51
|
Tahara T, Tahara S, Horiguchi N, Kawamura T, Okubo M, Nagasaka M, Nakagawa Y, Shibata T, Urano M, Tsukamoto T, Kuroda M, Ohmiya N. Gastric Mucosal Microarchitectures Associated with Irreversibility with Helicobacter pylori Eradication and Downregulation of Micro RNA (miR)-124a. Cancer Invest 2019; 37:417-426. [PMID: 31483161 DOI: 10.1080/07357907.2019.1663207] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
To investigate the molecular mechanisms of gastric carcinogenesis after Helicobacter pylori (H. pylori) eradication, expression of miR-124a, miR-34b, and miR-34c was examined in nonneoplastic gastric specimens after successful H. pylori eradication. The magnifying narrow-band imaging (NBI) endoscopic features of gastric mucosa were also examined. The atrophic type, an informative endoscopic feature for histological intestinal metaplasia, showed lower expression of miR-124a. Lower expression of miR-124a correlated with hypermethylation of the miR-124a3 locus. The atrophic type represents gastric microarchitectures associated with irreversibility with H. pylori eradication and downregulation of miR-124a.
Collapse
Affiliation(s)
- Tomomitsu Tahara
- Department of Gastroenterology, Fujita Health University School of Medicine , Toyoake , Japan
| | - Sayumi Tahara
- Department of Diagnostic Pathology I, School of Medicine, Fujita Health University , Toyoake , Japan
| | - Noriyuki Horiguchi
- Department of Gastroenterology, Fujita Health University School of Medicine , Toyoake , Japan
| | - Tomohiko Kawamura
- Department of Gastroenterology, Fujita Health University School of Medicine , Toyoake , Japan
| | - Masaaki Okubo
- Department of Gastroenterology, Fujita Health University School of Medicine , Toyoake , Japan
| | - Mitsuo Nagasaka
- Department of Gastroenterology, Fujita Health University School of Medicine , Toyoake , Japan
| | - Yoshihito Nakagawa
- Department of Gastroenterology, Fujita Health University School of Medicine , Toyoake , Japan
| | - Tomoyuki Shibata
- Department of Gastroenterology, Fujita Health University School of Medicine , Toyoake , Japan
| | - Makoto Urano
- Department of Diagnostic Pathology I, School of Medicine, Fujita Health University , Toyoake , Japan
| | - Tetsuya Tsukamoto
- Department of Diagnostic Pathology I, School of Medicine, Fujita Health University , Toyoake , Japan
| | - Makoto Kuroda
- Department of Diagnostic Pathology I, School of Medicine, Fujita Health University , Toyoake , Japan
| | - Naoki Ohmiya
- Department of Gastroenterology, Fujita Health University School of Medicine , Toyoake , Japan
| |
Collapse
|
52
|
Eslami M, Yousefi B, Kokhaei P, Arabkari V, Ghasemian A. Current information on the association of Helicobacter pylori with autophagy and gastric cancer. J Cell Physiol 2019; 234:14800-14811. [PMID: 30784066 DOI: 10.1002/jcp.28279] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 11/23/2018] [Accepted: 11/30/2018] [Indexed: 01/24/2023]
Abstract
Helicobacter pylori (H. pylori) is a Gram-negative bacterium and causative agent of gastric cancer. H. pylori induce defective autophagy or inhibit it by means of CagA and vacuolating cytotoxin A (VacA) toxins leading to the gastric cancer induction. Impaired or defective autophagy leads to the accumulation of cytotoxic materials, such as ROS and P62 that lead to increased mutations in the DNA, genome instability, and risk of cancer formation. H. pylori CagA may inhibit autophagy through the c-Met-PI3k/Akt-mTOR signaling pathway. However, VacA induces autophagy by some signaling pathways. In the gastric epithelial cells, VacA is a necessary and sufficient factor for the creation of autophagy. While CagA is a negative regulator of this phenomenon, the elimination of this gene from H. pylori has increased autophagy and the production of inflammatory cytokines is reduced. In gastrointestinal cancers, some of the microRNAs (miRNAs) act as tumor suppressors and some other are oncogenes by regulating various genes expression. H. pylori can also modify autophagy through a mechanism that includes the function of miRNAs. In autophagy, oncogenic miRNAs inhibit activation of some tumor suppressor signaling pathways (e.g., ULK1 complex, Beclin-1 function, and Atg4 messaging), whereas tumor suppressor miRNAs can block the activation of oncogenic signaling pathways. For instance, Beclin-1 is negatively regulated by miRNA-376b (oncogenic miRNA) and miRNA-30a (tumor suppressor miRNA). Similarly, Atg4 by miRNA-376b (oncogenic miRNA) and miRNA-101 (tumor suppressor miRNA). So, this apparent paradox can be explained as that both Beclin-1 and Atg4 play different roles in a particular cell or tissue.
