microbiota-inflammation axis-derived biomarkers be the answer to the unmet challenge of functional upper endoscopy? To address this question, in this review, the available evidence on the role of gastric dysbiosis and chronic inflammation in precancerous conditions of the stomach is summarized, particularly targeting the nuclear factor-κB (NF-κB), toll-like receptors (TLRs) and cyclooxygenase-2 (COX-2) pathways. Additionally, the potential of liquid biopsies as a non-invasive source and the clinical utility of studied biomarkers is also explored. Overall, and although most studies offer a mechanistic perspective linking a strong proinflammatory Th1 cell response associated with, but not limited to, chronic infection with Helicobacter pylori, promising data recently published highlights not only the diagnostic value of microbial biomarkers but also the potential of gastric juice as a liquid biopsy pushing forward the concept of functional endoscopy and personalized care in gastric cancer early diagnosis and surveillance.
Collapse
Affiliation(s)
- Catarina Lopes
- Precancerous Lesions and Early Cancer Management Group, Research Center of IPO Porto (CI‐IPOP)/Rise@CI‐IPOP (Health Research Group), Portuguese Institute of Oncology of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
- CINTESIS – Center for Health Technology and Services Research, University of Porto, Porto, Portugal
- ICBAS-UP – Institute of Biomedical Sciences Abel Salazar, University of Porto, Porto, Portugal
| | - Tatiana C. Almeida
- Precancerous Lesions and Early Cancer Management Group, Research Center of IPO Porto (CI‐IPOP)/Rise@CI‐IPOP (Health Research Group), Portuguese Institute of Oncology of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
| | - Pedro Pimentel-Nunes
- Department of Surgery and Physiology, Faculty of Medicine, University of Porto (FMUP), Porto, Portugal
- Department of Gastroenterology, Unilabs, Porto, Portugal
| | - Mário Dinis-Ribeiro
- Precancerous Lesions and Early Cancer Management Group, Research Center of IPO Porto (CI‐IPOP)/Rise@CI‐IPOP (Health Research Group), Portuguese Institute of Oncology of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
- Department of Gastroenterology, Portuguese Institute of Oncology of Porto, Porto, Portugal
| | - Carina Pereira
- Precancerous Lesions and Early Cancer Management Group, Research Center of IPO Porto (CI‐IPOP)/Rise@CI‐IPOP (Health Research Group), Portuguese Institute of Oncology of Porto (IPO Porto)/Porto Comprehensive Cancer Center (Porto.CCC), Porto, Portugal
- CINTESIS – Center for Health Technology and Services Research, University of Porto, Porto, Portugal
- *Correspondence: Carina Pereira,
| |
Collapse
|
10
|
Wizenty J, Sigal M. Gastric Stem Cell Biology and Helicobacter pylori Infection. Curr Top Microbiol Immunol 2023; 444:1-24. [PMID: 38231213 DOI: 10.1007/978-3-031-47331-9_1] [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 colonizes the human gastric mucosa and persists lifelong. An interactive network between the bacteria and host cells shapes a unique microbial niche within gastric glands that alters epithelial behavior, leading to pathologies such as chronic gastritis and eventually gastric cancer. Gland colonization by the bacterium initiates aberrant trajectories by inducing long-term inflammatory and regenerative gland responses, which involve various specialized epithelial and stromal cells. Recent studies using cell lineage tracing, organoids and scRNA-seq techniques have significantly advanced our knowledge of the molecular "identity" of epithelial and stromal cell subtypes during normal homeostasis and upon infection, and revealed the principles that underly stem cell (niche) behavior under homeostatic conditions as well as upon H. pylori infection. The activation of long-lived stem cells deep in the gastric glands has emerged as a key prerequisite of H. pylori-associated gastric site-specific pathologies such as hyperplasia in the antrum, and atrophy or metaplasia in the corpus, that are considered premalignant lesions. In addition to altering the behaviour of bona fide stem cells, injury-driven de-differentiation and trans-differentation programs, such as "paligenosis", subsequently allow highly specialized secretory cells to re-acquire stem cell functions, driving gland regeneration. This plastic regenerative capacity of gastric glands is required to maintain homeostasis and repair mucosal injuries. However, these processes are co-opted in the context of stepwise malignant transformation in chronic H. pylori infection, causing the emergence, selection and expansion of cancer-promoting stem cells.
Collapse
Affiliation(s)
- Jonas Wizenty
- Division of Gastroenterology and Hepatology, Medical Department, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Michael Sigal
- Division of Gastroenterology and Hepatology, Medical Department, Charité-Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
11
|
Wang H, Zhao M, Shi F, Zheng S, Xiong L, Zheng L. A review of signal pathway induced by virulent protein CagA of Helicobacter pylori. Front Cell Infect Microbiol 2023; 13:1062803. [PMID: 37124036 PMCID: PMC10140366 DOI: 10.3389/fcimb.2023.1062803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 03/24/2023] [Indexed: 05/02/2023] Open
Abstract
Gastric cancer (GC), a common and high-mortality disease, still occupies an important position in current cancer research, and Helicobacter pylori (H. pylori) infection as its important risk factor has been a hot and challenging research area. Among the numerous pathogenic factors of H. pylori, the virulence protein CagA has been widely studied as the only bacterial-derived oncoprotein. It was found that CagA entering into gastric epithelial cells (GECs) can induce the dysregulation of multiple cellular pathways such as MAPK signaling pathway, PI3K/Akt signaling pathway, NF-κB signaling pathway, Wnt/β-catenin signaling pathway, JAK-STAT signaling pathway, Hippo signaling pathway through phosphorylation and non-phosphorylation. These disordered pathways cause pathological changes in morphology, adhesion, polarity, proliferation, movement, and other processes of GECs, which eventually promotes the occurrence of GC. With the deepening of H. pylori-related research, the research on CagA-induced abnormal signaling pathway has been updated and deepened to some extent, so the key signaling pathways activated by CagA are used as the main stem to sort out the pathogenesis of CagA in this paper, aiming to provide new strategies for the H. pylori infection and treatment of GC in the future.
Collapse
Affiliation(s)
- Haiqiang Wang
- Department of Internal Medicine, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Mei Zhao
- Graduate School of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Fan Shi
- Graduate School of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Shudan Zheng
- Graduate School of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Li Xiong
- Graduate School of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Lihong Zheng
- Department of Internal Medicine, Fourth Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
- *Correspondence: Lihong Zheng,
| |
Collapse
|
12
|
Host Cell Antimicrobial Responses against Helicobacter pylori Infection: From Biological Aspects to Therapeutic Strategies. Int J Mol Sci 2022; 23:ijms231810941. [PMID: 36142852 PMCID: PMC9504325 DOI: 10.3390/ijms231810941] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/13/2022] [Accepted: 09/16/2022] [Indexed: 02/07/2023] Open
Abstract
The colonization of Helicobacter pylori (H. pylori) in human gastric mucosa is highly associated with the occurrence of gastritis, peptic ulcer, and gastric cancer. Antibiotics, including amoxicillin, clarithromycin, furazolidone, levofloxacin, metronidazole, and tetracycline, are commonly used and considered the major treatment regimens for H. pylori eradication, which is, however, becoming less effective by the increasing prevalence of H pylori resistance. Thus, it is urgent to understand the molecular mechanisms of H. pylori pathogenesis and develop alternative therapeutic strategies. In this review, we focus on the virulence factors for H. pylori colonization and survival within host gastric mucosa and the host antimicrobial responses against H. pylori infection. Moreover, we describe the current treatments for H. pylori eradication and provide some insights into new therapeutic strategies for H. pylori infection.
Collapse
|
13
|
Miri AH, Kamankesh M, Llopis-Lorente A, Liu C, Wacker MG, Haririan I, Asadzadeh Aghdaei H, Hamblin MR, Yadegar A, Rad-Malekshahi M, Zali MR. The Potential Use of Antibiotics Against Helicobacter pylori Infection: Biopharmaceutical Implications. Front Pharmacol 2022; 13:917184. [PMID: 35833028 PMCID: PMC9271669 DOI: 10.3389/fphar.2022.917184] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 05/31/2022] [Indexed: 11/13/2022] Open
Abstract
Helicobacter pylori (H. pylori) is a notorious, recalcitrant and silent germ, which can cause a variety of debilitating stomach diseases, including gastric and duodenal ulcers and gastric cancer. This microbe predominantly colonizes the mucosal layer of the human stomach and survives in the inhospitable gastric microenvironment, by adapting to this hostile milieu. In this review, we first discuss H. pylori colonization and invasion. Thereafter, we provide a survey of current curative options based on polypharmacy, looking at pharmacokinetics, pharmacodynamics and pharmaceutical microbiology concepts, in the battle against H. pylori infection.
Collapse
Affiliation(s)
- Amir Hossein Miri
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mojtaba Kamankesh
- Polymer Chemistry Department, School of Science, University of Tehran, Tehran, Iran
| | - Antoni Llopis-Lorente
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, Netherlands
| | - Chenguang Liu
- College of Marine Life Science, Ocean University of China, Qingdao, China
| | - Matthias G. Wacker
- Department of Pharmacy, National University of Singapore, Singapore, Singapore
| | - Ismaeil Haririan
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamid Asadzadeh Aghdaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Michael R. Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, South Africa
- *Correspondence: Michael R. Hamblin, ; Abbas Yadegar, ; Mazda Rad-Malekshahi, ; Mohammad Reza Zali,
| | - Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- *Correspondence: Michael R. Hamblin, ; Abbas Yadegar, ; Mazda Rad-Malekshahi, ; Mohammad Reza Zali,
| | - Mazda Rad-Malekshahi
- Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- *Correspondence: Michael R. Hamblin, ; Abbas Yadegar, ; Mazda Rad-Malekshahi, ; Mohammad Reza Zali,
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- *Correspondence: Michael R. Hamblin, ; Abbas Yadegar, ; Mazda Rad-Malekshahi, ; Mohammad Reza Zali,
| |
Collapse
|
14
|
Oster P, Vaillant L, McMillan B, Velin D. The Efficacy of Cancer Immunotherapies Is Compromised by Helicobacter pylori Infection. Front Immunol 2022; 13:899161. [PMID: 35677057 PMCID: PMC9168074 DOI: 10.3389/fimmu.2022.899161] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 04/26/2022] [Indexed: 12/12/2022] Open
Abstract
Helicobacter pylori infects the gastric mucosa of a large number of humans. Although asymptomatic in the vast majority of cases, H pylori infection can lead to the development of peptic ulcers gastric adenocarcinoma and mucosa-associated lymphoid tissue (MALT) lymphoma. Using a variety of mechanisms, H pylori locally suppresses the function of the host immune system to establish chronic infection. Systemic immunomodulation has been observed in both clinical and pre-clinical studies, which have demonstrated that H pylori infection is associated with reduced incidence of inflammatory diseases, such as asthma and Crohn’s disease. The introduction of immunotherapies in the arsenal of anti-cancer drugs has revealed a new facet of H pylori-induced immune suppression. In this review, we will describe the intimate interactions between H pylori and its host, and formulate hypothtyeses describing the detrimental impact of H pylori infection on the efficacy of cancer immunotherapies.
