1
|
Xie R, You N, Chen WY, Zhu P, Wang P, Lv YP, Yue GY, Xu XL, Wu JB, Xu JY, Liu SX, Lü MH, Yang SQ, Cheng P, Mao FY, Teng YS, Peng LS, Zhang JY, Liao YL, Yang SM, Zhao YL, Chen W, Zou QM, Zhuang Y. Helicobacter pylori-Induced Angiopoietin-Like 4 Promotes Gastric Bacterial Colonization and Gastritis. RESEARCH (WASHINGTON, D.C.) 2024; 7:0409. [PMID: 39022746 PMCID: PMC11254415 DOI: 10.34133/research.0409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 05/22/2024] [Indexed: 07/20/2024]
Abstract
Helicobacter pylori infection is characterized as progressive processes of bacterial persistence and chronic gastritis with features of infiltration of mononuclear cells more than granulocytes in gastric mucosa. Angiopoietin-like 4 (ANGPTL4) is considered a double-edged sword in inflammation-associated diseases, but its function and clinical relevance in H. pylori-associated pathology are unknown. Here, we demonstrate both pro-colonization and pro-inflammation roles of ANGPTL4 in H. pylori infection. Increased ANGPTL4 in the infected gastric mucosa was produced from gastric epithelial cells (GECs) synergistically induced by H. pylori and IL-17A in a cagA-dependent manner. Human gastric ANGPTL4 correlated with H. pylori colonization and the severity of gastritis, and mouse ANGPTL4 from non-bone marrow-derived cells promoted bacteria colonization and inflammation. Importantly, H. pylori colonization and inflammation were attenuated in Il17a -/-, Angptl4 -/-, and Il17a -/- Angptl4 -/- mice. Mechanistically, ANGPTL4 bound to integrin αV (ITGAV) on GECs to suppress CXCL1 production by inhibiting ERK, leading to decreased gastric influx of neutrophils, thereby promoting H. pylori colonization; ANGPTL4 also bound to ITGAV on monocytes to promote CCL5 production by activating PI3K-AKT-NF-κB, resulting in increased gastric influx of regulatory CD4+ T cells (Tregs) via CCL5-CCR4-dependent migration. In turn, ANGPTL4 induced Treg proliferation by binding to ITGAV to activate PI3K-AKT-NF-κB, promoting H. pylori-associated gastritis. Overall, we propose a model in which ANGPTL4 collectively ensures H. pylori persistence and promotes gastritis. Efforts to inhibit ANGPTL4-associated pathway may prove valuable strategies in treating H. pylori infection.
Collapse
Affiliation(s)
- Rui Xie
- Department ofEndoscopy and Digestive System, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Nan You
- Department of Hepatobiliary Surgery, XinQiao Hospital,
Third Military Medical University, Chongqing, China
| | - Wan-Yan Chen
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine,
Third Military Medical University, Chongqing, China
| | - Peng Zhu
- Department of Gastroenterology, Suining First People’s Hospital, Suining, Sichuan, China
| | - Pan Wang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine,
Third Military Medical University, Chongqing, China
| | - Yi-Pin Lv
- Department of Infection, The General Hospital of Western Theater Command, Chengdu, Sichuan, China
| | - Geng-Yu Yue
- Department ofEndoscopy and Digestive System, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Xiao-Lin Xu
- Department ofEndoscopy and Digestive System, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Jiang-Bo Wu
- Department ofEndoscopy and Digestive System, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Jing-Yu Xu
- Department ofEndoscopy and Digestive System, Guizhou Provincial People’s Hospital, Guiyang, China
| | - Si-Xu Liu
- Department of Gastroenterology,
Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Mu-Han Lü
- Department of Gastroenterology,
Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Sheng-Qian Yang
- Chongqing Engineering Research Center for Pharmacodynamics Evaluation, Department of Pharmaceutics, College of Pharmacy and Laboratory Medicine,
Third Military Medical University, Chongqing, China
| | - Ping Cheng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine,
Third Military Medical University, Chongqing, China
| | - Fang-Yuan Mao
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine,
Third Military Medical University, Chongqing, China
| | - Yong-Sheng Teng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine,
Third Military Medical University, Chongqing, China
| | - Liu-Sheng Peng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine,
Third Military Medical University, Chongqing, China
| | - Jin-Yu Zhang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine,
Third Military Medical University, Chongqing, China
| | - Ya-Ling Liao
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine,
Third Military Medical University, Chongqing, China
| | - Shi-Ming Yang
- Department of Gastroenterology, XinQiao Hospital,
Third Military Medical University, Chongqing, China
| | - Yong-Liang Zhao
- Department of General Surgery and Center of Minimal Invasive Gastrointestinal Surgery, Southwest Hospital,
Third Military Medical University, Chongqing, China
| | - Weisan Chen
- La Trobe Institute of Molecular Science,
La Trobe University, Bundoora, Victoria 3085, Australia
| | - Quan-Ming Zou
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine,
Third Military Medical University, Chongqing, China
| | - Yuan Zhuang
- Department ofEndoscopy and Digestive System, Guizhou Provincial People’s Hospital, Guiyang, China
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine,
Third Military Medical University, Chongqing, China
- Department of Gastroenterology,
Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, China
| |
Collapse
|
2
|
Matysiak-Budnik T, Priadko K, Bossard C, Chapelle N, Ruskoné-Fourmestraux A. Clinical Management of Patients with Gastric MALT Lymphoma: A Gastroenterologist's Point of View. Cancers (Basel) 2023; 15:3811. [PMID: 37568627 PMCID: PMC10417821 DOI: 10.3390/cancers15153811] [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: 06/23/2023] [Revised: 07/25/2023] [Accepted: 07/26/2023] [Indexed: 08/13/2023] Open
Abstract
Gastric mucosa-associated lymphoid tissue (MALT) lymphomas (GML) are non-Hodgkin lymphomas arising from the marginal zone of the lymphoid tissue of the stomach. They are usually induced by chronic infection with Helicobacter pylori (H. pylori); however, H. pylori-negative GML is of increasing incidence. The diagnosis of GML is based on histological examination of gastric biopsies, but the role of upper endoscopy is crucial since it is the first step in the diagnostic process and, with currently available novel endoscopic techniques, may even allow an in vivo diagnosis of GML per se. The treatment of GML, which is usually localized, always includes the eradication of H. pylori, which should be performed even in H. pylori-negative GML. In the case of GML persistence after eradication of the bacteria, low-dose radiotherapy may be proposed, while systemic treatments (immunochemotherapy) should be reserved for very rare disseminated cases. In GML patients, at diagnosis but even after complete remission, special attention must be paid to an increased risk of gastric adenocarcinoma, especially in the presence of associated gastric precancerous lesions (gastric atrophy and gastric intestinal metaplasia), which requires adequate endoscopic surveillance of these patients.
Collapse
Affiliation(s)
- Tamara Matysiak-Budnik
- IMAD, Hepato-Gastroenterology & Digestive Oncology, University Hospital of Nantes, 44093 Nantes, France; (K.P.); (N.C.)
- Inserm, CHU Nantes, University of Nantes, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, 44000 Nantes, France
| | - Kateryna Priadko
- IMAD, Hepato-Gastroenterology & Digestive Oncology, University Hospital of Nantes, 44093 Nantes, France; (K.P.); (N.C.)
- Hepato-Gastroenterology Unit, University Hospital Universita degli Studi della Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | | | - Nicolas Chapelle
- IMAD, Hepato-Gastroenterology & Digestive Oncology, University Hospital of Nantes, 44093 Nantes, France; (K.P.); (N.C.)
- Inserm, CHU Nantes, University of Nantes, Centre de Recherche en Transplantation et Immunologie, UMR 1064, ITUN, 44000 Nantes, France
| | | |
Collapse
|
3
|
Increased IL-17A Serum Levels and Gastric Th17 Cells in Helicobacter pylori-Infected Patients with Gastric Premalignant Lesions. Cancers (Basel) 2023; 15:cancers15061662. [PMID: 36980548 PMCID: PMC10046233 DOI: 10.3390/cancers15061662] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 02/28/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023] Open
Abstract
Background: Helicobacter pylori infection is characterized by an inflammatory infiltrate that might be an important antecedent of gastric cancer. The purpose of this study was to evaluate whether interleukin (IL)-17 inflammation is elicited by gastric T cells in Helicobacter pylori patients with gastric intestinal metaplasia and dysplasia (IM/DYS). We also investigated the serum IL-17A levels in Helicobacter pylori patients with gastric intestinal metaplasia and dysplasia, and patients with Helicobacter pylori non-atrophic gastritis (NAG). Methods: the IL-17 cytokine profile of gastric T cells was investigated in six patients with IM/DYS and Helicobacter pylori infection. Serum IL-17A levels were measured in 45 Helicobacter pylori-infected IM/DYS patients, 45 Helicobacter pylori-infected patients without IM/DYS and in 45 healthy controls (HC). Results: gastric T cells from all IM/DYS patients with Helicobacter pylori were able to proliferate in response to Helicobacter pylori and to produce IL-17A. The Luminex analysis revealed that IL-17A levels were significantly increased in Helicobacter pylori IM/DYS patients compared to healthy controls and to Helicobacter pylori gastritis patients without IM/DYS (452.34 ± 369.13 pg/mL, 246.82 ± 156.06 pg/mL, 169.26 ± 73.82 pg/mL, respectively; p < 0.01, p < 0.05). Conclusions: the results obtained indicate that Helicobacter pylori is able to drive gastric IL-17 inflammation in IM/DYS Helicobacter pylori-infected patients, and that IL-17A serum levels are significantly increased in Helicobacter pylori-infected patients with IM/DYS.