Collapse
Affiliation(s)
- Majid Eslami
- Department of Bacteriology and Virology, Semnan University of Medical Sciences, Semnan, Iran
| | - Bahman Yousefi
- Department of Immunology, Semnan University of Medical Sciences, Semnan, Iran
| | - Parviz Kokhaei
- Cancer Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Immune and Gene Therapy Lab, Cancer Centre Karolinska, Karolinska University Hospital, Stockholm, Sweden
| | - Vahid Arabkari
- Discipline of Pathology, Lambe Institute for Translational Research, Clinical Science Institute, School of Medicine, National University of Ireland, Galway, Ireland
| | | |
Collapse
|
53
|
Peng C, Li NS, Hu Y, Lu NH. Impact factors that modulate gastric cancer risk in Helicobacter pylori-infected rodent models. Helicobacter 2019; 24:e12580. [PMID: 30950162 DOI: 10.1111/hel.12580] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 02/08/2019] [Accepted: 02/26/2019] [Indexed: 12/24/2022]
Abstract
Gastric cancer causes a large social and economic burden to humans. Helicobacter pylori (H pylori) infection is a major risk factor for distal gastric cancer. Detailed elucidation of H pylori pathogenesis is significant for the prevention and treatment of gastric cancer. Animal models of H pylori-induced gastric cancer have provided an invaluable resource to help elucidate the mechanisms of H pylori-induced carcinogenesis as well as the interaction between host and the bacterium. Rodent models are commonly used to study H pylori infection because H pylori-induced pathological processes in the stomachs of rodents are similar to those in the stomachs of humans. The risk of gastric cancer in H pylori-infected animal models is greatly dependent on host factors, bacterial determinants, environmental factors, and microbiota. However, the related mechanisms and the effects of the interactions among these impact factors on gastric carcinogenesis remain unclear. In this review, we summarize the impact factors mediating gastric cancer risk when establishing H pylori-infected animal models. Clarifying these factors and their potential interactions will provide insights to construct animal models of gastric cancer and investigate the in-depth mechanisms of H pylori pathogenesis, which might contribute to the management of H pylori-associated gastric diseases.
Collapse
Affiliation(s)
- Chao Peng
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Nian-Shuang Li
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yi Hu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Nong-Hua Lu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
54
|
Suk FM, Wang MH, Ho YS. A historical review of classic Helicobacter pylori publications in science citation index expanded. COLLNET JOURNAL OF SCIENTOMETRICS AND INFORMATION MANAGEMENT 2019; 13:313-330. [DOI: 10.1080/09737766.2020.1716645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Fat-Moon Suk
- Division of Gastroenterology, Department of Internal Medicine, Taipei Municipal Wan Fang Hospital, Taipei Medical University, Taipei City 110, Taiwan, R.O.C
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei City 110, Taiwan, R.O.C
| | - Ming-Huang Wang
- Trend Research Centre, Asia University, No. 500, Lioufeng Road, Wufeng, Taichung County 41354, Taiwan, R.O.C
| | - Yuh-Shan Ho
- Trend Research Centre, Asia University, No. 500, Lioufeng Road, Wufeng, Taichung County 41354, Taiwan, R.O.C
| |
Collapse
|
55
|
Sethi V, Vitiello GA, Saxena D, Miller G, Dudeja V. The Role of the Microbiome in Immunologic Development and its Implication For Pancreatic Cancer Immunotherapy. Gastroenterology 2019; 156:2097-2115.e2. [PMID: 30768986 DOI: 10.1053/j.gastro.2018.12.045] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 12/09/2018] [Accepted: 12/17/2018] [Indexed: 12/20/2022]
Abstract
Our understanding of the microbiome and its role in immunity, cancer initiation, and cancer progression has evolved significantly over the past century. The "germ theory of cancer" was first proposed in the early 20th century, and shortly thereafter the bacterium Helicobacter pylori, and later Fusobacterium nucleatum, were implicated in the development of gastric and colorectal cancers, respectively. However, with the development of reliable mouse models and affordable sequencing technologies, the most fascinating aspect of the microbiome-cancer relationship, where microbes undermine cancer immune surveillance and indirectly promote oncogenesis, has only recently been described. In this review, we highlight the essential role of the microbiome in immune system development and maturation. We review how microbe-induced immune activation promotes oncogenesis, focusing particularly on pancreatic carcinogenesis, and show that modulation of the microbiome augments the anti-cancer immune response and enables successful immunotherapy against pancreatic cancer.
Collapse
Affiliation(s)
- Vrishketan Sethi
- Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida
| | - Gerardo A Vitiello
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York
| | - Deepak Saxena
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York; Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, New York
| | - George Miller
- S. Arthur Localio Laboratory, Department of Surgery, New York University School of Medicine, New York, New York
| | - Vikas Dudeja
- Department of Surgery, Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida.