Collapse
|
15
|
Altanbayar O, Amgalanbaatar A, Battogtokh C, Bayarjargal N, Belick D, Kohns Vasconcelos M, Mackenzie CR, Pfeffer K, Henrich B. Characterization of the cagA-gene in Helicobacter pylori in Mongolia and detection of two EPIYA-A enriched CagA types. Int J Med Microbiol 2022; 312:151552. [DOI: 10.1016/j.ijmm.2022.151552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 01/28/2022] [Accepted: 02/22/2022] [Indexed: 11/30/2022] Open
|
16
|
Sirt3, 6, and 7 Genes Expression in Gastric Antral Epithelial Cells of Patients with Helicobacter pylori Infection. Curr Microbiol 2022; 79:114. [PMID: 35195783 DOI: 10.1007/s00284-022-02775-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 01/20/2022] [Indexed: 11/03/2022]
Abstract
Sirtuins, known as the intracellular acylation enzymes, play a major role in regulating the cell's physiological activities. The relevant studies have shown diversely sirtuin genes expression in various cancers in humans. This study has surveyed the transcription of sirt3, 6, and 7 genes in gastric antral epithelial cells (GAECs) of gastritis and gastric adenocarcinoma patients with and without Helicobacter pylori infection. First of all, a case-control study was conducted, including 50 and 53 gastric antral biopsy samples collected from gastritis and gastric adenocarcinoma patients with and without H. pylori infection referred to hospitals of Sanandaj City during 2018-2019. Total RNAs were extracted from biopsy samples, then cDNAs were synthesized by using TaKaRa kits. Quality essay of H. pylori virulence genes expression and relative quantitative essay of sirt3, 6, and 7 genes expressions in gastric antral biopsy samples were performed using the real-time RT-PCR method. The statistical analysis showed the significant correlations between H. pylori vacA s1m2 and sabA cDNAs with sirt3 geneś expression in GAECs (P < 0.05, 0.05 respectively). In addition, sirt6 gene's expression decreased along increasing age in gastric adenocarcinoma patients (P < 0.05). The samples of gastritis patients with gastric antral epithelial biopsy containing H. pylori hopQII, oipA, and sabB cDNA showed an increased amount of sirt7 genes expression (P < 0.05, 0.05, and 0.05 respectively). In conclusion, the H. pylori virulence genes expression and increasing age of patients showed the significant correlations with sirt3, 6, and 7 genes expressions in GAECs of gastric and gastric cancer patients.
Collapse
|
17
|
Prashar A, Capurro MI, Jones NL. Under the Radar: Strategies Used by Helicobacter pylori to Evade Host Responses. Annu Rev Physiol 2021; 84:485-506. [PMID: 34672717 DOI: 10.1146/annurev-physiol-061121-035930] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The body depends on its physical barriers and innate and adaptive immune responses to defend against the constant assault of potentially harmful microbes. In turn, successful pathogens have evolved unique mechanisms to adapt to the host environment and manipulate host defenses. Helicobacter pylori (Hp), a human gastric pathogen that is acquired in childhood and persists throughout life, is an example of a bacterium that is very successful at remodeling the host-pathogen interface to promote a long-term persistent infection. Using a combination of secreted virulence factors, immune subversion, and manipulation of cellular mechanisms, Hp can colonize and persist in the hostile environment of the human stomach. Here, we review the most recent and relevant information regarding how this successful pathogen overcomes gastric epithelial host defense responses to facilitate its own survival and establish a chronic infection. Expected final online publication date for the Annual Review of Physiology, Volume 84 is February 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Akriti Prashar
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada;
| | - Mariana I Capurro
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada;
| | - Nicola L Jones
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, Ontario, Canada; .,Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children, Toronto, Ontario, Canada.,Departments of Paediatrics and Physiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
18
|
Abdi E, Latifi-Navid S, Abedi Sarvestani F, Esmailnejad MH. Emerging therapeutic targets for gastric cancer from a host- Helicobacter pylori interaction perspective. Expert Opin Ther Targets 2021; 25:685-699. [PMID: 34410200 DOI: 10.1080/14728222.2021.1971195] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Gastric cancer (GC) has the higher genetic, cytologic, and architectural heterogeneity compared to other gastrointestinal cancers. By inducing gastric inflammation, Helicobacter pylori (HP) may lead to GC through combining bacterial factors with host factors. In this regard, identification of the major therapeutic targets against the host-HP interactions plays a critical role in GC prevention, diagnosis, and treatment. AREAS COVERED This study offers new insights into the promising therapeutic targets against the angiogenesis, invasion, or metastasis of GC from a host-HP interaction perspective. To this end, MEDLINE, EMBASE, LILACS, AIM, and IndMed databases were searched for relevant articles since 1992. EXPERT OPINION Wnt signaling and COX pathway have a well-documented history in the genesis of GC by HP and might be considered as the most promising targets for early GC treatment. Destroying HP may decrease the risk of GC, but it cannot fully hinder the GC development induced by HP infection. Therefore, targeting HP-activated pathways, especially COX-2/Wnt/beta-catenin/VEGF, TLR2/TLR9/COX-2, COX2-PGE2, and NF-κB/COX-2, as well as EPHA2, MMPs, and miR-543/SIRT1 axis, can be an effective measure in the early treatment of GC. However, different clinical trials and large, multi-center cohorts are required to validate these potentially effective targets for GC therapy.
Collapse
Affiliation(s)
- Esmat Abdi
- Department of Biology, Faculty of Sciences, University of Mohaghegh Ardabili, Ardabil, Iran
| | - Saeid Latifi-Navid
- Department of Biology, Faculty of Sciences, University of Mohaghegh Ardabili, Ardabil, Iran
| | | | | |
Collapse
|
19
|
Baltierra-Uribe SL, Montañez-Barragán A, Romero-Ramírez H, Klimov-Kravtchenko K, Martínez-Pedro KI, Sánchez-Salguero E, Camorlinga-Ponce M, Torres J, Santos-Argumedo L. Colostrum IgA1 antibodies recognize antigens from Helicobacter pylori and prevent cytoskeletal changes in human epithelial cells. Eur J Immunol 2021; 51:2641-2650. [PMID: 34398472 DOI: 10.1002/eji.202049117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 06/11/2021] [Accepted: 08/13/2021] [Indexed: 11/06/2022]
Abstract
Helicobacter pylori is a gram-negative bacterium found on the luminal surface of the gastric mucosa in at least 50% of the world's human population. The protective effect of breastfeeding against H. pylori infection has been extensively reported; however, the mechanisms behind this protection remain poorly understood. Human IgA from colostrum has reactivity against H. pylori antigens. Despite that IgA1 and IgA2 display structural and functional differences, their reactivity against H. pylori had not been previously determined. We attested titers and reactivity of human colostrum-IgA subclasses by ELISA, immunoblot, and flow cytometry. Colostrum samples from healthy mothers had higher titers of IgA; and IgA1 mostly recognized H. pylori antigens. Moreover, we found a correlation between IgA1 reactivity and their neutralizing effect determined by inhibition of cytoskeletal changes in AGS cells infected with H. pylori. In conclusion, colostrum-IgA reduces H. pylori infection of epithelial gastric cells, suggesting an important role in preventing the bacteria establishment during the first months of life. As a whole, these results suggest that IgA1 from human colostrum provides protection that may help in the development of the mucosal immune system of newborn children. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Shantal Lizbeth Baltierra-Uribe
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Alejandra Montañez-Barragán
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Héctor Romero-Ramírez
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Ksenia Klimov-Kravtchenko
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico.,University Center of Biological and Agricultural Sciences (CUCBA), University of Guadalajara, Zapopan, Jalisco, Mexico
| | - Karla Ivette Martínez-Pedro
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico.,"La Cañada" University (UNCA), Oaxaca, Mexico
| | - Erick Sánchez-Salguero
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Margarita Camorlinga-Ponce
- Infectious and Parasitic Disease Medical Research Unit (UIMEIP) at Pediatric Hospital in National Medical Center (CMN- SIGLO XXI) Mexican Institute of Social Security (IMSS), Mexico City, Mexico
| | - Javier Torres
- Infectious and Parasitic Disease Medical Research Unit (UIMEIP) at Pediatric Hospital in National Medical Center (CMN- SIGLO XXI) Mexican Institute of Social Security (IMSS), Mexico City, Mexico
| | - Leopoldo Santos-Argumedo
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| |
Collapse
|
20
|
Taxauer K, Hamway Y, Ralser A, Dietl A, Mink K, Vieth M, Singer BB, Gerhard M, Mejías-Luque R. Engagement of CEACAM1 by Helicobacterpylori HopQ Is Important for the Activation of Non-Canonical NF-κB in Gastric Epithelial Cells. Microorganisms 2021; 9:1748. [PMID: 34442827 PMCID: PMC8400456 DOI: 10.3390/microorganisms9081748] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/04/2021] [Accepted: 08/10/2021] [Indexed: 01/01/2023] Open
Abstract
The gastric pathogen Helicobacter pylori infects half of the world's population and is a major risk factor for gastric cancer development. In order to attach to human gastric epithelial cells and inject the oncoprotein CagA into host cells, H. pylori utilizes the outer membrane protein HopQ that binds to the cell surface protein CEACAM, which can be expressed on the gastric mucosa. Once bound, H. pylori activates a number of signaling pathways, including canonical and non-canonical NF-κB. We investigated whether HopQ-CEACAM interaction is involved in activating the non-canonical NF-κB signaling pathway. Different gastric cancer cells were infected with the H. pylori wild type, or HopQ mutant strains, and the activation of non-canonical NF-κB was related to CEACAM expression levels. The correlation between CEACAM levels and the activation of non-canonical NF-κB was confirmed in human gastric tissue samples. Taken together, our findings show that the HopQ-CEACAM interaction is important for activation of the non-canonical NF-κB pathway in gastric epithelial cells.