Collapse
|
4
|
Naumann M, Ferino L, Sharafutdinov I, Backert S. Gastric Epithelial Barrier Disruption, Inflammation and Oncogenic Signal Transduction by Helicobacter pylori. Curr Top Microbiol Immunol 2023; 444:207-238. [PMID: 38231220 DOI: 10.1007/978-3-031-47331-9_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
Helicobacter pylori exemplifies one of the most favourable bacterial pathogens worldwide. The bacterium colonizes the gastric mucosa in about half of the human population and constitutes a major risk factor for triggering gastric diseases such as stomach cancer. H. pylori infection represents a prime example of chronic inflammation and cancer-inducing bacterial pathogens. The microbe utilizes a remarkable set of virulence factors and strategies to control cellular checkpoints of inflammation and oncogenic signal transduction. This chapter emphasizes on the pathogenicity determinants of H. pylori such as the cytotoxin-associated genes pathogenicity island (cagPAI)-encoded type-IV secretion system (T4SS), effector protein CagA, lipopolysaccharide (LPS) metabolite ADP-glycero-β-D-manno-heptose (ADP-heptose), cytotoxin VacA, serine protease HtrA, and urease, and how they manipulate various key host cell signaling networks in the gastric epithelium. In particular, we highlight the H. pylori-induced disruption of cell-to-cell junctions, pro-inflammatory activities, as well as proliferative, pro-apoptotic and anti-apoptotic responses. Here we review these hijacked signal transduction events and their impact on gastric disease development.
Collapse
Affiliation(s)
- Michael Naumann
- Institute of Experimental Internal Medicine, Medical Faculty, Otto Von Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany.
| | - Lorena Ferino
- Institute of Experimental Internal Medicine, Medical Faculty, Otto Von Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
| | - Irshad Sharafutdinov
- Dept. Biology, Division of Microbiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Staudtstr. 5, 91058, Erlangen, Germany
| | - Steffen Backert
- Dept. Biology, Division of Microbiology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Staudtstr. 5, 91058, Erlangen, Germany.
| |
Collapse
|
5
|
Investigating the Active Substance and Mechanism of San-Jiu-Wei-Tai Granules via UPLC-QE-Orbitrap-MS and Network Pharmacology. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:1487903. [PMID: 36299773 PMCID: PMC9592199 DOI: 10.1155/2022/1487903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 09/21/2022] [Accepted: 10/07/2022] [Indexed: 11/17/2022]
Abstract
San-Jiu-Wei-Tai granules (SJWTG) are a significant Chinese patent medicine for the treatment of chronic gastritis (CG), having outstanding advantages in long-term treatment; however, the chemical composition and potential mechanism have not been investigated until now. In this study, a rapid separation and identification method based on UPLC-QE-Orbitrap-MS was established, and 95 chemical components from SJWTGs were identified, including 6 chemical components of an unknown source that are not derived from the 8 herbs included in SJWTGs. The identified chemical components were subsequently analysed by network pharmacology, suggesting that the core targets for the treatment of CG with SJWTGs were EGFR, SRC, AKT1, HSP90AA1, MAPK1, and MAPK3 and thus indicating that SJWTGs could reduce the inflammatory response of gastric epithelial cells and prevent persistent chronic inflammation that induces cancerization by regulating the MAPK signalling pathway and the C-type lectin receptor signalling pathway as well as their upstream and downstream pathways in the treatment of CG. The key bioactive components in SJWTGs were identified as 2,6-bis(4-ethylphenyl)perhydro-1,3,5,7-tetraoxanaphth-4-ylethane-1,2-diol, a chemical component of an unknown source, murrangatin, meranzin hydrate, paeoniflorin, and albiflorin. The results of molecular docking showed the strong binding interaction between the key bioactive components and the core targets, demonstrating that the key bioactive components deserve to be further studied and considered as Q-markers. By acting on multiple targets, SJWTG is less susceptible to drug resistance during the long-term treatment of CG, indicating the advantage of Chinese patent medicines. Furthermore, the preventive effect of SJWTGs on gastric cancer also demonstrates the superiority of preventive treatment of disease with traditional Chinese medicine.
Collapse
|
6
|
Della Bella C, Antico A, Panozzo MP, Capitani N, Petrone L, Benagiano M, D’Elios S, Sparano C, Azzurri A, Pratesi S, Cianchi F, Ortiz-Princz D, Bergman M, Bizzaro N, D’Elios MM. Gastric Th17 Cells Specific for H+/K+-ATPase and Serum IL-17 Signature in Gastric Autoimmunity. Front Immunol 2022; 13:952674. [PMID: 35911678 PMCID: PMC9328118 DOI: 10.3389/fimmu.2022.952674] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 06/16/2022] [Indexed: 11/13/2022] Open
Abstract
Human gastric autoimmunity [autoimmune gastritis (AIG)] is characterized by inflammation of the gastric mucosa and parietal cell loss. The gastric parietal cell proton pump H+/K+-adenosine triphosphatase (H+/K+-ATPase) is the major autoantigen in AIG. Our work aimed to investigate the gastric H+/K+-ATPase-specific T helper 17 (Th17) responses in AIG and serum interleukin (IL)-17 cytokine subfamily in AIG patients, in healthy subjects [healthy controls (HCs)], and in patients with iron deficiency anemia (IDA) without AIG. We analyzed the activation of gastric lamina propria mononuclear cells (LPMCs) by H+/K+-ATPase and the IL-17A and IL-17F cytokine production in eight patients with AIG and four HCs. Furthermore, we compared serum levels of IL-17A, IL-17F, IL-21, IL-17E, IL-22, and IL-23 in 43 AIG patients, in 47 HCs, and in 20 IDA patients without AIG. Gastric LPMCs from all AIG patients, but not those from HCs, were activated by H+/K+-ATPase and were able to proliferate and produce high levels of IL-17A and IL-17F. AIG patients have significantly higher serum IL-17A, IL-17F, IL-21, and IL-17E (393.3 ± 410.02 pg/ml, 394.0 ± 378.03 pg/ml, 300.46 ± 303.45 pg/ml, 34.92 ± 32.56 pg/ml, respectively) than those in HCs (222.99 ± 361.24 pg/ml, 217.49 ± 312.1 pg/ml, 147.43 ± 259.17 pg/ml, 8.69 ± 8.98 pg/ml, respectively) and those in IDA patients without AIG (58.06 ± 107.49 pg/ml, 74.26 ± 178.50 pg/ml, 96.86 ± 177.46 pg/ml, 10.64 ± 17.70 pg/ml, respectively). Altogether, our results indicate that IL-17A and IL-17F are produced in vivo in the stomach of AIG patients following activation with H+/K+-ATPase and that serum IL-17A, IL-17F, IL-21, and IL-17E levels are significantly elevated in AIG patients but not in patients without AIG. These data suggest a Th17 signature in AIG and that IL-17A, IL-17F, IL-21, and IL-17E may represent a relevant tool for AIG management.