| |
Collapse
|
56
|
Efficacy and Long-Term Safety of H. pylori Eradication for Gastric Cancer Prevention. Cancers (Basel) 2019; 11:cancers11050593. [PMID: 31035365 PMCID: PMC6562927 DOI: 10.3390/cancers11050593] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/12/2019] [Accepted: 04/25/2019] [Indexed: 02/07/2023] Open
Abstract
Helicobacter pylori (H. pylori) has been shown to be a causal factor of gastric cancer in cohort studies and animal models. Meta-analysis of case-control studies nested within prospective cohorts showed that H. pylori infection was associated with a 5.9-fold increased risk of non-cardia gastric cancer. Prospective cohort studies showed that gastric cancer developed in 1–4% of H. pylori-infected subjects. Gastric cancer was successfully induced in Mongolian gerbils and insulin-gastrin (INS-GAS) transgenic mice after inoculation of H. pylori. Meta-analysis of randomized control trials also showed that eradication of H. pylori may reduce the risk of gastric cancer. However, there are several concerns regarding the widespread use of antibiotics to prevent gastric cancer, including the emergence of antibiotic resistance and the perturbation of gut microbiota after H. pylori eradication. Recent studies showed that eradication of H. pylori resulted in an increase in the bacterial diversity and restoration of the relative abundance of other bacteria to levels similar to H. pylori non-infected subjects in the gastric microbiota. The administration of antibiotics may also alter the composition of intestinal microbiota. The α-diversity and β-diversity of fecal microbiota are significantly altered immediately after H. pylori eradication but are gradually restored to levels similar to those before therapy. Yet, the rate of recovery varies with regimens. The diversity was restored at week 8 after triple therapy but was not yet fully recovered at 1 year after concomitant and quadruple therapies. Some studies showed that supplementation of probiotics may reduce the dysbiosis during H. pylori eradication therapy. Although some earlier studies showed high levels of macrolide resistance after triple therapy, recent studies showed that the increased antibiotic resistance rate may be restored 2–12 months after eradication therapy. These results collectively provide evidence of the long-term safety of H. pylori eradication. Yet, more prospective cohort studies and randomized trials are warranted to assess the efficacy and long-term safety of H. pylori eradication for gastric cancer prevention.
Collapse
|
57
|
Wang J, Yao Y, Zhang Q, Li S, Tang L. Inflammatory responses induced by Helicobacter pylori on the carcinogenesis of gastric epithelial GES‑1 cells. Int J Oncol 2019; 54:2200-2210. [PMID: 31081048 DOI: 10.3892/ijo.2019.4775] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 01/01/2019] [Indexed: 11/06/2022] Open
Abstract
Helicobacter pylori (HP) is a pathogenic bacterium associated with chronic gastritis, gastric ulcer and gastric cancer. In the present study, the primary carcinogenesis process of normal gastric epithelial cells (GES‑1) infected with HP was investigated. It was determined that infected gastric mucosal epithelial GES‑1 cells secreted increased interleukin‑8 (IL‑8) and IL‑23, and exhibited enhanced expression of inducible nitric oxide synthase and cyclooxygenase‑2, inducing inflammatory reactions and resulting in apoptosis. The bacterial infection significantly increased the expression of carcinogenesis‑associated genes, including p16, c‑Myc, p53 and p21, as well as the expression of cell surface signaling molecules cluster of differentiation 44 (CD44) and CD54 in GES‑1 cells or tissues of patients with gastritis and gastric cancer in vitro or in vivo. Simultaneously, the migration and invasion abilities of normal gastric epithelial GES‑1 cells were increased following HP infection. These observations demonstrated that the inflammatory response of HP infection could cause normal gastric epithelial cells to undergo significant cancerous reactions, indicating that HP is a risk factor for gastric cancer.
Collapse
Affiliation(s)
- Jianjun Wang
- Department of Clinical Laboratory, Kunshan First People's Hospital, Affiliated to Jiangsu University, Kunshan, Jiangsu 215300, P.R. China
| | - Yongliang Yao
- Department of Clinical Laboratory, Kunshan First People's Hospital, Affiliated to Jiangsu University, Kunshan, Jiangsu 215300, P.R. China
| | - Qinghui Zhang
- Department of Clinical Laboratory, Kunshan First People's Hospital, Affiliated to Jiangsu University, Kunshan, Jiangsu 215300, P.R. China
| | - Shasha Li
- Department of Clinical Laboratory, Kunshan First People's Hospital, Affiliated to Jiangsu University, Kunshan, Jiangsu 215300, P.R. China
| | - Lijun Tang
- Molecular Biology Research Center, School of Life Science, Central South University, Changsha, Hunan 410078, P.R. China
| |
Collapse
|
58
|
Li G, Yu S, Xu J, Zhang X, Ye J, Wang Z, He Y. The prognostic role of Helicobacter pylori in gastric cancer patients: A meta-analysis. Clin Res Hepatol Gastroenterol 2019; 43:216-224. [PMID: 30361060 DOI: 10.1016/j.clinre.2018.08.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 07/04/2018] [Accepted: 08/21/2018] [Indexed: 02/04/2023]
Abstract
BACKGROUND The prognostic value of Helicobacter pylori (H. pylori) infection in gastric cancer patients has been investigated over many years; however, the results remain inconclusive. Thus, we performed a comprehensive review of currently available evidence via a systemic meta-analysis to evaluate the effects of H. pylori infection on the prognosis of gastric cancer patients. METHODS Studies that evaluated the prognostic value of H. pylori infection in gastric cancer were extracted in March 2016 by searching PubMed, EMBASE, and the Cochrane Central Register of Controlled Trials. We obtained or calculated hazard ratios (HRs) and the associated 95% confidence intervals (CIs) from the identified studies, and conducted random-effects model analyses of overall survival and progression-free survival. Twenty-four studies with a cumulative sample size of 7191 patients were included in our analysis. RESULTS Our meta-analysis revealed that H. pylori infection is an indicator of improved overall survival in gastric cancer patients (HR, 0.79; 95% CI, 0.64-0.99); however, this was only true for European patients. The benefits of H. pylori infection were not detected in Asian gastric cancer patients (HR, 1.01; 95% CI, 0.91-1.12) or those in the United States (HR, 0.88; 95% CI, 0.73-1.05). Subgroup analyses revealed that the prognostic significance of H. pylori infection differed with respect to the year of study publication, number of patients, H. pylori detection method, tumor stage, H. pylori-positive rate, and risk of bias. The prognostic value of H. pylori infection on progression-free survival was unclear (HR, 0.84; 95% CI, 0.70-1.01). CONCLUSIONS These data provide limited, moderate-quality evidence that H. pylori infection is an indicator of good prognosis in European gastric cancer patients. However, this is not necessarily true for other populations.