Collapse
Affiliation(s)
- Karin Taxauer
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University Munich, 81675 Munich, Germany; (K.T.); (Y.H.); (A.R.); (A.D.); (K.M.); (M.G.)
| | - Youssef Hamway
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University Munich, 81675 Munich, Germany; (K.T.); (Y.H.); (A.R.); (A.D.); (K.M.); (M.G.)
| | - Anna Ralser
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University Munich, 81675 Munich, Germany; (K.T.); (Y.H.); (A.R.); (A.D.); (K.M.); (M.G.)
| | - Alisa Dietl
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University Munich, 81675 Munich, Germany; (K.T.); (Y.H.); (A.R.); (A.D.); (K.M.); (M.G.)
| | - Karin Mink
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University Munich, 81675 Munich, Germany; (K.T.); (Y.H.); (A.R.); (A.D.); (K.M.); (M.G.)
| | - Michael Vieth
- Institute of Pathology, Friedrich-Alexander University Erlangen-Nuremberg, Klinikum Bayreuth, 95445 Bayreuth, Germany;
| | - Bernhard B. Singer
- Institute of Anatomy, Medical Faculty, University of Duisburg-Essen, 45147 Essen, Germany;
| | - Markus Gerhard
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University Munich, 81675 Munich, Germany; (K.T.); (Y.H.); (A.R.); (A.D.); (K.M.); (M.G.)
| | - Raquel Mejías-Luque
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University Munich, 81675 Munich, Germany; (K.T.); (Y.H.); (A.R.); (A.D.); (K.M.); (M.G.)
| |
Collapse
|
21
|
Helicobacter pylori BabA-SabA Key Roles in the Adherence Phase: The Synergic Mechanism for Successful Colonization and Disease Development. Toxins (Basel) 2021; 13:toxins13070485. [PMID: 34357957 PMCID: PMC8310295 DOI: 10.3390/toxins13070485] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/03/2021] [Accepted: 07/07/2021] [Indexed: 01/21/2023] Open
Abstract
Helicobacter pylori is a pathogenic microorganism that successfully inhabits the human stomach, colonizing it by producing several virulence factors responsible for preventing host self-defense mechanisms. The adherence mechanism to gastric mucosal tissue is one of the most important processes for effective colonization in the stomach. The blood group antigen-binding adhesion (BabA) and sialic acid-binding adherence (SabA) are two H. pylori outer membrane proteins able to interact with antigens in the gastroduodenal tract. H. pylori possesses several mechanisms to control the regulation of both BabA and SabA in either the transcriptional or translational level. BabA is believed to be the most important protein in the early infection phase due to its ability to interact with various Lewis antigens, whereas SabA interaction with sialylated Lewis antigens may prove important for the adherence process in the inflamed gastric mucosal tissue in the ongoing-infection phase. The adherence mechanisms of BabA and SabA allow H. pylori to anchor in the gastric mucosa and begin the colonization process.
Collapse
|
22
|
Knorr J, Sharafutdinov I, Fiedler F, Soltan Esmaeili D, Rohde M, Rottner K, Backert S, Tegtmeyer N. Cortactin Is Required for Efficient FAK, Src and Abl Tyrosine Kinase Activation and Phosphorylation of Helicobacter pylori CagA. Int J Mol Sci 2021; 22:ijms22116045. [PMID: 34205064 PMCID: PMC8199859 DOI: 10.3390/ijms22116045] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/28/2021] [Accepted: 06/01/2021] [Indexed: 12/11/2022] Open
Abstract
Cortactin is a well-known regulatory protein of the host actin cytoskeleton and represents an attractive target of microbial pathogens like Helicobacter pylori. H. pylori manipulates cortactin's phosphorylation status by type-IV secretion-dependent injection of its virulence protein CagA. Multiple host tyrosine kinases, like FAK, Src, and Abl, are activated during infection, but the pathway(s) involved is (are) not yet fully established. Among them, Src and Abl target CagA and stimulate tyrosine phosphorylation of the latter at its EPIYA-motifs. To investigate the role of cortactin in more detail, we generated a CRISPR/Cas9 knockout of cortactin in AGS gastric epithelial cells. Surprisingly, we found that FAK, Src, and Abl kinase activities were dramatically downregulated associated with widely diminished CagA phosphorylation in cortactin knockout cells compared to the parental control. Together, we report here a yet unrecognized cortactin-dependent signaling pathway involving FAK, Src, and Abl activation, and controlling efficient phosphorylation of injected CagA during infection. Thus, the cortactin status could serve as a potential new biomarker of gastric cancer development.
Collapse
Affiliation(s)
- Jakob Knorr
- Department of Biology, Division of Microbiology, Friedrich-Alexander University of Erlangen-Nuremberg, 91058 Erlangen, Germany; (J.K.); (I.S.); (F.F.); (D.S.E.); (S.B.)
| | - Irshad Sharafutdinov
- Department of Biology, Division of Microbiology, Friedrich-Alexander University of Erlangen-Nuremberg, 91058 Erlangen, Germany; (J.K.); (I.S.); (F.F.); (D.S.E.); (S.B.)
| | - Florian Fiedler
- Department of Biology, Division of Microbiology, Friedrich-Alexander University of Erlangen-Nuremberg, 91058 Erlangen, Germany; (J.K.); (I.S.); (F.F.); (D.S.E.); (S.B.)
| | - Delara Soltan Esmaeili
- Department of Biology, Division of Microbiology, Friedrich-Alexander University of Erlangen-Nuremberg, 91058 Erlangen, Germany; (J.K.); (I.S.); (F.F.); (D.S.E.); (S.B.)
| | - Manfred Rohde
- Central Facility for Microscopy, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany;
| | - Klemens Rottner
- Department of Cell Biology, Helmholtz Centre for Infection Research, 38124 Braunschweig, Germany;
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, 38106 Braunschweig, Germany
| | - Steffen Backert
- Department of Biology, Division of Microbiology, Friedrich-Alexander University of Erlangen-Nuremberg, 91058 Erlangen, Germany; (J.K.); (I.S.); (F.F.); (D.S.E.); (S.B.)
| | - Nicole Tegtmeyer
- Department of Biology, Division of Microbiology, Friedrich-Alexander University of Erlangen-Nuremberg, 91058 Erlangen, Germany; (J.K.); (I.S.); (F.F.); (D.S.E.); (S.B.)
- Correspondence:
| |
Collapse
|
23
|
CagL polymorphisms between East Asian and Western Helicobacter pylori are associated with different abilities to induce IL-8 secretion. J Microbiol 2021; 59:763-770. [PMID: 34061339 DOI: 10.1007/s12275-021-1136-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 12/17/2022]
Abstract
Helicobacter pylori colonizes human gastric mucosa. Its infection is associated with gastric diseases including gastric cancer. CagA is one of the most important toxins produced by H. pylori. It is related to gastric cancer which can be injected into host cells via a type IV secretion system (T4SS). CagL is a structural component of T4SS apparatus, which triggers host cell signaling pathway. It has been reported that CagL polymorphisms may influence the severity of disease development. To explore the contribution of CagL polymorphisms between East Asian and Western H. pylori in pathogenesis, cagL gene in G27 H. pylori was swapped by K74 cagL which is identical to East Asian CagL consensus sequence and by Western 26695 H. pylori, resulting in G27 ΔcagL/cagLK74 and G27 ΔcagL/cagL26695, respectively. Intriguingly, G27 ΔcagL/cagLK74 showed significantly less ability of IL-8 induction than G27 ΔcagL/cagL26695 while displayed similar abilities of CagA phosphorylation, and cell elongation. Taken together, this study suggests that the CagL polymorphism may influence IL-8 induction, and K74 CagL has less ability to induce IL-8 secretion than G27 or 26695 CagL. Further research should address how the different capabilities of IL-8 induction between intraspecies-CagL are associated with the large differences of the incidence of gastric cancer between East Asian and Western countries.
Collapse
|
24
|
Do AD, Chang CC, Su CH, Hsu YM. Lactobacillus rhamnosus JB3 inhibits Helicobacter pylori infection through multiple molecular actions. Helicobacter 2021; 26:e12806. [PMID: 33843101 DOI: 10.1111/hel.12806] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/22/2021] [Accepted: 03/26/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Eradication of Helicobacter pylori infection is the most direct and effective way for preventing gastric cancer. Lactic acid bacteria are considered as alternative therapeutic agents against H. pylori infection. METHODS Effects of Lactobacillus rhamnosus JB3 (LR-JB3) on the virulence gene expression of H. pylori and infection-induced cellular responses of AGS cells were investigated by co-cultivating infected AGS cells with different multiplicity of infections (MOIs) of LR-JB3. RESULTS LR-JB3, specifically at a MOI of 25, suppressed the association ability of H. pylori and its induced IL-8 levels, as well as the mRNA levels of vacA, sabA, and fucT of H. pylori, infection-induced Lewis (Le)x antigen and Toll-like receptor 4 (TLR4) expressions in AGS cells. However, the apoptosis mediated by infection was inhibited by LR-JB3 in a dose-dependent manner. In addition, autoinducer (AI)-2 was observed to have increased the association ability and fucT expression of H. pylori, and Lex antigen and TLR4 expression of AGS cells. Interestingly, an unknown bioactive cue was hypothesized to have been secreted from LR-JB3 at a MOI of 25 to act as an antagonist of AI-2. CONCLUSIONS LR-JB3 possesses various means to interfere with H. pylori pathogenesis and infection-induced cellular responses of AGS cells to fight against infection.