Collapse
Affiliation(s)
- Chiara Della Bella
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Antonio Antico
- Laboratory of Clinical Pathology, ULSS7 Pedemontana, Hospital Alto Vicentino, Santorso, Italy
| | - Maria Piera Panozzo
- Laboratory of Clinical Pathology, ULSS7 Pedemontana, Hospital Alto Vicentino, Santorso, Italy
| | - Nagaja Capitani
- Department of Life Sciences, University of Siena, Siena, Italy
| | - Luisa Petrone
- Endocrinology Unit, Careggi Hospital, Florence, Italy
| | - Marisa Benagiano
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Sofia D’Elios
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Annalisa Azzurri
- Laboratory of Clinical Pathology, Toscana Centro Hospital, Florence, Italy
| | - Sara Pratesi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Fabio Cianchi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Diana Ortiz-Princz
- Laboratory of Molecular Microbiology, Autonomous Service Institute of Biomedicine “Dr. Jacinto Convit”, Caracas, Venezuela
| | - Mathijs Bergman
- Molecular Microbiology, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Nicola Bizzaro
- Laboratory of Clinical Pathology, San Antonio Hospital, Tolmezzo, Italy
- Laboratory of Clinical Pathology, Azienda Sanitaria Universitaria Integrata, Udine, Italy
| | - Mario Milco D’Elios
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- *Correspondence: Mario Milco D’Elios,
| |
Collapse
|
7
|
Della Bella C, Antico A, Panozzo MP, Capitani N, Benagiano M, Petrone L, Azzurri A, Pratesi S, D’Elios S, Cianchi F, Ortiz-Princz D, Bizzaro N, D’Elios MM. Elevated IL-19 Serum Levels in Patients With Pernicious Anemia and Autoimmune Gastritis. Front Immunol 2022; 13:887256. [PMID: 35479078 PMCID: PMC9036483 DOI: 10.3389/fimmu.2022.887256] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 03/21/2022] [Indexed: 12/30/2022] Open
Abstract
Pernicious anemia (PA) is a megaloblastic anemia consisting of hematological, gastric and immunological alterations. The immunopathogenesis of PA is sustained by both autoantibodies (e.g. intrinsic factor (IFA) antibodies and anti parietal cell (PCA) antibodies and autoreactive T cells specific for IFA and the parietal cell proton pump ATPase. Iron deficient anemia (IDA) is a microcytic anemia and represents the most common cause of anemia worldwide. Our work aimed to investigate serum levels of several interleukins (IL) of the IL-20 cytokine subfamily in patients with PA, with IDA and in healthy subjects (HC). We compared serum levels of IL-19, IL-20, IL-26, IL-28A and IL-29 in 43 patients with PA and autoimmune gastritis, in 20 patients with IDA and no autoimmune gastritis, and in 47 HC. Furthermore, we analyzed the IL-19 cytokine production by gastric lamina propria mononuclear cells (LPMC) in eight patients with PA and four HC. We found that patients with PA have significantly higher serum levels of IL-19 (163.68 ± 75.96 pg/ml) than patients with IDA (35.49 ± 40.97 pg/ml; p<0.001) and healthy subjects (55.68 ± 36.75 pg/ml; p<0.001). Gastric LPMC from all PA patients were able to produce significantly higher levels of IL-19 (420.67 ± 68.14 pg/ml) than HC (53.69 ± 10.92 pg/ml) (p<0.01). Altogether, our results indicate that IL-19 serum levels are significantly increased in patients with PA but not with IDA and that IL-19 is produced in vivo in the stomach of PA patients. These data open a new perspective on PA pathogenesis and suggest that IL-19 may represent a novel important tool for the management of patients with PA.
Collapse
Affiliation(s)
- Chiara Della Bella
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Antonio Antico
- Laboratory of Clinical Pathology, ULSS7 Pedemontana, Hospital Alto Vicentino, Santorso, Italy
| | - Maria Piera Panozzo
- Laboratory of Clinical Pathology, ULSS7 Pedemontana, Hospital Alto Vicentino, Santorso, Italy
| | - Nagaja Capitani
- Department of Life Sciences, University of Siena, Siena, Italy
| | - Marisa Benagiano
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Luisa Petrone
- Endocrinology Unit, Careggi Hospital, Florence, Italy
| | - Annalisa Azzurri
- Laboratory of Clinical Pathology, Toscana Centro Hospital, Firenze, Italy
| | - Sara Pratesi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Sofia D’Elios
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Fabio Cianchi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Diana Ortiz-Princz
- Laboratory of Molecular Microbiology, Autonomous Service Institute of Biomedicine “Dr. Jacinto Convit”, Caracas, Venezuela
| | - Nicola Bizzaro
- Laboratory of Clinical Pathology, San Antonio Hospital, Tolmezzo, Italy
- Laboratory of Clinical Pathology, Azienda Sanitaria Universitaria Integrata, Udine, Italy
| | - Mario Milco D’Elios
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- *Correspondence: Mario Milco D’Elios,
| |
Collapse
|
8
|
Deng Y, Su W, Zhu J, Ji H, Zhou X, Geng J, Zhu J, Zhang Q. Helicobacter pylori infection disturbs the tumor immune microenvironment and is associated with a discrepant prognosis in gastric de novo diffuse large B-cell lymphoma. J Immunother Cancer 2021; 9:jitc-2021-002947. [PMID: 34645670 PMCID: PMC8515460 DOI: 10.1136/jitc-2021-002947] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/09/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Gastric diffuse large B-cell lymphoma (gDLBCL) related to Helicobacter pylori infection exhibits a wide spectrum of prognosis, and the tumor immune microenvironment (TIME) affects tumor progression. However, there are few studies on the correlation between prognosis and changes of TIME induced by H. pylori infection in de novo gDLBCL. METHODS A retrospective study was performed to determine the prognostic value of TIME related to H. pylori infection in de novo gDLBCL. A total of 252 patients were included and have been treated with standard rituximab to cyclophosphamide, doxorubicin, vincristine, and prednisone chemotherapy or other similar regimens in addition to H. pylori eradication (HPE). All patients were stratified by H. pylori infection, HPE efficacy, and preliminary TIME evaluation using conventional criteria. Statistical analyses were conducted. To assess the mechanism, 30 subjects were assessed for H. pylori infection. The components and spatial distributions of TIME were analyzed. RESULTS The median follow-up of the 252 patients was 66.6 months (range 0.7-119.2), and the 5-year overall survival (OS) was 78.0%. A total of 109 H. pylori-positive cases with pathological complete remission and high tumor-infiltrating T lymphocytes (cohort 1) had significantly higher 5-year progression-free survival (88.1% vs 70.5%, p<0.001) and OS (89.2% vs 76.6%, p<0.001) than the other 143 patients (cohort 2). Among 30 patients, 19 were cytotoxin-associated gene A-marked as the cohort 1 subset. Compared with cohort 2, cohort 1 exhibited increased inflammatory factors (tumor necrosis factor-α, interferon γ, etc) and decreased immunosuppressive components (PD-L1, PD-1, IL-10, etc). There was reduced NF-kB activation. Cancer-promoting immune cells (PD-1hiTim-3+ CTL, Tregs, M2-like macrophages, etc) occupied a minor spatial distribution, while the antitumor subtypes increased, corresponding to favorable survival. CONCLUSION H. pylori-evoked inflammatory responses disturb the TIME, causing a differential prognosis in de novo gDLBCL, which can be used to identify patients who could benefit from HPE and immunochemotherapy.
Collapse
Affiliation(s)
- Yuwei Deng
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, People's Republic of China
| | - Wenjia Su
- Department of Hematology, First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Junwen Zhu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, People's Republic of China
| | - Hongfei Ji
- Institute of Cancer Prevention and Treatment, Heilongjiang Academy of Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Xiaoping Zhou
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, People's Republic of China
| | - Jingshu Geng
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, People's Republic of China
| | - Jiayu Zhu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, People's Republic of China
| | - Qingyuan Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, People's Republic of China
| |
Collapse
|
9
|
Mahant S, Chakraborty A, Som A, Mehra S, Das K, Mukhopadhyay AK, Gehlot V, Bose S, Das R. The Synergistic Role of Tip α, Nucleolin and Ras in Helicobacter pylori Infection Regulates the Cell Fate Towards Inflammation or Apoptosis. Curr Microbiol 2021; 78:3720-3732. [PMID: 34468852 DOI: 10.1007/s00284-021-02626-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 08/03/2021] [Indexed: 12/22/2022]
Abstract
Infection with Helicobacter pylori (H. pylori) leads to a fork in the road situation where it is critical and complex to judge the fate of the cell. We propose for the first time an in silico representation of a protein level network model that can unfold the mystery behind the cell fate decision between inflammation or cell proliferation or cell death. Upon infection TNF inducible protein α (Tip α) is internalised after binding with the cell surface receptor Nucleolin which is overexpressed on the cell surface thereby activating the Ras pathway. Tip α, Nucleolin and Ras decides the cell fate for apoptosis or abnormal cell proliferation along with ulcers in the gastric tract, hence we term it as the "death triad", which otherwise triggers the inflammatory pathway through downstream signalling of NF-κβ. A series of proteins involved in the signalling cascade are portrayed through compartmentalization of the bacteria and the gut wall. The depicted network works synchronously toward an overarching goal of deciding between apoptosis or inflammation or proliferation. The model has been validated by simulating it with existing transcriptomic data along with clinical findings from patients infected with H. pylori across different regions in India. The results clearly indicate that for a short period of time there is increased binding of Tip α to Nucleolin and the receptor starts to saturate. This increases the tenacity of binding and the cell triggers an inflammatory cascade reaction which involves proinflammatory cytokines such as TNF α thereby progressing to inflammation by activating NF-κβ downstream. On the other hand, Ras involved in interaction with nucleolin can be present both in its activated or inactivated state. Binding of Tip α as a monomer leads to desensitization of Nucleolin leading to cell survival and proliferation.