Collapse
Affiliation(s)
- Guanghua Li
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Sun-Yat-sen University, No. 58, Zhongshan 2nd street, 510080 Guangzhou, Guangdong, PR China.
| | - Shuangjin Yu
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Sun-Yat-sen University, No. 58, Zhongshan 2nd street, 510080 Guangzhou, Guangdong, PR China
| | - Jianbo Xu
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Sun-Yat-sen University, No. 58, Zhongshan 2nd street, 510080 Guangzhou, Guangdong, PR China
| | - Xinhua Zhang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Sun-Yat-sen University, No. 58, Zhongshan 2nd street, 510080 Guangzhou, Guangdong, PR China
| | - Jinning Ye
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Sun-Yat-sen University, No. 58, Zhongshan 2nd street, 510080 Guangzhou, Guangdong, PR China.
| | - Zhao Wang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Sun-Yat-sen University, No. 58, Zhongshan 2nd street, 510080 Guangzhou, Guangdong, PR China.
| | - Yulong He
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Sun-Yat-sen University, No. 58, Zhongshan 2nd street, 510080 Guangzhou, Guangdong, PR China.
| |
Collapse
|
59
|
Mishra KK, Srivastava S, Aayyagari A, Ghosh K. Development of an animal model of Helicobacter pylori (Indian strain) infection. Indian J Gastroenterol 2019; 38:167-172. [PMID: 30911993 DOI: 10.1007/s12664-018-0905-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 09/23/2018] [Indexed: 02/04/2023]
Abstract
To develop an animal model for Indian strain Helicobacter pylori (H. pylori) infection. This model will allow one to study many facts of H. pylori infection in a more controlled manner. Mongolian gerbils were orogastric inoculated with two different Indian strains of H. pylori at different time points. Animals were sacrificed and looked for the presence of infection up to 52 weeks post-inoculation using a variety of techniques. Simultaneously, serums from these animals were also tested for antibody, and changes in the histopathology of stomach on H&E (hematoxylin and eosin) stains were also noted. Experimental sets of Mongolian gerbils were orally fed two strains of H. pylori obtained from human case by culture of different cagA and vacA strains three times daily on days 0, 2, and 4. H. pylori ATCC26695 strain was used for antisera preparation; three animals from each group were sacrificed at different time periods 2, 4, 8, 12, 26, 38 and 52 weeks after infection along with one control animal. Infections with H. pylori were confirmed in all the animals from 4 weeks onwards up to 52 weeks with histopathological changes in conformity with H. pylori gastritis. Wild Mongolian gerbils can be infected with Indian strains of H. pylori, and the infection persists at least 1 year. However, intensity of gastritis was milder than that seen in human case.
Collapse
Affiliation(s)
- Kanchan K Mishra
- Surat Raktadan Kendra and Research Centre, 1st Floor, Khatodara Health Centre, Near ChosathJoganiya Mata Mandir, Udhana Magdalla Road, Khatodara, Surat, 394 210, India
| | - Shashikant Srivastava
- Center for Infectious Diseases Research and Experimental Therapeutics, Dallas, TX, USA
| | - Archana Aayyagari
- Sanjay Gandhi Postgraduate Institute of Medical Sciences, Raebareli Road, Lucknow, 226 014, India
| | - Kanjaksha Ghosh
- Surat Raktadan Kendra and Research Centre, 1st Floor, Khatodara Health Centre, Near ChosathJoganiya Mata Mandir, Udhana Magdalla Road, Khatodara, Surat, 394 210, India.