Collapse
Affiliation(s)
- Anh Duy Do
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Chun-Chi Chang
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Chiu-Hsian Su
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Yuan-Man Hsu
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| |
Collapse
|
25
|
Qaria MA, Qumar S, Sepe LP, Ahmed N. Cholesterol glucosylation-based survival strategy in Helicobacter pylori. Helicobacter 2021; 26:e12777. [PMID: 33368895 DOI: 10.1111/hel.12777] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/09/2020] [Accepted: 11/12/2020] [Indexed: 12/14/2022]
Abstract
Helicobacter pylori is a major chronic health problem, infecting more than half of the population worldwide. H. pylori infection is linked with various clinical complications ranging from gastritis to gastric cancer. The resolution of gastritis and peptic ulcer appears to be linked with the eradication of H. pylori. However, resistance to antibiotics and eradication failure rates are reaching alarmingly high levels. This calls for urgent action in finding alternate methods for H. pylori eradication. Here, we discuss the recently identified mechanism of H. pylori known as cholesterol glucosylation, mediated by the enzyme cholesterol-α-glucosyltransferase, encoded by the gene cgt. Cholesterol glucosylation serves several functions that include promoting immune evasion, enhancing antibiotic resistance, maintaining the native helical morphology, and supporting functions of prominent virulence factors such as CagA and VacA. Consequently, strategies aiming at inhibition of the cholesterol glucosylation process have the potential to attenuate the potency of H. pylori infection and abrogate H. pylori immune evasion capabilities. Knockout of H. pylori cgt results in unsuccessful colonization and elimination by the host immune responses. Moreover, blocking cholesterol glucosylation can reverse antibiotic susceptibility in H. pylori. In this work, we review the main roles of cholesterol glucosylation in H. pylori and evaluate whether this mechanism can be targeted for the development of alternate methods for eradication of H. pylori infection.
Collapse
Affiliation(s)
- Majjid A Qaria
- Pathogen Biology Laboratory, Department of Biotechnology and Bioinformatics, University of Hyderabad, Hyderabad, India
| | - Shamsul Qumar
- Pathogen Biology Laboratory, Department of Biotechnology and Bioinformatics, University of Hyderabad, Hyderabad, India
| | - Ludovico P Sepe
- Department of Biological Safety, German Federal Institute for Risk Assessment (BfR), Berlin, Germany
| | - Niyaz Ahmed
- Pathogen Biology Laboratory, Department of Biotechnology and Bioinformatics, University of Hyderabad, Hyderabad, India
| |
Collapse
|
26
|
Traulsen J, Zagami C, Daddi AA, Boccellato F. Molecular modelling of the gastric barrier response, from infection to carcinogenesis. Best Pract Res Clin Gastroenterol 2021; 50-51:101737. [PMID: 33975688 DOI: 10.1016/j.bpg.2021.101737] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 02/15/2021] [Accepted: 02/18/2021] [Indexed: 02/07/2023]
Abstract
The lining of the stomach is a tight monolayer of epithelial cells performing functions in digestion and a protective barrier against gastric acid, toxic metabolites and infectious agents, including Helicobacter pylori. The response of the epithelial barrier to infections underlies gastric pathologies, including gastric cancer. H. pylori has the unique capacity to colonise the gastric mucosa while evading the immune system. The colonised mucosa initiates an inflammatory response to fight the infection and a strong regenerative program to avoid barrier failure and ulceration. This response changes the morphology and cell composition of the gastric epithelium and in parallel it might contribute to the accumulation of somatic mutations leading to cellular transformation. Genetically modified mice, cell lines and human-derived organoids are the main biological models to study the gastric epithelial barrier. With these models it is possible to dissect the stepwise process of tissue adaptation to infection that places the epithelium at risk of malignant transformation.
Collapse
Affiliation(s)
- Jan Traulsen
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, United Kingdom.
| | - Claudia Zagami
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, United Kingdom.
| | - Alice Anna Daddi
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, United Kingdom.
| | - Francesco Boccellato
- Ludwig Institute for Cancer Research, Nuffield Department of Medicine, University of Oxford, United Kingdom.
| |
Collapse
|
27
|
de Oliveira IA, Corvelo TCDO. ABH and Lewis blood group systems and their relation to diagnosis and risk of Helicobacter pylori infection. Microb Pathog 2020; 152:104653. [PMID: 33253856 DOI: 10.1016/j.micpath.2020.104653] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 11/01/2020] [Accepted: 11/23/2020] [Indexed: 11/18/2022]
Abstract
BACKGROUND Helicobacter pylori infection occurs in 50% of the world's population and represents a major risk factor for chronic gastritis, gastroduodenal ulcer and gastric cancer in developed and developing countries. The distribution of H. pylori virulence factors is diverse and varies geographically, such as the CagA and VacA genes, which have revealed association with disease status. Some findings show increased frequencies of these diseases in O Le (a-b +) and A Le (a-b +) blood type individuals, but other studies not found any relationship between these blood groups and H. pylori infection. AIM This study aimed to elucidate probable controversies described in the relationship between the ABH/Lewis blood groups and H. pylori, contributing to the severity of gastric diseases in northern the population of Belém -Pará.-Brazil. METHODS This cross-sectional study included 288 samples of patients separate into two groups with gastric cancer and chronic gastritis. Blood, saliva, and gastric biopsy were analyzed using modified Gram and hematoxylin-eosin staining techniques, the enzyme immunoassay Elisa and Multiplex PCR. The antigens expression of ABH and Lewis systems was determined through Dot-ELISA and direct hemagglutination. Proportions were compared in univariate analysis, while the relation between putative risk factors including H. pylori status and ABO/Lewis phenotype was performed using multivariable logistic regression analyses, P-value < 0.05 was considered significant. RESULTS The findings of this study demonstrate that the likelihood of developing gastric cancer increases threefold if the individual is from A1 Le (a-b +) blood group, has premalignant changes, and infection with H. pylori virulent strains (cagA+/vacA + s1m1). CONCLUSION Therefore, this study found a significant association between ABO and Lewis phenotypes and H. pylori cagA status into the relevance of the development of gastric carcinogenesis.
Collapse
|
28
|
Resuscitation of the Helicobacter pylori Coccoid Forms by Resuscitation Promoter Factor Obtained from Micrococcus Luteus. Curr Microbiol 2020; 77:2093-2103. [DOI: 10.1007/s00284-020-02043-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 05/20/2020] [Indexed: 02/07/2023]
|
29
|
Prevalence of the Helicobacter pylori babA2 Gene in Children Mainly Depends on the PCR Primer Set Used. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2020; 2020:4080248. [PMID: 32855749 PMCID: PMC7443014 DOI: 10.1155/2020/4080248] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/20/2020] [Accepted: 07/28/2020] [Indexed: 02/07/2023]
Abstract
Various polymerase chain reaction- (PCR-) based methods with varying positivity rates were designed to detect the Helicobacter pylori babA2 gene. To compare different primer sets, babA2 prevalence was determined in 279 H. pylori-positive pediatric samples using the 832 bp, 139 bp, and 271 bp PCR primer sets, resulting in 34.0%, 51.3%, and 79.6% prevalence of the babA2 gene, respectively. The babA2 status determined using the 832 bp and 139 bp PCR primer sets significantly correlated with bacterial density and activity of inflammation, whereas no such correlations were found using the 271 bp PCR primer set. The 139 and 832 bp PCR primer sets concordantly detected the babA2 gene in 93 cases; however, in comparison to the 832 bp PCR primer set, the 139 bp PCR primer set detected additional 50 babA2 cases, whereas only two 832 bp positive cases were missed. The 271 bp PCR primer set missed 32 babA2 cases that were 832 bp and/or 139 bp PCR positive, but tested solely positive in 109 cases. Interestingly, cloning of a subset of 271 bp PCR positive samples revealed amplification of the babA/B gene chimera. Hence, in our opinion, the 271 bp PCR protocol is not a reliable diagnostic tool for detecting the babA2 gene in children. Our results reaffirm previous observations that the use of certain babA2 PCR primer sets can significantly impact estimation of the prevalence and clinical relevance of the H. pylori babA2 gene in children, suggesting babA2 detection methods should be carefully selected.
Collapse
|
30
|
Sukhan DS, Vernygorodskyi SV, Haidukov NV, Ludkevich HP. Molecular and Genetic Aspects of Helicobacter pylori Interaction with Cells of Gastric Mucosa. CYTOL GENET+ 2020. [DOI: 10.3103/s0095452720020139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
31
|
Wizenty J, Tacke F, Sigal M. Responses of gastric epithelial stem cells and their niche to Helicobacter pylori infection. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:568. [PMID: 32775369 PMCID: PMC7347775 DOI: 10.21037/atm.2020.02.178] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Helicobacter pylori (H. pylori) are gram-negative bacteria that are able to colonize and persist in the stomach. Gastric cancer is tightly linked to chronic infection with this bacterium. Research over the last decades has illuminated the molecular interactions between H. pylori and host cells. It is now well established that H. pylori have multiple sophisticated means to adhere to epithelial cells and to manipulate their behavior. This interaction with the epithelium can lead to altered cell signaling, DNA damage and aberrant epithelial immunity. H. pylori are known to colonize the mucus layer of the stomach and surface epithelial cells. In addition, it has recently become clear that they can also penetrate the glands and directly interact with specialized epithelial cells deep in the glands. Understanding the biogeography of infection is important because gastric epithelial glands are composed of various types of short-lived differentiated cells that are constantly regenerated by a limited pool of long-lived stem cells located in base of gastric glands. Recent advances in gastric stem cell research not only led to identification of stem cell populations using specific markers but has also uncovered specific regulatory pathways and principles that govern gastric stem cell behavior and regeneration. Particularly, the stem cell state is largely dependent on signals from the niche cells that surround the stem cell compartment. The subpopulation of H. pylori that colonizes in the stem cell compartment triggers specific inflammatory responses and drives epithelial pathology. Colonization of gastric glands induces responses of the stem cell niche, simultaneously enhancing the cell turnover kinetics and driving the formation of antimicrobial cells in the gland base. These data reveal the high plasticity of the epithelium and its ability to adapt to the environment, which is necessary to regenerate and counterbalance infection, but simultaneously lays the grounds for development of gastric pathology and carcinogenesis.