Collapse
Affiliation(s)
- Shweta Mahant
- Amity Institute of Biotechnology, Amity University, Noida, U.P, 201313, India
| | - Amlan Chakraborty
- Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
- Victorian Heart Institute, Monash University, Melbourne, VIC, 3800, Australia
| | - Anup Som
- Centre of Bioinformatics, University of Allahabad, Prayagraj, U.P, 211002, India
| | - Shubham Mehra
- Amity Institute of Biotechnology, Amity University, Noida, U.P, 201313, India
| | - Kunal Das
- Department of Gastroenterology, Manipal Hospital, Dwarka, New Delhi, 110075, India
| | - Asish Kumar Mukhopadhyay
- Division of Bacteriology, National Institute of Cholera and Enteric Diseases, Kolkata, 700010, India
| | - Valentina Gehlot
- Amity Institute of Biotechnology, Amity University, Noida, U.P, 201313, India
| | - Sudeep Bose
- Amity Institute of Biotechnology, Amity University, Noida, U.P, 201313, India.
| | - Rajashree Das
- Amity Institute of Biotechnology, Amity University, Noida, U.P, 201313, India.
| |
Collapse
|
10
|
Della Bella C, Soluri MF, Puccio S, Benagiano M, Grassi A, Bitetti J, Cianchi F, Sblattero D, Peano C, D’Elios MM. The Helicobacter pylori CagY Protein Drives Gastric Th1 and Th17 Inflammation and B Cell Proliferation in Gastric MALT Lymphoma. Int J Mol Sci 2021; 22:ijms22179459. [PMID: 34502367 PMCID: PMC8431018 DOI: 10.3390/ijms22179459] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 08/23/2021] [Accepted: 08/26/2021] [Indexed: 01/22/2023] Open
Abstract
Background: the neoplastic B cells of the Helicobacter pylori-related low-grade gastric mucosa-associated lymphoid tissue (MALT) lymphoma proliferate in response to H. pylori, however, the nature of the H. pylori antigen responsible for proliferation is still unknown. The purpose of the study was to dissect whether CagY might be the H. pylori antigen able to drive B cell proliferation. Methods: the B cells and the clonal progeny of T cells from the gastric mucosa of five patients with MALT lymphoma were compared with those of T cell clones obtained from five H. pylori–infected patients with chronic gastritis. The T cell clones were assessed for their specificity to H. pylori CagY, cytokine profile and helper function for B cell proliferation. Results: 22 of 158 CD4+ (13.9%) gastric clones from MALT lymphoma and three of 179 CD4+ (1.7%) clones from chronic gastritis recognized CagY. CagY predominantly drives Interferon-gamma (IFN-γ) and Interleukin-17 (IL-17) secretion by gastric CD4+ T cells from H. pylori-infected patients with low-grade gastric MALT lymphoma. All MALT lymphoma-derived clones dose dependently increased their B cell help, whereas clones from chronic gastritis lost helper activity at T-to-B-cell ratios greater than 1. Conclusion: the results obtained indicate that CagY drives both B cell proliferation and T cell activation in gastric MALT lymphomas.
Collapse
Affiliation(s)
- Chiara Della Bella
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (C.D.B.); (M.B.); (A.G.); (J.B.); (F.C.)
| | - Maria Felicia Soluri
- Department of Health Sciences & IRCAD, Università del Piemonte Orientale, 28100 Novara, Italy;
- Center for Translational Research on Autoimmune and Allergic Disease, Università del Piemonte Orientale, 28100 Novara, Italy
| | - Simone Puccio
- Genomic Unit, IRCCS, Humanitas Clinical and Research Center, 20090 Milan, Italy;
| | - Marisa Benagiano
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (C.D.B.); (M.B.); (A.G.); (J.B.); (F.C.)
| | - Alessia Grassi
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (C.D.B.); (M.B.); (A.G.); (J.B.); (F.C.)
| | - Jacopo Bitetti
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (C.D.B.); (M.B.); (A.G.); (J.B.); (F.C.)
| | - Fabio Cianchi
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (C.D.B.); (M.B.); (A.G.); (J.B.); (F.C.)
| | - Daniele Sblattero
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy;
| | - Clelia Peano
- Institute of Genetic and Biomedical Research, UoS Milan, National Research Council, 20090 Milan, Italy;
- Human Technopole, 20157 Milan, Italy
| | - Mario Milco D’Elios
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy; (C.D.B.); (M.B.); (A.G.); (J.B.); (F.C.)
- Correspondence: ; Tel.: +39-055-275-8331
| |
Collapse
|
11
|
Shafiee S, Cegolon L, Khafaei M, Gholami N, Zhao S, Khalesi N, Moosavian H, Fathi S, Izadi M, Ghadian A, Javanbakht M, Javanbakht A, Akhavan-Sigari R. Gastrointestinal cancers, ACE-2/TMPRSS2 expression and susceptibility to COVID-19. Cancer Cell Int 2021; 21:431. [PMID: 34399734 PMCID: PMC8365127 DOI: 10.1186/s12935-021-02129-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 07/30/2021] [Indexed: 12/22/2022] Open
Abstract
Recent studies on the pathophysiology of COVID-19 are indicating that the Angiotensin convertase enzyme 2 (ACE-2) and transmembrane serine protease 2 (TMPRSS2) can act as a major component in the fusion of SARS-Cov-2 with target cells. It has also been observed that the expression of ACE-2 and TMPRSS2 can be altered in malignancies. Shedding light on this matter could be crucial since the COVID-19 pandemic interfered with many gastrointestinal cancer screening programs. Herein we discuss the possibility of severe forms of COVID-19 in patients with gastrointestinal cancers due to the gastrointestinal entry route of SARS-CoV-2 into the human body. The disruption of cancer screening programs caused by the current COVID-19 pandemic could therefore have massive negative health impact on patients affected by gastrointestinal malignancies.
Collapse
Affiliation(s)
- Sepehr Shafiee
- Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Luca Cegolon
- Public Health Department, Local Health Unit N.2 "Marca Trevigiana", 31100, Treviso, Italy
| | - Mostafa Khafaei
- Human Genetics Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Nasrin Gholami
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Shi Zhao
- JC School of Public Health and Primary Care, Chinese University of Hong Kong, Hong Kong, China
| | - Nasrin Khalesi
- Department of Pediatrics, Iran University of Medical Sciences, Tehran, Iran
| | - Hamidreza Moosavian
- Department of Clinical Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Saeid Fathi
- Department of Parasite Vaccine Research and Production, Razi Vaccine and Serum Research Institute, Agriculture Research, Education and Extension Organization (AREEO), Karaj, Iran
| | - Morteza Izadi
- Health Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Alireza Ghadian
- Nephrology and Urology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mohammad Javanbakht
- Nephrology and Urology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | | | | |
Collapse
|
12
|
Zhu R, Lang T, Yan W, Zhu X, Huang X, Yin Q, Li Y. Gut Microbiota: Influence on Carcinogenesis and Modulation Strategies by Drug Delivery Systems to Improve Cancer Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2003542. [PMID: 34026439 PMCID: PMC8132165 DOI: 10.1002/advs.202003542] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 01/05/2021] [Indexed: 05/05/2023]
Abstract
Gut microbiota have close interactions with the host. It can affect cancer progression and the outcomes of cancer therapy, including chemotherapy, immunotherapy, and radiotherapy. Therefore, approaches toward the modulation of gut microbiota will enhance cancer prevention and treatment. Modern drug delivery systems (DDS) are emerging as rational and promising tools for microbiota intervention. These delivery systems have compensated for the obstacles associated with traditional treatments. In this review, the essential roles of gut microbiota in carcinogenesis, cancer progression, and various cancer therapies are first introduced. Next, advances in DDS that are aimed at enhancing the efficacy of cancer therapy by modulating or engineering gut microbiota are highlighted. Finally, the challenges and opportunities associated with the application of DDS targeting gut microbiota for cancer prevention and treatment are briefly discussed.