| |
Collapse
|
60
|
Whisner CM, Athena Aktipis C. The Role of the Microbiome in Cancer Initiation and Progression: How Microbes and Cancer Cells Utilize Excess Energy and Promote One Another's Growth. Curr Nutr Rep 2019; 8:42-51. [PMID: 30758778 PMCID: PMC6426824 DOI: 10.1007/s13668-019-0257-2] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW We use an ecological lens to understand how microbes and cancer cells coevolve inside the ecosystems of our bodies. We describe how microbe-cancer cell interactions contribute to cancer progression, including cooperation between microbes and cancer cells. We discuss the role of the immune system in preventing this apparent 'collusion' and describe how microbe-cancer cell interactions lead to opportunities and challenges in treating cancer. RECENT FINDINGS Microbiota influence many aspects of our health including our cancer risk. Since both microbes and cancer cells rely on incoming resources for their survival and replication, excess energy and nutrient input from the host can play a role in cancer initiation and progression. Certain microbes enhance cancer cell fitness by promoting proliferation and protecting cancer cells from the immune system. How diet influences these interactions remains largely unknown but recent evidence suggests a role for nutrients across the cancer continuum.
Collapse
Affiliation(s)
- Corrie M Whisner
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA
| | - C Athena Aktipis
- Department of Psychology, Center for Social Dynamics and Complexity, Center for Evolution and Medicine, Biodesign Institute, Arizona State University, PO Box 871104, Tempe, AZ, 85287-1104, USA.
| |
Collapse
|
61
|
Chen J, Zhu C, Wang C, Zhang X, Ni J, Czajkowsky DM, Liu B, Guo Y. Discovery and genetic characterization of intestinal metaplasia in the Helicobacter felis-infected mouse model of gastric cancer. Acta Biochim Biophys Sin (Shanghai) 2019; 51:219-222. [PMID: 30576406 DOI: 10.1093/abbs/gmy160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 11/20/2018] [Indexed: 11/12/2022] Open
Affiliation(s)
- Jiangrong Chen
- Bio-ID Center, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Chunchao Zhu
- Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chaojie Wang
- Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaodan Zhang
- Bio-ID Center, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Jian Ni
- Bio-ID Center, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Daniel M Czajkowsky
- Bio-ID Center, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Bingya Liu
- Shanghai Key Laboratory of Gastric Neoplasms, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Guo
- Bio-ID Center, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
- State Key Laboratory for Oncogenes & Related Genes, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
62
|
Chan KL, Machuca MA, Rahman MM, Khan MF, Andrews D, Roujeinikova A. Purification, crystallization and preliminary X-ray crystallographic studies on the C-terminal domain of the flagellar protein FliL from Helicobacter pylori. Biosci Trends 2019; 12:630-635. [PMID: 30674764 DOI: 10.5582/bst.2018.01218] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
FliL is an inner membrane protein, occupying a position between the rotor and the stator of the bacterial flagellar motor. Its proximity to, and interactions with, the MS (membrane and supramembranous) ring, the switch complex and the stator proteins MotA/B suggests a role in recruitment and/or stabilization of the stator around the rotor, although the precise role of FliL in the flagellum remains to be established. In this study, recombinant C-terminal domain of Helicobacter pylori FliL (amino-acid residues 81-183) has been expressed in Escherichia coli and purified to > 98% homogeneity. Purified recombinant protein behaved as a monomer in solution. Crystals were obtained by the hanging-drop vapour-diffusion method using ammonium phosphate monobasic as a precipitant. These crystals belong to space group P1, with unit-cell parameters a = 62.5, b = 82.6, c = 97.8 Å, α = 67.7, ꞵ = 83.4, γ = 72.8°. A complete data set has been collected to 2.8 Å resolution using synchrotron radiation. This is an important step towards elucidation of the function of FliL in the bacterial flagellar motor.
Collapse
Affiliation(s)
- Kar Lok Chan
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University
| | - Mayra A Machuca
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University
| | - Mohammad Mizanur Rahman
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University
| | - Mohammad Firoz Khan
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University
| | - Daniel Andrews
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University
| | - Anna Roujeinikova
- Infection and Immunity Program, Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University.,Department of Biochemistry and Molecular Biology, Monash University
| |
Collapse
|
63
|
Zamperone A, Cohen D, Stein M, Viard C, Müsch A. Inhibition of polarity-regulating kinase PAR1b contributes to Helicobacter pylori inflicted DNA Double Strand Breaks in gastric cells. Cell Cycle 2019; 18:299-311. [PMID: 30580666 DOI: 10.1080/15384101.2018.1560121] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The serine/threonine kinase Par1 is a core component of the machinery that sets up polarity in the embryo and regulates cell fate decisions but its role in the homeostasis of adult tissues is poorly understood. Inhibition of Par1 by the bacterium Helicobacter pylori (H. pylori) represents the only established pathology that affects Par1 function in an adult epithelium. Thus, during chronic H. pylori infection of the gastric mucosa Par1 is one of the targets of the non-obligate H.pylori cytotoxic protein and oncogene CagA, which stimulates inflammation and triggers morphological changes, both believed to contribute to the gastric cancer risk imposed by H. pylori infection. Based on Par1's role in cell polarity, it has been speculated that Par1 inhibition affects epithelial polarity. Here we report the unexpected finding that CagA-mediated Par1-inhibition promotes the generation of DNA Double Strand Breaks in primary gastric epithelial cells, which likely contributes to the reported accumulation of mutations in chronically infected mucosal cells. Abbreviations: AGS: human gastric adenocarcinoma cell line; CM: CagA Multimerization (and Par1 binding) domain; H. pylori: Helicobacter pylori; DSB: Double Strand Break; HGECs: human (primary) gastric epithelial cells; IB: immunoblot; IF: immunofluorescence; MOI: Multiplicity of Infection; ROS: reactive oxygen species; Par1: Partitioning Defective 1 kinase; WT: wild type.