Collapse
Affiliation(s)
- Jonas Wizenty
- Department of Hepatology & Gastroenterology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Frank Tacke
- Department of Hepatology & Gastroenterology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Michael Sigal
- Department of Hepatology & Gastroenterology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| |
Collapse
|
32
|
Jan HM, Chen YC, Yang TC, Ong LL, Chang CC, Muthusamy S, Abera AB, Wu MS, Gervay-Hague J, Mong KKT, Lin CH. Cholesteryl α-D-glucoside 6-acyltransferase enhances the adhesion of Helicobacter pylori to gastric epithelium. Commun Biol 2020; 3:120. [PMID: 32170208 PMCID: PMC7069968 DOI: 10.1038/s42003-020-0855-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 02/20/2020] [Indexed: 12/18/2022] Open
Abstract
Helicobacter pylori, the most common etiologic agent of gastric diseases including gastric cancer, is auxotrophic for cholesterol and has to hijack it from gastric epithelia. Upon uptake, the bacteria convert cholesterol to cholesteryl 6′-O-acyl-α-D-glucopyranoside (CAG) to promote lipid raft clustering in the host cell membranes. However, how CAG appears in the host to exert the pathogenesis still remains ambiguous. Herein we identified hp0499 to be the gene of cholesteryl α-D-glucopyranoside acyltransferase (CGAT). Together with cholesteryl glucosyltransferase (catalyzing the prior step), CGAT is secreted via outer membrane vesicles to the host cells for direct synthesis of CAG. This significantly enhances lipid rafts clustering, gathers adhesion molecules (including Lewis antigens and integrins α5, β1), and promotes more bacterial adhesion. Furthermore, the clinically used drug amiodarone was shown as a potent inhibitor of CGAT to effectively reduce the bacterial adhesion, indicating that CGAT is a potential target of therapeutic intervention. Jan et al. identify cholesteryl α-D- glucopyranoside acyltransferase as a key enzyme in Helicobacter pylori’s synthesis of cholesteryl 6’-O-acyl-α-D-glucopyranoside, which promotes bacterial adhesion. This study provides insights into the H. pylori-induced pathogenesis and therapeutic strategies against it.
Collapse
Affiliation(s)
- Hau-Ming Jan
- Institute of Biological Chemistry, Academia Sinica, No. 128 Academic Road Section 2, Nan-Kang, Taipei, 11529, Taiwan
| | - Yi-Chi Chen
- Institute of Biological Chemistry, Academia Sinica, No. 128 Academic Road Section 2, Nan-Kang, Taipei, 11529, Taiwan.,Department of Chemistry and Institute of Biochemical Sciences, National Taiwan University, Taipei, 10617, Taiwan
| | - Tsai-Chen Yang
- Institute of Biological Chemistry, Academia Sinica, No. 128 Academic Road Section 2, Nan-Kang, Taipei, 11529, Taiwan
| | - Lih-Lih Ong
- Institute of Biological Chemistry, Academia Sinica, No. 128 Academic Road Section 2, Nan-Kang, Taipei, 11529, Taiwan.,Department of Applied Chemistry, National Chiao Tung University, Hsin-Chu, 30010, Taiwan.,Sustainable Chemical Science and Technology, Taiwan International Graduate Program, Academia Sinica and National Chiao Tung University, Taipei, 11529, Taiwan
| | - Chia-Chen Chang
- Department of Applied Chemistry, National Chiao Tung University, Hsin-Chu, 30010, Taiwan
| | - Sasikala Muthusamy
- Institute of Biological Chemistry, Academia Sinica, No. 128 Academic Road Section 2, Nan-Kang, Taipei, 11529, Taiwan.,Molecular and Biological Agricultural Sciences, Taiwan International Graduate Program, Academia Sinica and National Chung-Hsing University, Taipei, 11529, Taiwan.,Graduate Institute of Biotechnology, National Chung-Hsing University, Taichung, 40227, Taiwan
| | - Andualem Bahiru Abera
- Institute of Biological Chemistry, Academia Sinica, No. 128 Academic Road Section 2, Nan-Kang, Taipei, 11529, Taiwan.,Molecular and Biological Agricultural Sciences, Taiwan International Graduate Program, Academia Sinica and National Chung-Hsing University, Taipei, 11529, Taiwan.,Graduate Institute of Biotechnology, National Chung-Hsing University, Taichung, 40227, Taiwan
| | - Ming-Shiang Wu
- Division of Gastroenterology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, 10002, Taiwan
| | | | - Kwok-Kong Tony Mong
- Department of Applied Chemistry, National Chiao Tung University, Hsin-Chu, 30010, Taiwan.
| | - Chun-Hung Lin
- Institute of Biological Chemistry, Academia Sinica, No. 128 Academic Road Section 2, Nan-Kang, Taipei, 11529, Taiwan. .,Department of Chemistry and Institute of Biochemical Sciences, National Taiwan University, Taipei, 10617, Taiwan.
| |
Collapse
|
33
|
Kashyap D, Baral B, Verma TP, Sonkar C, Chatterji D, Jain AK, Jha HC. Oral rinses in growth inhibition and treatment of Helicobacter pylori infection. BMC Microbiol 2020; 20:45. [PMID: 32131741 PMCID: PMC7055109 DOI: 10.1186/s12866-020-01728-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Accepted: 02/14/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Helicobacter pylori (H. pylori) is well-known for its role in chronic gastritis and gastric cancer. Eradication of these carcinogenic bacteria from the gut is one of the challenges for clinicians. The complexity of treatment mainly owes to antibiotic resistance and relapse due to an additional reservoir in the oral cavity. Our study emphases the isolation of H. pylori from distinct habitats of the gut microenvironment (gastric biopsy and gastric juice) and its subsequent characterization. We have also evaluated the effect of various oral rinses on isolated H. pylori from different anatomical locations of included subjects. RESULTS The possible strains isolated from two different habitats of the same subject shows a striking difference in their growth pattern. Promisingly, some of the included oral rinses are efficient in growth inhibition as per recommended 30 s treatment. The subsequent evaluation shows that oral rinse B (among A-E) is most effective and down-regulates the expression of one of the potent H. pylori gene, CagA, in the infected gastric adenocarcinoma (AGS) cells. CONCLUSION Our study, for the first time, revealed that H. pylori, isolated from the different habitat of the same subject, show a different growth pattern. The expression of H. pylori pathogenic gene (CagA) was down-regulated by the use of oral rinses. Hence, oral rinses will reduce the H. pylori in the oral cavity and help to control its migration from oral to the gastric compartment and may be used as an adjuvant treatment option for its re-infection.
Collapse
Affiliation(s)
- Dharmendra Kashyap
- Discipline of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, 453552, Indore, Madhya Pradesh, India
| | - Budhadev Baral
- Discipline of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, 453552, Indore, Madhya Pradesh, India
| | - Tarun Prakash Verma
- Discipline of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, 453552, Indore, Madhya Pradesh, India
| | - Charu Sonkar
- Discipline of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, 453552, Indore, Madhya Pradesh, India
| | - Debi Chatterji
- Choithram Hospital and Research Centre, 452014, Indore, Madhya Pradesh, India
| | - Ajay Kumar Jain
- Choithram Hospital and Research Centre, 452014, Indore, Madhya Pradesh, India
| | - Hem C Jha
- Discipline of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, 453552, Indore, Madhya Pradesh, India.
| |
Collapse
|
34
|
O’Callaghan AA, Corr SC. Establishing Boundaries: The Relationship That Exists between Intestinal Epithelial Cells and Gut-Dwelling Bacteria. Microorganisms 2019; 7:microorganisms7120663. [PMID: 31818022 PMCID: PMC6956261 DOI: 10.3390/microorganisms7120663] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 12/05/2019] [Accepted: 12/07/2019] [Indexed: 12/16/2022] Open
Abstract
The human gastrointestinal (GI) tract is a highly complex organ in which various dynamic physiological processes are tightly coordinated while interacting with a complex community of microorganisms. Within the GI tract, intestinal epithelial cells (IECs) create a structural interface that separates the intestinal lumen from the underlying lamina propria. In the lumen, gut-dwelling microbes play an essential role in maintaining gut homeostasis and functionality. Whether commensal or pathogenic, their interaction with IECs is inevitable. IECs and myeloid immune cells express an array of pathogen recognition receptors (PRRs) that define the interaction of both pathogenic and beneficial bacteria with the intestinal mucosa and mount appropriate responses including induction of barrier-related factors which enhance the integrity of the epithelial barrier. Indeed, the integrity of this barrier and induction of appropriate immune responses is critical to health status, with defects in this barrier and over-activation of immune cells by invading microbes contributing to development of a range of inflammatory and infectious diseases. This review describes the complexity of the GI tract and its interactions with gut bacteria.
Collapse
|
35
|
Molecular anatomy and pathogenic actions of Helicobacter pylori CagA that underpin gastric carcinogenesis. Cell Mol Immunol 2019; 17:50-63. [PMID: 31804619 PMCID: PMC6952403 DOI: 10.1038/s41423-019-0339-5] [Citation(s) in RCA: 133] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 11/13/2019] [Accepted: 11/13/2019] [Indexed: 12/15/2022] Open
Abstract
Chronic infection with Helicobacter pylori cagA-positive strains is the strongest risk factor for gastric cancer. The cagA gene product, CagA, is delivered into gastric epithelial cells via the bacterial type IV secretion system. Delivered CagA then undergoes tyrosine phosphorylation at the Glu-Pro-Ile-Tyr-Ala (EPIYA) motifs in its C-terminal region and acts as an oncogenic scaffold protein that physically interacts with multiple host signaling proteins in both tyrosine phosphorylation-dependent and -independent manners. Analysis of CagA using in vitro cultured gastric epithelial cells has indicated that the nonphysiological scaffolding actions of CagA cell-autonomously promote the malignant transformation of the cells by endowing the cells with multiple phenotypic cancer hallmarks: sustained proliferation, evasion of growth suppressors, invasiveness, resistance to cell death, and genomic instability. Transgenic expression of CagA in mice leads to in vivo oncogenic action of CagA without any overt inflammation. The in vivo oncogenic activity of CagA is further potentiated in the presence of chronic inflammation. Since Helicobacter pylori infection triggers a proinflammatory response in host cells, a feedforward stimulation loop that augments the oncogenic actions of CagA and inflammation is created in CagA-injected gastric mucosa. Given that Helicobacter pylori is no longer colonized in established gastric cancer lesions, the multistep nature of gastric cancer development should include a “hit-and-run” process of CagA action. Thus, acquisition of genetic and epigenetic alterations that compensate for CagA-directed cancer hallmarks may be required for completion of the “hit-and-run” process of gastric carcinogenesis.