Collapse
Affiliation(s)
- Runqi Zhu
- State Key Laboratory of Drug Research and Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of Sciences501 Haike RoadShanghai201203China
- School of PharmacyUniversity of Chinese Academy of SciencesBeijing100049China
| | - Tianqun Lang
- State Key Laboratory of Drug Research and Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of Sciences501 Haike RoadShanghai201203China
- School of PharmacyUniversity of Chinese Academy of SciencesBeijing100049China
- Yantai Key Laboratory of Nanomedicine and Advanced PreparationsYantai Institute of Materia MedicaYantai264000China
| | - Wenlu Yan
- State Key Laboratory of Drug Research and Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of Sciences501 Haike RoadShanghai201203China
- School of PharmacyUniversity of Chinese Academy of SciencesBeijing100049China
| | - Xiao Zhu
- State Key Laboratory of Drug Research and Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of Sciences501 Haike RoadShanghai201203China
- School of PharmacyUniversity of Chinese Academy of SciencesBeijing100049China
| | - Xin Huang
- State Key Laboratory of Drug Research and Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of Sciences501 Haike RoadShanghai201203China
- School of PharmacyUniversity of Chinese Academy of SciencesBeijing100049China
| | - Qi Yin
- State Key Laboratory of Drug Research and Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of Sciences501 Haike RoadShanghai201203China
- School of PharmacyUniversity of Chinese Academy of SciencesBeijing100049China
- Yantai Key Laboratory of Nanomedicine and Advanced PreparationsYantai Institute of Materia MedicaYantai264000China
| | - Yaping Li
- State Key Laboratory of Drug Research and Center of PharmaceuticsShanghai Institute of Materia MedicaChinese Academy of Sciences501 Haike RoadShanghai201203China
- School of PharmacyUniversity of Chinese Academy of SciencesBeijing100049China
- Yantai Key Laboratory of Nanomedicine and Advanced PreparationsYantai Institute of Materia MedicaYantai264000China
- School of PharmacyYantai UniversityYantai264005China
| |
Collapse
|
13
|
Wei N, Zhou M, Lei S, Zhong Z, Shi R. A meta-analysis and systematic review on subtypes of gastric intestinal metaplasia and neoplasia risk. Cancer Cell Int 2021; 21:173. [PMID: 33731114 PMCID: PMC7968216 DOI: 10.1186/s12935-021-01869-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 03/06/2021] [Indexed: 12/20/2022] Open
Abstract
Background Gastric intestinal metaplasia (GIM) is a significant risk factor for gastric cancer. Risk of gastric cancer/dysplasia between complete intestinal metaplasia (CIM) and incomplete intestinal metaplasia (IIM) was controversial. Our study aimed to pool relative risk (RR) of cancer/dysplasia of IIM compared with CIM in GIM patients. Methods PubMed, EMBASE, Cochrane Library and Web of Science were searched for studies concerning cancer/dysplasia in GIM patients. Random-effects or fixed-effects model was utilized for pooling RR. Sensitivity and publication bias analyses were conducted. Stability of results would be evaluated in case of publication bias. Results 12 studies were included. Compared with CIM, pooled RR of cancer/dysplasia in IIM patients was 4.48 (95% CI 2.50–8.03), and the RR was 4.96 (95% CI 2.72–9.04) for cancer, and 4.82 (95% CI 1.45–16.0) for dysplasia. The pooled RR for cancer/dysplasia in type III IM was 6.27 (95% CI 1.89–20.77) compared with type II + I IM, while it was 5.55 (95% CI 2.07–14.92) compared with type II IM. Pooled RR between type II IM and type I IM was 1.62 (95% CI 1.16–2.27). Subgroup analyses showed that IIM was associated with a higher risk of gastric cancer/dysplasia in Western population (pooled RR = 4.65 95% CI 2.30–9.42), but not in East Asian population (pooled RR = 4.01 95% CI 0.82–19.61). Conclusions IIM was related to a higher risk of cancer/dysplasia compared with CIM. Risk of developing cancer/dysplasia from type I, II, and III intestinal metaplasia increased gradually. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-01869-0.
Collapse
Affiliation(s)
- Ning Wei
- Medical School of Southeast University, Nanjing, 210009, China.,Department of Gastroenterology, Southeast University Affiliated Zhongda Hospital, No. 87 Dingjiaqiao, Nanjing, 210009, China
| | - Mengyue Zhou
- Medical School of Southeast University, Nanjing, 210009, China.,Department of Gastroenterology, Southeast University Affiliated Zhongda Hospital, No. 87 Dingjiaqiao, Nanjing, 210009, China
| | - Siyu Lei
- Medical School of Southeast University, Nanjing, 210009, China.,Department of Gastroenterology, Southeast University Affiliated Zhongda Hospital, No. 87 Dingjiaqiao, Nanjing, 210009, China
| | - Zhiheng Zhong
- Medical School of Southeast University, Nanjing, 210009, China.,Department of Gastroenterology, Southeast University Affiliated Zhongda Hospital, No. 87 Dingjiaqiao, Nanjing, 210009, China
| | - Ruihua Shi
- Medical School of Southeast University, Nanjing, 210009, China. .,Department of Gastroenterology, Southeast University Affiliated Zhongda Hospital, No. 87 Dingjiaqiao, Nanjing, 210009, China.
| |
Collapse
|
14
|
Xu C, Xie J, Liu Y, Tang F, Long Z, Wang Y, Luo J, Li J, Li G. MicroRNA expression profiling and target gene analysis in gastric cancer. Medicine (Baltimore) 2020; 99:e21963. [PMID: 32925730 PMCID: PMC7489646 DOI: 10.1097/md.0000000000021963] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
This study aims to identify differentially expressed microRNAs (miRNAs) in gastric cancer by comparing gastric cancerous tissues with normal tissues, explore the potential roles.The miRNA expression microarray was employed on gastric cancer tissues, and apparently normal para-cancerous tissues from 3 patients undergoing radical surgery were matched. Quantitative RT-PCR was performed on the other 7 patients to validate the findings of the microarray. Furthermore, Gene Ontology (GO) analysis and enrichment analysis of KEGG Pathway were performed for 5 dysregulated candidate miRNAs, including 3 upregulated (miR-31-3p, miR-6736-3p, and miR-147b) and 2 downregulated (miR-3065-5p and miR-3921) miRNAs, in order to determine the role of miRNAs in tumorigenesis and development.Among these miRNAs, 17 miRNAs were found to be upregulated, and 19 miRNAs were found to be downregulated. The dysregulated expression of 5 candidate miRNAs, including miR-31-3p, miR-147b, miR-6736-3p, miR-3065-5p, and miR-3921, were verified by quantitative RT-PCR in the validation set. Among these miRNAs, miR-31-3p, miR-6736-3p, miR-3065-5p, and miR-3921 had 551 target gene intersections. The GO and KEGG Pathway analyses Revealed that miR-31-3p, miR-6736-3p, miR-3065-5p, and miR-3921 may participate in multiple pathophysiological processes, such as foreign substance metabolism and chemical carcinogenesis.The profile of differentially expressed miRNAs was successfully screened, and 4 miRNAs (i.e., miR-31-3p, miR-6736-3p, miR-3065-5p, and miR-3921) appeared to be involved in gastric carcinogenesis. These might serve as promising biomarkers for gastric cancer.
Collapse
Affiliation(s)
- Chenguang Xu
- Department of Gastroenterology, the Second Affiliated Hospital, University of South China, Hengyang
- Department of Gastroenterology, People's Hospital of Longhua District, Shenzhen
| | - Juan Xie
- Research Lab of translational medicine
- Institute of Pharmacy and Pharmacology, Hengyang Medical College, University of South China, Hengyang
| | - Yanping Liu
- Department of Gastroenterology, the Second Affiliated Hospital, University of South China, Hengyang
| | - Fenfen Tang
- Department of Gastroenterology, the Second Affiliated Hospital, University of South China, Hengyang
| | - Zhi Long
- Department of Gastroenterology, the Second Affiliated Hospital, University of South China, Hengyang
| | - Yaodong Wang
- Department of Gastroenterology, the Second Affiliated Hospital, University of South China, Hengyang
| | - Jiangyan Luo
- Department of Ultrasonography, the Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, China
| | - Junda Li
- Department of Gastroenterology, People's Hospital of Longhua District, Shenzhen
| | - Guoqing Li
- Department of Gastroenterology, the Second Affiliated Hospital, University of South China, Hengyang
| |
Collapse
|
15
|
What are the effects of IL-1β (rs1143634), IL-17A promoter (rs2275913) and TLR4 (rs4986790) gene polymorphism on the outcomes of infection with H. pylori within as Iranian population; A systematic review and meta-analysis. GENE REPORTS 2020. [DOI: 10.1016/j.genrep.2020.100735] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
16
|
Abstract
Understanding the mechanisms involved in induction and regulation of the immune and inflammatory response to Helicobacter pylori is extremely important in determining disease outcomes. H pylori expresses a plethora of factors that influence the host response. Vaccines against H pylori are desperately needed for the prevention of gastric carcinogenesis, especially with the increasing trends in antimicrobial resistance. This review summarizes some important findings, published between 1 April 2019 and 31 March 2020, in the areas of H pylori-mediated inflammation, immunity and vaccines.