Collapse
Affiliation(s)
- Andrea Zamperone
- a Department of Developmental & Molecular Biology , Albert Einstein College Medicine , Bronx , NY , USA
| | - David Cohen
- a Department of Developmental & Molecular Biology , Albert Einstein College Medicine , Bronx , NY , USA
| | - Markus Stein
- b Department of Health Sciences , Albany College of Pharmacy and Health Sciences , Albany , NY , USA
| | - Charlotte Viard
- a Department of Developmental & Molecular Biology , Albert Einstein College Medicine , Bronx , NY , USA
| | - Anne Müsch
- a Department of Developmental & Molecular Biology , Albert Einstein College Medicine , Bronx , NY , USA
| |
Collapse
|
64
|
Shi YY, Zhang J, Zhang T, Zhou M, Wang Y, Zhang HJ, Ding SG. Cellular stress and redox activity proteins are involved in gastric carcinogenesis associated with Helicobacter pylori infection expressing high levels of thioredoxin-1. J Zhejiang Univ Sci B 2018; 19:750-763. [PMID: 30269443 PMCID: PMC6194355 DOI: 10.1631/jzus.b1700456] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 02/07/2018] [Indexed: 12/12/2022]
Abstract
Helicobacter pylori infection is related to the development of gastric diseases. Our previous studies showed that high thioredoxin-1 (Trx1) expression in H. pylori can promote gastric carcinogenesis. To explore the underlying molecular mechanisms, we performed an isobaric tags for relative and absolute quantitation (iTRAQ)-based quantitative proteomic analysis of stomach tissues from Mongolian gerbil infected with H. pylori expressing high and low Trx1. Differences in the profiles of the expressed proteins were analyzed by bioinformatics and verified using Western blot analysis. We found three candidate proteins, 14-3-3α/β, glutathione-S-transferase (GST), and heat shock protein 70 (HSP70), in high Trx1 tissues compared with low Trx1 tissues and concluded that cellular stress and redox activity-related proteins were involved in the pathogenesis of gastric cancer associated with H. pylori Trx1.
Collapse
Affiliation(s)
- Yan-yan Shi
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing 100191, China
| | - Jing Zhang
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China
| | - Ting Zhang
- Department of Microbiology, Peking University Health Science Center, Beijing 100191, China
| | - Man Zhou
- Department of Biomedical Engineering, College of Engineering, Peking University, Beijing 100871, China
| | - Ye Wang
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China
| | - He-jun Zhang
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China
| | - Shi-gang Ding
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
65
|
Mills JC, Samuelson LC. Past Questions and Current Understanding About Gastric Cancer. Gastroenterology 2018; 155:939-944. [PMID: 29964037 PMCID: PMC6174109 DOI: 10.1053/j.gastro.2018.06.044] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 06/18/2018] [Accepted: 06/18/2018] [Indexed: 02/06/2023]
Affiliation(s)
- Jason C Mills
- Departments of Medicine, Developmental Biology, Pathology & Immunology, Washington University School of Medicine, St. Louis, Missouri.
| | - Linda C Samuelson
- Departments of Molecular & Integrative Physiology and Internal Medicine, the University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
66
|
Lin Y, Liu Z, Qiu Y, Zhang J, Wu H, Liang R, Chen G, Qin G, Li Y, Zou D. Clinical significance of plasma D-dimer and fibrinogen in digestive cancer: A systematic review and meta-analysis. Eur J Surg Oncol 2018; 44:1494-1503. [DOI: 10.1016/j.ejso.2018.07.052] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 07/23/2018] [Indexed: 12/20/2022] Open
|
67
|
Yu S, Yang M, Lim KM, Cho Y, Kim H, Lee K, Jeong SH, Coffey RJ, Goldenring JR, Nam KT. Expression of LRIG1, a Negative Regulator of EGFR, Is Dynamically Altered during Different Stages of Gastric Carcinogenesis. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:2912-2923. [PMID: 30248341 DOI: 10.1016/j.ajpath.2018.08.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 08/07/2018] [Accepted: 08/14/2018] [Indexed: 12/12/2022]
Abstract
Leucine-rich repeats and immunoglobulin-like domains (LRIG)-1 is a transmembrane protein that antagonizes epidermal growth factor receptor signaling in epithelial tissues. LRIG1 is down-regulated in various epithelial cancers, including bladder, breast, and colorectal cancer, suggesting that it functions as a tumor suppressor. However, its role in gastric carcinogenesis is not well understood. Here, we investigated the changes in LRIG1 expression during the stages of gastric cancer. We used a DMP-777-induced spasmolytic polypeptide-expressing metaplasia mouse model and a tissue array of human gastric cancer lesions. The effects of LRIG1 knockdown were also assessed using the human gastric cancer cell line SNU638 in a xenograft model. LRIG1 expression varied over the course of gastric carcinogenesis, increasing in spasmolytic polypeptide-expressing metaplasia lesions but disappearing in intestinal metaplasia and cancer lesions, and the increase was concurrent with the up-regulation of epidermal growth factor receptor. In addition, LRIG1 knockdown promoted the tumorigenic potential in vitro, which was manifested as increased proliferation, invasiveness, and migration as well as increased tumor size in vivo in the xenograft model. Furthermore, LRIG1 expression was determined to be a positive prognostic biomarker for the survival of gastric cancer patients. Collectively, our findings indicate that LRIG1 expression is closely related wto gastric carcinogenesis and may play a vital role as a tumor suppressor through the modulation of epidermal growth factor receptor activity.