Collapse
|
36
|
Activity and Functional Importance of Helicobacter pylori Virulence Factors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1149:35-56. [PMID: 31016624 DOI: 10.1007/5584_2019_358] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Helicobacter pylori is a very successful Gram-negative pathogen colonizing the stomach of humans worldwide. Infections with this bacterium can generate pathologies ranging from chronic gastritis and peptic ulceration to gastric cancer. The best characterized H. pylori virulence factors that cause direct cell damage include an effector protein encoded by the cytotoxin-associated gene A (CagA), a type IV secretion system (T4SS) encoded in the cag-pathogenicity island (cag PAI), vacuolating cytotoxin A (VacA), γ-glutamyl transpeptidase (GGT), high temperature requirement A (HtrA, a serine protease) and cholesterol glycosyl-transferase (CGT). Since these H. pylori factors are either surface-exposed, secreted or translocated, they can directly interact with host cell molecules and are able to hijack cellular functions. Studies on these bacterial factors have progressed substantially in recent years. Here, we review the current status in the characterization of signaling cascades by these factors in vivo and in vitro, which comprise the disruption of cell-to-cell junctions, induction of membrane rearrangements, cytoskeletal dynamics, proliferative, pro-inflammatory, as well as, pro-apoptotic and anti-apoptotic responses or immune evasion. The impact of these signal transduction modules in the pathogenesis of H. pylori infections is discussed.
Collapse
|
37
|
Blaser N, Backert S, Pachathundikandi SK. Immune Cell Signaling by Helicobacter pylori: Impact on Gastric Pathology. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1149:77-106. [PMID: 31049845 DOI: 10.1007/5584_2019_360] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Helicobacter pylori represents a highly successful colonizer of the human stomach. Infections with this Gram-negative bacterium can persist lifelong, and although in the majority of cases colonization is asymptomatic, it can trigger pathologies ranging from chronic gastritis and peptic ulceration to gastric cancer. The interaction of the bacteria with the human host modulates immune responses in different ways to enable bacterial survival and persistence. H. pylori uses various pathogenicity-associated factors such as VacA, NapA, CGT, GGT, lipopolysaccharide, peptidoglycan, heptose 1,7-bisphosphate, ADP-heptose, cholesterol glucosides, urease and a type IV secretion system for controlling immune signaling and cellular functions. It appears that H. pylori manipulates multiple extracellular immune receptors such as integrin-β2 (CD18), EGFR, CD74, CD300E, DC-SIGN, MINCLE, TRPM2, T-cell and Toll-like receptors as well as a number of intracellular receptors including NLRP3, NOD1, NOD2, TIFA and ALPK1. Consequently, downstream signaling pathways are hijacked, inducing tolerogenic dendritic cells, inhibiting effector T cell responses and changing the gastrointestinal microbiota. Here, we discuss in detail the interplay of bacterial factors with multiple immuno-regulatory cells and summarize the main immune evasion and persistence strategies employed by H. pylori.
Collapse
Affiliation(s)
- Nicole Blaser
- Department of Biology, Institute for Microbiology, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Steffen Backert
- Department of Biology, Institute for Microbiology, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - Suneesh Kumar Pachathundikandi
- Department of Biology, Institute for Microbiology, Friedrich Alexander University Erlangen-Nuremberg, Erlangen, Germany.
| |
Collapse
|
38
|
Hu W, Chan H, Lu L, Wong KT, Wong SH, Li MX, Xiao ZG, Cho CH, Gin T, Chan MTV, Wu WKK, Zhang L. Autophagy in intracellular bacterial infection. Semin Cell Dev Biol 2019; 101:41-50. [PMID: 31408699 DOI: 10.1016/j.semcdb.2019.07.014] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 06/06/2019] [Accepted: 07/30/2019] [Indexed: 12/11/2022]
Abstract
Autophagy is a conserved intracellular degradation process enclosing the bulk of cytosolic components for lysosomal degradation to maintain cellular homeostasis. Accumulating evidences showed that a specialized form of autophagy, known as xenophagy, could serve as an innate immune response to defend against pathogens invading inside the host cells. Correspondingly, infectious pathogens have developed a variety of strategies to disarm xenophagy, leading to a prolonged and persistent intracellular colonization. In this review, we first summarize the current knowledge about the general mechanisms of intracellular bacterial infections and xenophagy. We then focus on the ongoing battle between these two processes.
Collapse
Affiliation(s)
- Wei Hu
- Department of Gastroenterology, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, PR China; Department of Anesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong, China
| | - Hung Chan
- Department of Anesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong, China
| | - Lan Lu
- Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, Sichuan, PR China
| | - Kam Tak Wong
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong, China
| | - Sunny H Wong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China; State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, and Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong, China
| | - Ming X Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, PR China
| | - Zhan G Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, PR China
| | - Chi H Cho
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, PR China
| | - Tony Gin
- Department of Anesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong, China
| | - Matthew T V Chan
- Department of Anesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong, China.
| | - William K K Wu
- Department of Anesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong, China; State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, and Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong, China.
| | - Lin Zhang
- Department of Anesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong, China; State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Sciences, and Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
39
|
Marinoni C, Ribaldone DG, Rosso C, Astegiano M, Caviglia GP. Diagnosis of Helicobacter pylori infection: a look into molecular aspects of urea breath test. MINERVA BIOTECNOL 2019. [DOI: 10.23736/s1120-4826.19.02555-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
40
|
Tserentogtokh T, Gantuya B, Subsomwong P, Oyuntsetseg K, Bolor D, Erdene-Ochir Y, Azzaya D, Davaadorj D, Uchida T, Matsuhisa T, Yamaoka Y. Western-Type Helicobacter pylori CagA are the Most Frequent Type in Mongolian Patients. Cancers (Basel) 2019; 11:cancers11050725. [PMID: 31137742 PMCID: PMC6562502 DOI: 10.3390/cancers11050725] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 04/27/2019] [Accepted: 05/22/2019] [Indexed: 12/21/2022] Open
Abstract
Helicobacter pylori infection possessing East-Asian-type CagA is associated with carcinogenesis. Mongolia has the highest mortality rate from gastric cancer. Therefore, we evaluated the CagA status in the Mongolian population. High risk and gastric cancer patients were determined using endoscopy and histological examination. H. pylori strains were isolated from different locations in Mongolia. The CagA subtypes (East-Asian-type or Western-type, based on sequencing of Glu-Pro-Ile-Tyr-Ala (EPIYA) segments) and vacA genotypes (s and m regions) were determined using PCR-based sequencing and PCR, respectively. In total, 368 patients were examined (341 gastritis, 10 peptic ulcer, and 17 gastric cancer). Sixty-two (16.8%) strains were cagA-negative and 306 (83.1%) were cagA-positive (293 Western-type, 12 East-Asian-type, and one hybrid type). All cagA-negative strains were isolated from gastritis patients. In the gastritis group, 78.6% (268/341) had Western-type CagA, 2.9% (10/341) had East-Asian-type, and 18.2% (61/341) were cagA-negative. However, all H. pylori from gastric cancer patients possessed Western-type CagA. Histological analyses showed that East-Asian-type CagA was the most virulent strains, followed by Western-type and cagA-negative strains. This finding agreed with the current consensus. CagA-positive strains were the most virulent type. However, the fact that different CagA types can explain the high incidence of gastric cancer might be inapplicable in Mongolia.
Collapse
Affiliation(s)
- Tegshee Tserentogtokh
- Department of Gastroenterology, Mongolian National University of Medical Sciences, Ulaanbaatar city 14210, Mongolia.
- Department of Endoscopy, Medipas hospital, Orkhon province, Bayan-Undur soum, Zest bag 61029 , Mongolia.
| | - Boldbaatar Gantuya
- Department of Gastroenterology, Mongolian National University of Medical Sciences, Ulaanbaatar city 14210, Mongolia.
- Department of Environmental and Preventive Medicine, Oita University Faculty of Medicine, Yufu city 879-5593, Japan.
| | - Phawinee Subsomwong
- Department of Environmental and Preventive Medicine, Oita University Faculty of Medicine, Yufu city 879-5593, Japan.
| | - Khasag Oyuntsetseg
- Department of Gastroenterology, Mongolian National University of Medical Sciences, Ulaanbaatar city 14210, Mongolia.
| | - Dashdorj Bolor
- Department of Endoscopy, National Cancer Center, Ulaanbaatar city 13370, Mongolia.
| | - Yansan Erdene-Ochir
- Department of General Surgery National Cancer Center, Ulaanbaatar city 13370, Mongolia.
| | - Dashdorj Azzaya
- Department of Environmental and Preventive Medicine, Oita University Faculty of Medicine, Yufu city 879-5593, Japan.
| | - Duger Davaadorj
- Department of Gastroenterology, Mongolian National University of Medical Sciences, Ulaanbaatar city 14210, Mongolia.
| | - Tomohisa Uchida
- Department of Molecular Pathology, Oita University Faculty of Medicine, Yufu city 879-5593, Japan.
| | - Takeshi Matsuhisa
- Department of Endoscopy, Nippon Medical University Tama Nagayama Hospital, Tokyo 113-8602, Japan.
| | - Yoshio Yamaoka
- Department of Environmental and Preventive Medicine, Oita University Faculty of Medicine, Yufu city 879-5593, Japan.
- Global Oita Medical Advanced Research Center for Health, Yufu city 879-5593, Japan.