Collapse
Affiliation(s)
- Karen Robinson
- School of Medicine, Nottingham Digestive Diseases Biomedical Research Centre, University of Nottingham, Nottingham, UK
| | - Philippe Lehours
- UMR1053 Bordeaux Research In Translational Oncology, INSERM, Univ. Bordeaux, BaRITOn, Bordeaux, France.,French National Reference Centre for Campylobacters & Helicobacters, Hôpital Pellegrin, Bordeaux, France
| |
Collapse
|
17
|
Yoo JY, Cho HJ, Moon S, Choi J, Lee S, Ahn C, Yoo KY, Kim I, Ko KP, Lee JE, Park SK. Pickled Vegetable and Salted Fish Intake and the Risk of Gastric Cancer: Two Prospective Cohort Studies and a Meta-Analysis. Cancers (Basel) 2020; 12:cancers12040996. [PMID: 32316595 PMCID: PMC7225928 DOI: 10.3390/cancers12040996] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/08/2020] [Accepted: 04/14/2020] [Indexed: 12/13/2022] Open
Abstract
An increased risk of gastric cancer for pickled vegetable and salted fish intake has been suggested, yet the lack of a dose-response association warrants a quantitative analysis. We conducted a meta-analysis, combining results from our analysis of two large Korean cohort studies and those from previous prospective cohort studies. We investigated the association of pickled vegetable and salted fish intake with gastric cancer in the Korean Genome Epidemiology Study and the Korean Multi-center Cancer Cohort Study using Cox proportional hazard models. We then searched for observational studies published until November 2019 and conducted both dose-response and categorical meta-analyses. The pooled relative risk (RR) of gastric cancer incidence was 1.15 (95% Confidence Interval (CI), 1.07–1.23) for 40 g/day increment in pickled vegetable intake in a dose-response manner (P for nonlinearity = 0.11). As for salted fish intake, the pooled risk of gastric cancer incidence was 1.17 (95% CI, 0.99–1.38) times higher, comparing the highest to the lowest intake. Our findings supported the evidence that high intake of pickled vegetable and salted fish is associated with elevated risk of gastric cancer incidence.
Collapse
Affiliation(s)
- Jin Young Yoo
- Department of Food and Nutrition, Seoul National University, Seoul 08826, Korea; (J.Y.Y.); (H.J.C.)
| | - Hyun Jeong Cho
- Department of Food and Nutrition, Seoul National University, Seoul 08826, Korea; (J.Y.Y.); (H.J.C.)
| | - Sungji Moon
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul 03080, Korea; (S.M.); (J.C.); (S.L.); (C.A.); (K.-Y.Y.); (S.K.P.)
- Cancer Research Institute, Seoul National University, Seoul 03080, Korea
- Interdisciplinary Program in Cancer Biology Major, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Jeoungbin Choi
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul 03080, Korea; (S.M.); (J.C.); (S.L.); (C.A.); (K.-Y.Y.); (S.K.P.)
- Department of Biomedical Science, Seoul National University Graduate School, Seoul 03080, Korea
| | - Sangjun Lee
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul 03080, Korea; (S.M.); (J.C.); (S.L.); (C.A.); (K.-Y.Y.); (S.K.P.)
- Cancer Research Institute, Seoul National University, Seoul 03080, Korea
- Department of Biomedical Science, Seoul National University Graduate School, Seoul 03080, Korea
| | - Choonghyun Ahn
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul 03080, Korea; (S.M.); (J.C.); (S.L.); (C.A.); (K.-Y.Y.); (S.K.P.)
- Cancer Research Institute, Seoul National University, Seoul 03080, Korea
- Department of Biomedical Science, Seoul National University Graduate School, Seoul 03080, Korea
| | - Keun-Young Yoo
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul 03080, Korea; (S.M.); (J.C.); (S.L.); (C.A.); (K.-Y.Y.); (S.K.P.)
| | - Inah Kim
- Department of Occupational and Environmental Medicine, Hanyang University College of Medicine, Seoul 04763, Korea;
| | - Kwang-Pil Ko
- Department of Preventive Medicine, Gachon University College of Medicine, Incheon 21565, Korea;
| | - Jung Eun Lee
- Department of Food and Nutrition, Seoul National University, Seoul 08826, Korea; (J.Y.Y.); (H.J.C.)
- Research Institute of Human Ecology, Seoul National University, Seoul 08826, Korea
- Correspondence: ; Tel.: +82-2-880-6834
| | - Sue K. Park
- Department of Preventive Medicine, Seoul National University College of Medicine, Seoul 03080, Korea; (S.M.); (J.C.); (S.L.); (C.A.); (K.-Y.Y.); (S.K.P.)
- Cancer Research Institute, Seoul National University, Seoul 03080, Korea
- Department of Biomedical Science, Seoul National University Graduate School, Seoul 03080, Korea
| |
Collapse
|
18
|
Zhu Q, Wu X, Tang M, Wu L. Observation of tumor-associated macrophages expression in gastric cancer and its clinical pathological relationship. Medicine (Baltimore) 2020; 99:e19839. [PMID: 32332633 PMCID: PMC7220635 DOI: 10.1097/md.0000000000019839] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The present study was designed to investigate the expression of tumor-associated macrophages (TAMs) in gastric cancer and its clinicopathological relationship. In addition, we also aimed to analyze the relationship between helicobacter pylori (HP) infection and TAMs in gastric cancer.The protein expression of CD16 and CD163 in 90 gastric cancer tissues and 30 margin tissues was detected by immunohistochemistry. HP infection was detected in 90 gastric cancer tissues and 30 margin tissues by gram staining and immunohistochemistry.There was no clear correlation between CD16 macrophages and gastric cancer. The density of CD163 macrophages was not correlated with the general condition of tumor patients, but with tumor size, tumor differentiation, lymphatic metastasis, depth of invasion and TNM stage. Additionally, the infection rate of HP in gastric cancer tissues was significantly higher.In summary, TAMs are associated with tumor size, degree of differentiation, depth of invasion, lymph node metastasis and TNM stage, suggesting their critical role in the invasion and metastasis of gastric cancer.
Collapse
Affiliation(s)
- Qing Zhu
- Departments of Pathology, The First Affiliated Hospital of Bengbu Medical College
- Department of Pathology, Bengbu Medical College
| | - Xia Wu
- Class 2018, Clinical Pathology, The Graduate School, Bengbu Medical College
| | - Mingyang Tang
- Class 2016, School of Clinical Medicine, Bengbu Medical College, Anhui, China
| | - Ligao Wu
- Departments of Pathology, The First Affiliated Hospital of Bengbu Medical College
- Department of Pathology, Bengbu Medical College
| |
Collapse
|
19
|
Xue W, Xu X, Tan Y, Wang Y, Wang H, Xu Y, Xi C, Jiang P, Ding W. Evaluation and validation of the prognostic value of nutrition and immunity parameters in gastric cancer after R0 resection. Medicine (Baltimore) 2020; 99:e19270. [PMID: 32080137 PMCID: PMC7034686 DOI: 10.1097/md.0000000000019270] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Precise predictive tools are critical for choosing the individualized treatment protocols and follow-up procedures for patients with gastric cancer (GC). In this study, we aimed to evaluate and validate the prognostic abilities of preoperative nutrition and immunity parameters in GC after curative R0 resection.We established two nomograms based on 437 patients who underwent curative radical gastrectomy for gastric cancer to predict the postoperative overall survival (OS) and recurrence-free survival (RFS), and then compared the predictive accuracy and discriminative ability of the nomograms with the TNM stage systems for GC. An internal validation cohort of 141 patients and an external validation cohort of 116 patients were used to validate the result.The independent predictive factors for OS or RFS, including T stage, N stage, differentiated degree, neutrophil monocyte lymphocyte ratio (NMLR) and albumin globulin ratio (AGR) were used to establish the 2 nomograms. The C-index of the OS nomogram was 0.802, which was higher than that of the AGR, the NMLR and the TNM stage. The C-index of the RFS nomogram was 0.850, which was higher than that of the AGR, the NMLR and the TNM stage. Analogously, the areas under the receiver operating characteristics curves (AUROCs, 0.920 for OS and 0.897 for RFS, respectively) of the two nomograms were higher than that of the NMLR, the AGR and the TNM stage. In the internal validation cohort, the C-indexes of the OS and RFS nomograms were 0.812 and 0.826, respectively. In the external validation cohort, the C-indexes of the OS and RFS nomograms were 0.866 and 0.880, respectively.The proposed nomograms including nutrition and immunity parameters were proved to have excellent predictive ability in survival and recurrence for patients with GC after R0 resection.