Collapse
Affiliation(s)
- Sungsook Yu
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Mijeong Yang
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Kyung-Min Lim
- College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| | - Yejin Cho
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyunji Kim
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Keunwook Lee
- Department of Biomedical Science, Hallym University, Chuncheon, Republic of Korea
| | - Sang-Ho Jeong
- Department of Surgery, Gyeongsang National University Changwon Hospital, Gyeongsang National University, Changwon, Republic of Korea
| | - Robert J Coffey
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee; Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - James R Goldenring
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, Tennessee; Section of Surgical Science, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Ki Taek Nam
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
68
|
Choi JM, Kim SG, Choi J, Park JY, Oh S, Yang HJ, Lim JH, Im JP, Kim JS, Jung HC. Effects of Helicobacter pylori eradication for metachronous gastric cancer prevention: a randomized controlled trial. Gastrointest Endosc 2018; 88:475-485.e2. [PMID: 29800546 DOI: 10.1016/j.gie.2018.05.009] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 05/12/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Whether eradication of Helicobacter pylori reduces the incidence of metachronous gastric cancer (MGC) is still debatable. We aimed to evaluate the long-term effect of H pylori eradication on the development of MGC after endoscopic gastric tumor resection. METHODS We undertook an open-label, prospective, randomized controlled trial at a tertiary hospital in Seoul, Korea. Participants were recruited during April 2005 to February 2011 and followed until December 2016. We assigned 898 patients with H pylori infection treated with endoscopic resection (ER) for gastric dysplasia or early gastric cancer to receive (n =442) or not receive (n =456) eradication therapy using a random-number chart. Eradication group patients received oral omeprazole 20 mg, amoxicillin 1 g, and clarithromycin 500 mg twice daily for a week, whereas control group patients received no H pylori treatment. The primary outcome was the incidence of MGC (intention-to-treat analysis). RESULTS The 877 patients who attended ≥1 follow-up examination (eradication group, 437; control group, 440) were analyzed. Median follow-up was 71.6 months (interquartile range, 42.1-90.0). MGC developed in 18 (4.1%) eradication and 36 (8.2%) control group patients (log-rank test, P = .01). In our yearly analysis, the effect of eradication showed a significant difference in 5 years after allocation (log-rank test, P = .02). The adjusted hazard ratio for the control group was 2.02 (95% CI, 1.14-3.56; P = .02), compared with the eradication group. CONCLUSIONS H pylori eradication significantly reduces the incidence of MGC after ER of gastric tumors and should be considered for H pylori-positive gastric tumor patients treated with ER. (Clinical trial registration number: NCT01510730.).