- Department of Medicine, Gastroenterology and Hepatology section, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
41
|
Gonciarz W, Walencka M, Moran AP, Hinc K, Obuchowski M, Chmiela M. Upregulation of MUC5AC production and deposition of LEWIS determinants by HELICOBACTER PYLORI facilitate gastric tissue colonization and the maintenance of infection. J Biomed Sci 2019; 26:23. [PMID: 30841890 PMCID: PMC6402143 DOI: 10.1186/s12929-019-0515-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 02/19/2019] [Indexed: 02/07/2023] Open
Abstract
Background Helicobacter pylori bacteria colonize human gastric mucosa, cause chronic inflammation, peptic ulcers and gastric cancer. Colonization is mediated by H. pylori adhesins, which preferentially bind mucin 5 (MUC5AC) and Lewis (Le) determinants. The aim of this study was to evaluate the influence of H. pylori and their components on MUC5AC production and deposition of LeX/LeY in gastric epithelial cells in relation to bacterial adhesion using Caviae porcellus primary gastric epithelial cells and an in vivo model of experimental H. pylori infection in these animals. Methods MUCA5C and LeX/LeY were induced in vitro by live H. pylori reference strain CCUG 17874 (2 × 107 CFU/ml), H. pylori glycine acid extract (GE), 10 μg/ml; cytotoxin associated gene A (CagA) protein, 1 μl/ml; UreA urease subunit, 5 μg/ml; lipopolysaccharide (LPS) 25 ng/ml and imaged by fluorescence microscopy after anti-MUC5AC or anti-LeX/LeY FITC antibody staining. Bacterial adhesion was imaged by using anti-H. pylori FITC antibodies. The animals were inoculated per os with H. pylori (3 times in 2 days intervals, 1 × 1010 CFU/ml). After 7 or 28 days an infection and inflammation were assessed by histological, serological and molecular methods. Gastric tissue sections of infected and control animals were screend for MUCA5C and LeX, and H. pylori adhesion as above. Results MUC5AC production and deposition of Lewis determinants, especially LeX were upregulated in the milieu of live H. pylori as well as GE, CagA, UreA or LPS in vitro and in vivo during infection, more effectively in the acute (7 days) than in the chronic (28 days) phase of infection. This was related to enhanced adhesion of H. pylori, which was abrogated by anti-MUC5AC and anti-LeX or anti-LeY antibody treatment. Conclusions Modulation of MUCA5C production and LeX/LeY deposition in the gastric mucosa by H. pylori can significantly increase gastric tissue colonization during H. pylori infection.
Collapse
Affiliation(s)
- Weronika Gonciarz
- Division of Gastroimmunology, Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Łódź, Banacha 12/16, 90-237, Łódź, Poland
| | - Maria Walencka
- Division of Gastroimmunology, Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Łódź, Banacha 12/16, 90-237, Łódź, Poland
| | - Anthony P Moran
- Department of Microbiology, School of Natural Sciences, National University of Ireland Galway, Galway, Ireland
| | - Krzysztof Hinc
- Laboratory of Molecular Bacteriology, Intercollegiate Faculty of Biotechnology UG-MUG, Medical University of Gdańsk, 80-210, Gdańsk, Poland
| | - Michał Obuchowski
- Laboratory of Molecular Bacteriology, Intercollegiate Faculty of Biotechnology UG-MUG, Medical University of Gdańsk, 80-210, Gdańsk, Poland
| | - Magdalena Chmiela
- Division of Gastroimmunology, Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology, Faculty of Biology and Environmental Protection, University of Łódź, Banacha 12/16, 90-237, Łódź, Poland.
| |
Collapse
|
42
|
Siemer S, Hahlbrock A, Vallet C, McClements DJ, Balszuweit J, Voskuhl J, Docter D, Wessler S, Knauer SK, Westmeier D, Stauber RH. Nanosized food additives impact beneficial and pathogenic bacteria in the human gut: a simulated gastrointestinal study. NPJ Sci Food 2018; 2:22. [PMID: 30882042 PMCID: PMC6420113 DOI: 10.1038/s41538-018-0030-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Nanotechnology provides the food industry with new ways to modulate various aspects of food. Hence, engineered nanoparticles (NPs) are increasingly added to food and beverage products as functional ingredients. However, the impact of engineered as well as naturally occurring NPs on both commensal and pathogenic microorganisms within the gastrointestinal tract (GI) is not fully understood. Here, well-defined synthetic NPs and bacterial models were used to probe nanoparticle–bacteria interactions, from analytical to in situ to in vitro. NP–bacteria complexation occurred most efficiently for small NPs, independent of their core material or surface charge, but could be reduced by NPs’ steric surface modifications. Adsorption to bacteria could also be demonstrated for naturally occurring carbon NPs isolated from beer. Complex formation affected the (patho)biological behavior of both the NPs and bacteria, including their cellular uptake into epithelial cells and phagocytes, pathogenic signaling pathways, and NP-induced cell toxicity. NP–bacteria complex formation was concentration-dependently reduced when the NPs became coated with biomolecule coronas with sequential simulation of first oral uptake and then the GI. However, efficient NP adsorption was restored when the pH was sufficiently low, such as in simulating the conditions of the stomach. Collectively, NP binding to enteric bacteria may impact their (patho)biology, particularly in the stomach. Nanosized-food additives as well as naturally occurring NPs may be exploited to (rationally) shape the microbiome. The information contained in this article should facilitate a “safe by design” strategy for the development and application of engineered NPs as functional foods ingredients. Engineered or naturally occurring nanoparticles could potentially affect the bacteria in the gut. A study led by Dana Westmeier and Roland Stauber from University Medical Center of Mainz, Germany probed the nanoparticle–bacteria interactions in situ. They found that NP–bacteria complex occurred most efficiently for small NPs, independent of their core material or surface charge. The complex formation affected the (patho)biological behavior of both the NPs and bacteria, particularly under conditions that simulate the stomach. The result shows that both engineered and naturally occurring nanoparticles could be exploited to shape the gut microbiome. The study can offer guidelines for future development and application of nanoparticles in food industry.
Collapse
Affiliation(s)
- Svenja Siemer
- Department of Nanobiomedicine/ENT, University Medical Center of Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Angelina Hahlbrock
- Department of Nanobiomedicine/ENT, University Medical Center of Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Cecilia Vallet
- Department of Molecular Biology II, Centre for Medical Biotechnology (ZMB), University Duisburg-Essen, Universitätsstraße 5, 45117 Essen, Germany
| | | | - Jan Balszuweit
- Institute for Organic Chemistry, University of Duisburg-Essen, Universitätsstraße 5, 45117 Essen, Germany
| | - Jens Voskuhl
- Institute for Organic Chemistry, University of Duisburg-Essen, Universitätsstraße 5, 45117 Essen, Germany
| | - Dominic Docter
- Department of Nanobiomedicine/ENT, University Medical Center of Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Silja Wessler
- Department of Microbiology, Paris-Lodron University of Salzburg, A-5020 Salzburg, Austria
| | - Shirley K Knauer
- Department of Molecular Biology II, Centre for Medical Biotechnology (ZMB), University Duisburg-Essen, Universitätsstraße 5, 45117 Essen, Germany
| | - Dana Westmeier
- Department of Nanobiomedicine/ENT, University Medical Center of Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| | - Roland H Stauber
- Department of Nanobiomedicine/ENT, University Medical Center of Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany
| |
Collapse
|
43
|
Chmiela M, Walczak N, Rudnicka K. Helicobacter pylori outer membrane vesicles involvement in the infection development and Helicobacter pylori-related diseases. J Biomed Sci 2018; 25:78. [PMID: 30409143 PMCID: PMC6225681 DOI: 10.1186/s12929-018-0480-y] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 10/24/2018] [Indexed: 02/07/2023] Open
Abstract
Helicobacter pylori - (H. pylori) play a role in the pathogenesis of gastritis, gastric and duodenal ulcers as well as gastric cancer. A possible involvement of outer membrane vesicles (OMVs) produced by H. pylori in the distribution of bacterial antigens through the gastric epithelial barrier and their role in the development of local and systemic host inflammatory and immune responses has been suggested. OMVs contain various biologically active compounds, which internalize into host cells affecting signaling pathways and promoting apoptosis of gastric epithelial and immunocompetent cells. OMVs-associated H. pylori virulence factors may strengthen or downregulate the immune responses leading to disease development. This review describes the biological importance of H. pylori OMVs and their role in the course of H. pylori infections, as well as H. pylori related local and systemic effects.
Collapse
Affiliation(s)
- Magdalena Chmiela
- Laboratory of Gastroimmunology, Department of Immunology and Infectious Biology, Faculty of Biology and Environmental Protection, University of Łódź, Banacha 12/16, 90-237, Łódź, Poland
| | - Natalia Walczak
- Laboratory of Gastroimmunology, Department of Immunology and Infectious Biology, Faculty of Biology and Environmental Protection, University of Łódź, Banacha 12/16, 90-237, Łódź, Poland
| | - Karolina Rudnicka
- Laboratory of Gastroimmunology, Department of Immunology and Infectious Biology, Faculty of Biology and Environmental Protection, University of Łódź, Banacha 12/16, 90-237, Łódź, Poland.
| |
Collapse
|
44
|
In silico proteomic and phylogenetic analysis of the outer membrane protein repertoire of gastric Helicobacter species. Sci Rep 2018; 8:15453. [PMID: 30337679 PMCID: PMC6194013 DOI: 10.1038/s41598-018-32476-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 09/03/2018] [Indexed: 12/16/2022] Open
Abstract
Helicobacter (H.) pylori is an important risk factor for gastric malignancies worldwide. Its outer membrane proteome takes an important role in colonization of the human gastric mucosa. However, in zoonotic non-H. pylori helicobacters (NHPHs) also associated with human gastric disease, the composition of the outer membrane (OM) proteome and its relative contribution to disease remain largely unknown. By means of a comprehensive survey of the diversity and distribution of predicted outer membrane proteins (OMPs) identified in all known gastric Helicobacter species with fully annotated genome sequences, we found genus- and species-specific families known or thought to be implicated in virulence. Hop adhesins, part of the Helicobacter-specific family 13 (Hop, Hor and Hom) were restricted to the gastric species H. pylori, H. cetorum and H. acinonychis. Hof proteins (family 33) were putative adhesins with predicted Occ- or MOMP-family like 18-stranded β-barrels. They were found to be widespread amongst all gastric Helicobacter species only sporadically detected in enterohepatic Helicobacter species. These latter are other members within the genus Helicobacter, although ecologically and genetically distinct. LpxR, a lipopolysaccharide remodeling factor, was also detected in all gastric Helicobacter species but lacking as well from the enterohepatic species H. cinaedi, H. equorum and H. hepaticus. In conclusion, our systemic survey of Helicobacter OMPs points to species and infection-site specific members that are interesting candidates for future virulence and colonization studies.