Collapse
Affiliation(s)
- Wenbo Xue
- Department of General Surgery, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu Province
- The Wujin Clinical college of Xuzhou Medical University, Changzhou
| | - Xuezhong Xu
- Department of General Surgery, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu Province
- The Wujin Clinical college of Xuzhou Medical University, Changzhou
| | - Yulin Tan
- Department of General Surgery, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu Province
- The Wujin Clinical college of Xuzhou Medical University, Changzhou
| | - Yibo Wang
- Department of General Surgery, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu Province
- The Wujin Clinical college of Xuzhou Medical University, Changzhou
| | - Hao Wang
- The Wujin Clinical college of Xuzhou Medical University, Changzhou
- Department of Medical Record, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu Province, China
| | - Yixin Xu
- Department of General Surgery, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu Province
- The Wujin Clinical college of Xuzhou Medical University, Changzhou
| | - Cheng Xi
- Department of General Surgery, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu Province
- The Wujin Clinical college of Xuzhou Medical University, Changzhou
| | - Peng Jiang
- Department of General Surgery, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu Province
- The Wujin Clinical college of Xuzhou Medical University, Changzhou
| | - Wei Ding
- Department of General Surgery, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu Province
- The Wujin Clinical college of Xuzhou Medical University, Changzhou
| |
Collapse
|
20
|
Ansari S, Yamaoka Y. Helicobacter pylori Virulence Factors Exploiting Gastric Colonization and its Pathogenicity. Toxins (Basel) 2019; 11:E677. [PMID: 31752394 PMCID: PMC6891454 DOI: 10.3390/toxins11110677] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 11/15/2019] [Accepted: 11/16/2019] [Indexed: 02/07/2023] Open
Abstract
Helicobacter pylori colonizes the gastric epithelial cells of at least half of the world's population, and it is the strongest risk factor for developing gastric complications like chronic gastritis, ulcer diseases, and gastric cancer. To successfully colonize and establish a persistent infection, the bacteria must overcome harsh gastric conditions. H. pylori has a well-developed mechanism by which it can survive in a very acidic niche. Despite bacterial factors, gastric environmental factors and host genetic constituents together play a co-operative role for gastric pathogenicity. The virulence factors include bacterial colonization factors BabA, SabA, OipA, and HopQ, and the virulence factors necessary for gastric pathogenicity include the effector proteins like CagA, VacA, HtrA, and the outer membrane vesicles. Bacterial factors are considered more important. Here, we summarize the recent information to better understand several bacterial virulence factors and their role in the pathogenic mechanism.
Collapse
Affiliation(s)
- Shamshul Ansari
- Department of Microbiology, Chitwan Medical College and Teaching Hospital, Bharatpur 44200, Chitwan, Nepal;
| | - Yoshio Yamaoka
- Department of Environmental and Preventive Medicine, Oita University Faculty of Medicine, Idaigaoka, Hasama-machi, Yufu, Oita 879-5593, Japan
- Global Oita Medical Advanced Research Center for Health, Idaigaoka, Hasama-machi, Yufu, Oita 879-5593, Japan
- Department of Medicine, Gastroenterology and Hepatology Section, Baylor College of Medicine, 2002 Holcombe Blvd., Houston, TX 77030, USA
- Borneo Medical and Health Research Centre, Universiti Malaysia Sabah, Kota Kinabaru, Sabah 88400, Malaysia
| |
Collapse
|
21
|
Effect of Peptides from Alaska Pollock on Intestinal Mucosal Immunity Function and Purification of Active Fragments. Nutrients 2019; 11:nu11102517. [PMID: 31635335 PMCID: PMC6835260 DOI: 10.3390/nu11102517] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 10/01/2019] [Accepted: 10/12/2019] [Indexed: 12/23/2022] Open
Abstract
The intestinal mucosal barrier plays an important role in systemic immune functions. This study aimed to find the mechanism of peptide from Alaska Pollock (APP) on intestinal mucosal immunity in mice induced by cyclophosphamide (Cy). Cy-induced decreases of body weight and index of immune organ were significantly improved by APP as compared with Cy group (p < 0.05). APP could promote the secretion of SIgA and IgA on intestinal mucosa (p < 0.05) and mainly had an impact on the final differentiation of IgA+ B cell, thereby promoting the secretion of plasma cells, which can be corroborated by the increases of IL-6 and IL-10 (p < 0.05). APP with high immune activity was separated and two peptides were purified and identified as Gly–Val–Ile–Lys and Ala–Cys–Asn–Gly–Arg. Therefore, APP can be considered as beneficial ingredients to protect the intestinal barrier disruption induced by Cy.
Collapse
|
22
|
Jafarzadeh A, Nemati M, Jafarzadeh S. The important role played by chemokines influence the clinical outcome of Helicobacter pylori infection. Life Sci 2019; 231:116688. [PMID: 31348950 DOI: 10.1016/j.lfs.2019.116688] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 06/30/2019] [Accepted: 07/22/2019] [Indexed: 02/07/2023]
Abstract
The extended infection with Helicobacter pylori (H. pylori), one of the most frequent infectious agents in humans, may cause gastritis, peptic ulcers, gastric mucosa-associated lymphoid tissue (MALT) lymphoma, and gastric cancer. During H. pylori infection, different kinds of inflammatory cells such as dendritic cells, macrophages, neutrophils, mast cells, eosinophils, T cells and B cells are accumulated into the stomach. The interactions between chemokines and their respective receptors recruit particular types of the leukocytes that ultimately determine the nature of immune response and therefore, have a main influence on the consequence of infection. The suitable production of chemokines especially in the early stages of H. pylori infection shapes appropriate immune responses that contribute to the H. pylori elimination. The unbalanced expression of the chemokines can contribute in the induction of inappropriate responses that result in the tissue damage or malignancy. Thus, chemokines and their receptors may be promising potential targets for designing the therapeutic strategies against various types H. pylori-related gastrointestinal disorders. In this review, a comprehensive explanation regarding the roles played by chemokines in H. pylori-mediated peptic ulcer, gastritis and gastric malignancies was provided while presenting the potential utilization of these chemoattractants as therapeutic elements.
Collapse
Affiliation(s)
- Abdollah Jafarzadeh
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran; Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| | - Maryam Nemati
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran; Department of Hematology and Laboratory Sciences, School of Para-Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Sara Jafarzadeh
- Student Research Committee, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
23
|
Pero R, Brancaccio M, Laneri S, Biasi MGD, Lombardo B, Scudiero O. A Novel View of Human Helicobacter pylori Infections: Interplay between Microbiota and Beta-Defensins. Biomolecules 2019; 9:biom9060237. [PMID: 31216758 PMCID: PMC6627275 DOI: 10.3390/biom9060237] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 06/11/2019] [Accepted: 06/13/2019] [Indexed: 02/07/2023] Open
Abstract
The gut microbiota is significantly involved in the preservation of the immune system of the host, protecting it against the pathogenic bacteria of the stomach. The correlation between gut microbiota and the host response supports human gastric homeostasis. Gut microbes may be shifted in Helicobacter pylori (Hp)-infected individuals to advance gastric inflammation and distinguished diseases. Particularly interesting is the establishment of cooperation between gut microbiota and antimicrobial peptides (AMPs) of the host in the gastrointestinal tract. AMPs have great importance in the innate immune reactions to Hp and participate in conservative co-evolution with an intricate microbiome. β-Defensins, a class of short, cationic, arginine-rich proteins belonging to the AMP group, are produced by epithelial and immunological cells. Their expression is enhanced during Hp infection. In this review, we discuss the impact of the gut microbiome on the host response, with particular regard to β-defensins in Hp-associated infections. In microbial infections, mostly in precancerous lesions induced by Hp infection, these modifications could lead to different outcomes.
Collapse
Affiliation(s)
- Raffaela Pero
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II", 80131 Napoli, Italy.
- Task Force sugli Studi del Microbioma, Università degli Studi di Napoli "Federico II", 80131 Napoli, Italy.
| | - Mariarita Brancaccio
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, 80121 Napoli, Italy.
| | - Sonia Laneri
- Dipartimento di Farmacia, Università degli Studi di Napoli "Federico II", Via Montesano 49, 80131 Napoli, Italy.
| | | | - Barbara Lombardo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II", 80131 Napoli, Italy.
- CEINGE-Biotecnologie Avanzate Scarl, Via G. Salvatore 486, 80145 Napoli, Italy.
| | - Olga Scudiero
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II", 80131 Napoli, Italy.
- Task Force sugli Studi del Microbioma, Università degli Studi di Napoli "Federico II", 80131 Napoli, Italy.