Collapse
|
45
|
Bagheri N, Salimzadeh L, Shirzad H. The role of T helper 1-cell response in Helicobacter pylori-infection. Microb Pathog 2018; 123:1-8. [PMID: 29936093 DOI: 10.1016/j.micpath.2018.06.033] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 06/19/2018] [Accepted: 06/21/2018] [Indexed: 12/11/2022]
Abstract
Helicobacter pylori (H. pylori) is a human pathogen affecting over 50% of the world population. This pathogen is usually associated with chronic inflammation of the gastric mucosa that can lead to peptic ulcer disease (PUD) and gastric cancer (GC), especially in susceptible individuals. These outcomes have been attributed to the interaction of several factors, including host genetic susceptibility, local innate and adaptive immune responses, virulence factors of H. pylori, and environmental factors. T helper (Th) cell subsets and their signature cytokines especially IFN-γ, contribute to anti-bacterial response, but at the mean time sustaining chronic inflammatory responses in the site of infection. It has been acknowledged that H. pylori-infection results in a Th1-dominant response and that inflammation of the gastric mucosa depends mainly on Th1 cell responses. But, the mechanism of the role of Th1 cell responses in H. pylori-infection has not yet been clearly explained. In this review, we will focus on the role of Th1 involved in H. pylori-infection, its interaction with Th17/Treg cells and its association with the clinical consequences of the infection.
Collapse
Affiliation(s)
- Nader Bagheri
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| | - Loghman Salimzadeh
- Department of Microbiology and Immunology Programme, National University of Singapore, Singapore
| | - Hedayatollah Shirzad
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| |
Collapse
|
46
|
Moonens K, Hamway Y, Neddermann M, Reschke M, Tegtmeyer N, Kruse T, Kammerer R, Mejías-Luque R, Singer BB, Backert S, Gerhard M, Remaut H. Helicobacter pylori adhesin HopQ disrupts trans dimerization in human CEACAMs. EMBO J 2018; 37:embj.201798665. [PMID: 29858229 DOI: 10.15252/embj.201798665] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 05/08/2018] [Accepted: 05/15/2018] [Indexed: 01/24/2023] Open
Abstract
The human gastric pathogen Helicobacter pylori is a major causative agent of gastritis, peptic ulcer disease, and gastric cancer. As part of its adhesive lifestyle, the bacterium targets members of the carcinoembryonic antigen-related cell adhesion molecule (CEACAM) family by the conserved outer membrane adhesin HopQ. The HopQ-CEACAM1 interaction is associated with inflammatory responses and enables the intracellular delivery and phosphorylation of the CagA oncoprotein via a yet unknown mechanism. Here, we generated crystal structures of HopQ isotypes I and II bound to the N-terminal domain of human CEACAM1 (C1ND) and elucidated the structural basis of H. pylori specificity toward human CEACAM receptors. Both HopQ alleles target the β-strands G, F, and C of C1ND, which form the trans dimerization interface in homo- and heterophilic CEACAM interactions. Using SAXS, we show that the HopQ ectodomain is sufficient to induce C1ND monomerization and thus providing H. pylori a route to influence CEACAM-mediated cell adherence and signaling events.
Collapse
Affiliation(s)
- Kristof Moonens
- Structural and Molecular Microbiology, Structural Biology Research Center, VIB, Brussels, Belgium.,Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Youssef Hamway
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München, Munich, Germany
| | - Matthias Neddermann
- Division of Microbiology, Department of Biology, Friedrich Alexander University Erlangen, Erlangen, Germany
| | - Marc Reschke
- Institute of Anatomy, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Nicole Tegtmeyer
- Division of Microbiology, Department of Biology, Friedrich Alexander University Erlangen, Erlangen, Germany
| | | | - Robert Kammerer
- Institute of Immunology, Friedrich-Loeffler Institut, Greifswald-Insel Riems, Germany
| | - Raquel Mejías-Luque
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München, Munich, Germany.,German Center for Infection Research, Partner Site Munich, Munich, Germany
| | - Bernhard B Singer
- Institute of Anatomy, Medical Faculty, University of Duisburg-Essen, Essen, Germany
| | - Steffen Backert
- Division of Microbiology, Department of Biology, Friedrich Alexander University Erlangen, Erlangen, Germany
| | - Markus Gerhard
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München, Munich, Germany.,German Center for Infection Research, Partner Site Munich, Munich, Germany
| | - Han Remaut
- Structural and Molecular Microbiology, Structural Biology Research Center, VIB, Brussels, Belgium .,Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
47
|
The Double Face of Mucin-Type O-Glycans in Lectin-Mediated Infection and Immunity. Molecules 2018; 23:molecules23051151. [PMID: 29751628 PMCID: PMC6100456 DOI: 10.3390/molecules23051151] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 05/03/2018] [Accepted: 05/07/2018] [Indexed: 02/07/2023] Open
Abstract
Epithelial human blood group antigens (HBGAs) on O-glycans play roles in pathogen binding and the initiation of infection, while similar structures on secretory mucins exert protective functions. These double-faced features of O-glycans in infection and innate immunity are reviewed based on two instructive examples of bacterial and viral pathogens. Helicobacter pylori represents a class 1 carcinogen in the human stomach. By expressing blood group antigen-binding adhesin (BabA) and LabA adhesins that bind to Lewis-b and LacdiNAc, respectively, H. pylori colocalizes with the mucin MUC5AC in gastric surface epithelia, but not with MUC6, which is cosecreted with trefoil factor family 2 (TFF2) by deep gastric glands. Both components of the glandular secretome are concertedly up-regulated upon infection. While MUC6 expresses GlcNAc-capped glycans as natural antibiotics for H. pylori growth control, TFF2 may function as a probiotic lectin. In viral infection human noroviruses of the GII genogroup interact with HBGAs via their major capsid protein, VP1. HBGAs on human milk oligosaccharides (HMOs) may exert protective functions by binding to the P2 domain pocket on the capsid. We discuss structural details of the P2 carbohydrate-binding pocket in interaction with blood group H/Lewis-b HMOs and fucoidan-derived oligofucoses as effective interactors for the most prevalent norovirus strains, GII.4 and GII.17.
Collapse
|
48
|
Backert S, Haas R, Gerhard M, Naumann M. The Helicobacter pylori Type IV Secretion System Encoded by the cag Pathogenicity Island: Architecture, Function, and Signaling. Curr Top Microbiol Immunol 2018. [DOI: 10.1007/978-3-319-75241-9_8] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
49
|
Westmeier D, Posselt G, Hahlbrock A, Bartfeld S, Vallet C, Abfalter C, Docter D, Knauer SK, Wessler S, Stauber RH. Nanoparticle binding attenuates the pathobiology of gastric cancer-associated Helicobacter pylori. NANOSCALE 2018; 10:1453-1463. [PMID: 29303193 DOI: 10.1039/c7nr06573f] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Enteric bacteria may cause severe diseases, including gastric cancer-associated Helicobacter pylori. Their infection paths overlap with the oro-gastrointestinal uptake route for nanoparticles, increasingly occurring during environmental or consumer/medical exposure. By comprehensive independent analytical methods, such as live cell fluorescence, electron as well as atomic force microscopy and elemental analysis, we show that a wide array of nanoparticles (NPs) but not microparticles form complexes with H. pylori and enteric pathogens without the need for specific functionalization. The NP-assembly that occurred rapidly was not influenced by variations in physiological temperature, though affected by the NPs' physico-chemical characteristics. Improved binding was observed for small NPs with a negative surface charge, whereas binding could be reduced by surface 'stealth' modifications. Employing human gastric epithelial cells and 3D-organoid models of the stomach, we show that NP-coating did not inhibit H. pylori's cellular attachment. However, even the assembly of non-bactericidal silica NPs attenuated H. pylori infection by reducing CagA phosphorylation, cytoskeletal rearrangement, and IL-8 secretion. Here we demonstrate that NP binding to enteric bacteria may impact their pathobiology which could be further exploited to rationally modulate the (patho)biology of microbes by nanomaterials.
Collapse
Affiliation(s)
- Dana Westmeier
- Department of Nanobiomedicine/ENT, University Medical Center of Mainz, Langenbeckstrasse 1, 55101 Mainz, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Westmeier D, Hahlbrock A, Reinhardt C, Fröhlich-Nowoisky J, Wessler S, Vallet C, Pöschl U, Knauer SK, Stauber RH. Nanomaterial–microbe cross-talk: physicochemical principles and (patho)biological consequences. Chem Soc Rev 2018; 47:5312-5337. [DOI: 10.1039/c6cs00691d] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
NPs’ characteristics impact their spontaneous binding to microbes, which may affect the (patho)biological identity of both NP and microbes.
Collapse
Affiliation(s)
- D. Westmeier
- Department of Nanobiomedicine/ENT
- University Medical Center of Mainz
- 55101 Mainz
- Germany
| | - A. Hahlbrock
- Department of Nanobiomedicine/ENT
- University Medical Center of Mainz
- 55101 Mainz
- Germany
| | - C. Reinhardt
- Center for Thrombosis and Hemostasis
- University Medical Center Mainz
- 55101 Mainz
- Germany
| | - J. Fröhlich-Nowoisky
- Multiphase Chemistry Department
- Max Planck Institute for Chemistry
- 55128 Mainz
- Germany
| | - S. Wessler
- Department of Molecular Biology
- Paris-Lodron University of Salzburg
- A-5020 Salzburg
- Austria
| | - C. Vallet
- Institute for Molecular Biology
- CENIDE
- University Duisburg-Essen
- 45117 Essen
- Germany
| | - U. Pöschl
- Multiphase Chemistry Department
- Max Planck Institute for Chemistry
- 55128 Mainz
- Germany
| | - S. K. Knauer
- Institute for Molecular Biology
- CENIDE
- University Duisburg-Essen
- 45117 Essen
- Germany
| | - R. H. Stauber
- Department of Nanobiomedicine/ENT
- University Medical Center of Mainz
- 55101 Mainz
- Germany
| |
Collapse
|