- CEINGE-Biotecnologie Avanzate Scarl, Via G. Salvatore 486, 80145 Napoli, Italy.
| |
Collapse
|
24
|
Roviello G, D’Angelo A, Generali D, Pittacolo M, Ganzinelli M, Iezzi G, Manzini ND, Sobhani N. Avelumab in gastric cancer. Immunotherapy 2019; 11:759-768. [DOI: 10.2217/imt-2019-0011] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer (GC) is the fifth most common malignancy and the third cause of cancer-related deaths worldwide. Currently, surgery and chemotherapy remain the main therapeutic options and the prognosis of the disease is still poor in the metastatic setting. Avelumab is a human IgG1 antibody directed against PD-L1 approved for Merkel cell carcinoma and urothelial carcinoma that could be useful also for the treatment of GC. This review describes the chemical structure, the pharmacologic properties and the current knowledge of the efficacy of avelumab in the treatment of GC from the data available on the first and later phase clinical trials. The ongoing studies testing this drug either alone or in combination with other drugs are also described.
Collapse
Affiliation(s)
- Giandomenico Roviello
- Department of Health Sciences, University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy
| | - Alberto D’Angelo
- Department of Biology & Biochemistry, University of Bath, Bath BA2 7AY, United Kingdom
| | - Daniele Generali
- Breast Cancer Unit, ASST Cremona, Viale Concordia 1, 26100 Cremona, Italy
| | - Matteo Pittacolo
- Department of Orthopedics & Orthopedic Oncology, University of Padova, Italy
| | - Monica Ganzinelli
- Thoracic Unit, Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | - Nicolò de Manzini
- General Surgery Unit, Department of Medical, Surgical and Health Sciences Cattinara University Hospital, Azienda Sanitaria Universitaria Integrata di Trieste, Trieste, Italy
| | - Navid Sobhani
- Breast Cancer Unit, ASST Cremona, Viale Concordia 1, 26100 Cremona, Italy
| |
Collapse
|
25
|
Bifidobacteria and Mucosal-Associated Invariant T (MAIT) Cells: A New Approach to Colorectal Cancer Prevention? GASTROINTESTINAL DISORDERS 2019. [DOI: 10.3390/gidisord1020022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Colorectal cancer is the most preventable form of cancer worldwide. The pathogenesis of colorectal cancer includes gut inflammation, genetic and microbial composition factors. İmpairment of the gut microbiota has been associated with development of colorectal cancer. The genus Bifidobacterium is an important component of the commensal gut microbiota. Bifidobacteria are considered to have important roles in multiple homeostatic functions: immunologic, hormonal and metabolic. Mucosal-associated invariant T cells (MAIT) are components of the immune system involved in protection against infectious pathogens and regulate the pathogenesis of various inflammatory diseases and, potentially, colorectal cancer. Engagement between Bifidobacterium and MAIT cells could exert a beneficial effect on colorectal cancer prevention and treatment.
Collapse
|
26
|
Caston RR, Loh JT, Voss BJ, McDonald WH, Scholz MB, McClain MS, Cover TL. Effect of environmental salt concentration on the Helicobacter pylori exoproteome. J Proteomics 2019; 202:103374. [PMID: 31063819 DOI: 10.1016/j.jprot.2019.05.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 04/20/2019] [Accepted: 05/02/2019] [Indexed: 02/07/2023]
Abstract
Helicobacter pylori infection and a high salt diet are each risk factors for gastric cancer. In this study, we tested the hypothesis that environmental salt concentration influences the composition of the H. pylori exoproteome. H. pylori was cultured in media containing varying concentrations of sodium chloride, and aliquots were fractionated and analyzed by liquid chromatography-tandem mass spectrometry (LC-MS/MS). We identified proteins that were selectively released into the extracellular space, and we identified selectively released proteins that were differentially abundant in culture supernatants, depending on the environmental salt concentration. We also used RNA-seq analysis to identify genes that were differentially expressed in response to environmental salt concentration. The salt-responsive proteins identified by proteomic analysis and salt-responsive genes identified by RNA-seq analysis were mostly non-concordant, but the secreted toxin VacA was salt-responsive in both analyses. Western blot analysis confirmed that VacA levels in the culture supernatant were increased in response to high salt conditions, and quantitative RT-qPCR experiments confirmed that vacA transcription was upregulated in response to high salt conditions. These results indicate that environmental salt concentration influences the composition of the H. pylori exoproteome, which could contribute to the increased risk of gastric cancer associated with a high salt diet. SIGNIFICANCE: Helicobacter pylori-induced alterations in the gastric mucosa have been attributed, at least in part, to the actions of secreted H. pylori proteins. In this study, we show that H. pylori growth in high salt concentrations leads to increased levels of a secreted VacA toxin. Salt-induced alterations in the composition of the H. pylori exoproteome is relevant to the increased risk of gastric cancer associated with consumption of a high salt diet.
Collapse
Affiliation(s)
- Rhonda R Caston
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - John T Loh
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Bradley J Voss
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - W Hayes McDonald
- Proteomics Laboratory, Mass Spectrometry Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA; Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Matthew B Scholz
- Vanderbilt Technologies for Advanced Genetics (VANTAGE), Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mark S McClain
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Timothy L Cover
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA; Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA; Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN, USA.
| |
Collapse
|
27
|
Troilo A, Grassi A, Petrone L, Cianchi F, Benagiano M, Bella CD, Capitani N, Bitetti J, D'Elios S, Tapinassi S, Azzurri A, Alnwaisri H, Romagnoli J, Bizzaro N, Bergman M, Baldari CT, D'Elios MM. Intrinsic factor recognition promotes T helper 17/T helper 1 autoimmune gastric inflammation in patients with pernicious anemia. Oncotarget 2019; 10:2921-2929. [PMID: 31080562 PMCID: PMC6499598 DOI: 10.18632/oncotarget.26874] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 04/03/2019] [Indexed: 02/06/2023] Open
Abstract
The intrinsic factor is the major humoral autoantigen in pernicious anemia/autoimmune gastritis. Although many studies have examined the autoantibody response to intrinsic factor and H+,K+-ATPase, no information is available on possible pathogenic mechanisms mediated by intrinsic factor - specific gastric T cells. Aim of this study was to investigate intrinsic factor-specific T cells in the gastric mucosa of pernicious anemia patients and define their functional properties. For the first time we provide evidence that gastric mucosa of pernicious anemia patients harbour a high proportion (20%) of autoreactive activated CD4+ T-cell clones that specifically recognize intrinsic factor. Most of these clones (94%) showed a T helper 17 or T helper 1 profile. All intrinsic factor-specific clones produced tumor necrosis factor-α, interleukin-21 and provided substantial help for B-cell immunoglobulin production. Most mucosa-derived intrinsic factor-specific T-cell clones expressed cytotoxicity against target cells. Our results indicate that activation of intrinsic factor-specific T helper 17 and T helper 1 T cells in the gastric mucosa represent a key effector mechanism in pernicious anemia suggesting that the T helper 17/T helper 1 pathway may represent a novel target for the prevention and treatment of the disease.
Collapse
Affiliation(s)
- Arianna Troilo
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Alessia Grassi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Luisa Petrone
- Endocrinology Unit, Careggi Hospital, Florence, Italy
| | - Fabio Cianchi
- Department of Surgery, University of Florence, Florence, Italy
| | - Marisa Benagiano
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Chiara Della Bella
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Nagaja Capitani
- Department of Life Sciences, University of Siena, Siena, Italy
| | - Jacopo Bitetti
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Sofia D'Elios
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Simona Tapinassi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | | | - Heba Alnwaisri
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | | | | | - Mathijs Bergman
- Amsterdam Institute for Molecules, Medicines and Systems, Vrije University, Amsterdam, The Netherlands
| | | | - Mario Milco D'Elios
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| |
Collapse
|
28
|
Mutated Rnf43 Aggravates Helicobacter Pylori-Induced Gastric Pathology. Cancers (Basel) 2019; 11:cancers11030372. [PMID: 30884828 PMCID: PMC6468876 DOI: 10.3390/cancers11030372] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 03/10/2019] [Accepted: 03/13/2019] [Indexed: 12/24/2022] Open
Abstract
The E3 ubiquitin ligase ring finger protein 43 (RNF43) is frequently mutated in gastric tumors and loss of RNF43 expression was suggested to be one of the key events during the transition from adenoma to gastric carcinoma. Functional studies on RNF43 have shown that it acts as a tumor suppressor by negatively regulating Wnt signaling. Interestingly, we observed that RNF43H292R/H295R mice bearing two point mutations in the ring domain displayed thickening of the mucosa at early age but did not develop neoplasia. In this study, we infected these mice for 6 months with Helicobacter pylori, which has been described as one of the major risk factors for gastric cancer. Mice bearing mutant RNF43H292R/H295R showed higher gastritis scores upon H. pylori infection compared to wild-type mice, accompanied by increased lymphocyte infiltration and Ifng levels. Furthermore, infected Rnf43 mutant mice developed atrophy, hyperplasia and MUC2 expressing metaplasia and displayed higher levels of the gastric stem cell marker CD44 and canonical NF-κB signaling. In summary, our results show that transactivating mutations in the tumor suppressor Rnf43 can worsen H. pylori induced pathology.
Collapse
|