Collapse
Affiliation(s)
- Ji Min Choi
- Department of Internal Medicine, Healthcare Research Institute, Seoul National University Hospital Healthcare System Gangnam Center, Seoul, Korea
| | - Sang Gyun Kim
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Jeongmin Choi
- Department of Internal Medicine, Sanggye Paik Hospital, Inje University College of Medicine, Seoul, Korea
| | - Jae Yong Park
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Sooyeon Oh
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hyo-Joon Yang
- Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Joo Hyun Lim
- Department of Internal Medicine, Healthcare Research Institute, Seoul National University Hospital Healthcare System Gangnam Center, Seoul, Korea
| | - Jong Pil Im
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Joo Sung Kim
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Hyun Chae Jung
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
69
|
Zu LD, Peng XC, Zeng Z, Wang JL, Meng LL, Shen WW, Hu CT, Yang Y, Fu GH. Gastrin inhibits gastric cancer progression through activating the ERK-P65-miR23a/27a/24 axis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:115. [PMID: 29866191 PMCID: PMC5987590 DOI: 10.1186/s13046-018-0782-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 05/01/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND To test the hypothesis that activated extracellular signal-regulated kinase (ERK) regulates P65-miR23a/27a/24 axis in gastric cancer (GC) and the ERK-P65-miR23a/27a/24 axis plays an important role in the development of GC, and to evaluate the role of gastrin in GC progression and ERK-P65-miR23a/27a/24 axis. METHODS The component levels of the ERK-P65-miR23a/27a/24 axis in four fresh GC tissues, 101 paraffin-embedded GC tissues and four GC cell lines were determined by Western blotting, immunohistochemistry (IHC) or qRT-PCR. The effects of gastrin on GC were first evaluated by measuring gastrin serum levels in 30 healthy and 70 GC patients and performing a correlation analysis between gastrin levels and survival time in 27 GC patients after eight years of follow-up, then evaluated on GC cell lines, GC cell xenograft models, and patient-derived xenografts (PDX) mouse models. The roles of ERK-P65-miR23a/27a/24 axis in GC progression and in the effects of gastrin on GC were examined. RESULTS ERK- P65-miR23a/27a/24 axis was proved to be present in GC cells. The levels of components of ERK-P65-miR23a/27a/24 axis were decreased in GC tissue samples and PGC cells. The decreased levels of components of ERK-P65-miR23a/27a/24 axis were associated with poor prognosis of GC, and ERK-P65-miR23a/27a/24 axis played a suppressive role in GC progression. Low blood gastrin was correlated with poor prognosis of the GC patients and decreased expression of p-ERK and p-P65 in GC tissues. Gastrin inhibited proliferation of poorly-differentiated GC (PGC) cells through activating the ERK-P65-miR23a/27a/24 axis. Gastrin inhibited GC growth and enhanced the suppression of GC by cisplatin in mice or PGC cell culture models through activating the ERK-P65-miR23a/27a/24 axis or its components. CONCLUSIONS ERK-P65-miR23a/27a/24 axis is down-regulated, leading to excess GC growth and poor prognosis of GC. Low gastrin promoted excess GC growth and contributed to the poor prognosis of the GC patients by down-regulating ERK-P65-miR23a/27a/24 axis. Gastrin inhibits gastric cancer growth through activating the ERK-P65-miR23a/27a/24 axis.
Collapse
Affiliation(s)
- Li-Dong Zu
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Institutes of Medical Sciences, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xing-Chun Peng
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Institutes of Medical Sciences, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhi Zeng
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jing-Long Wang
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Institutes of Medical Sciences, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li-Li Meng
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Institutes of Medical Sciences, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei-Wei Shen
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Institutes of Medical Sciences, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chun-Ting Hu
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Institutes of Medical Sciences, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ye Yang
- Department of Digestive Medicine, Ningbo No. 2 Hospital, Ningbo, 315010, China
| | - Guo-Hui Fu
- Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Institutes of Medical Sciences, Shanghai Key Laboratory of Gastric Neoplasms, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China. .,Pathology Center, Shanghai General Hospital/Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, No. 280, South Chong-Qing Road, Shanghai, 200025, People's Republic of China.
| |
Collapse
|
70
|
Beckett AC, Loh JT, Chopra A, Leary S, Lin AS, McDonnell WJ, Dixon BREA, Noto JM, Israel DA, Peek RM, Mallal S, Algood HMS, Cover TL. Helicobacter pylori genetic diversification in the Mongolian gerbil model. PeerJ 2018; 6:e4803. [PMID: 29796347 PMCID: PMC5961626 DOI: 10.7717/peerj.4803] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 04/30/2018] [Indexed: 12/12/2022] Open
Abstract
Helicobacter pylori requires genetic agility to infect new hosts and establish long-term colonization of changing gastric environments. In this study, we analyzed H. pylori genetic adaptation in the Mongolian gerbil model. This model is of particular interest because H. pylori-infected gerbils develop a high level of gastric inflammation and often develop gastric adenocarcinoma or gastric ulceration. We analyzed the whole genome sequences of H. pylori strains cultured from experimentally infected gerbils, in comparison to the genome sequence of the input strain. The mean annualized single nucleotide polymorphism (SNP) rate per site was 1.5e−5, which is similar to the rates detected previously in H. pylori-infected humans. Many of the mutations occurred within or upstream of genes associated with iron-related functions (fur, tonB1, fecA2, fecA3, and frpB3) or encoding outer membrane proteins (alpA, oipA, fecA2, fecA3, frpB3 and cagY). Most of the SNPs within coding regions (86%) were non-synonymous mutations. Several deletion or insertion mutations led to disruption of open reading frames, suggesting that the corresponding gene products are not required or are deleterious during chronic H. pylori colonization of the gerbil stomach. Five variants (three SNPs and two deletions) were detected in isolates from multiple animals, which suggests that these mutations conferred a selective advantage. One of the mutations (FurR88H) detected in isolates from multiple animals was previously shown to confer increased resistance to oxidative stress, and we now show that this SNP also confers a survival advantage when H. pylori is co-cultured with neutrophils. Collectively, these analyses allow the identification of mutations that are positively selected during H. pylori colonization of the gerbil model.
Collapse
Affiliation(s)
- Amber C Beckett
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - John T Loh
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Abha Chopra
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Australia
| | - Shay Leary
- Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Australia
| | - Aung Soe Lin
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Wyatt J McDonnell
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Beverly R E A Dixon
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Jennifer M Noto
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Dawn A Israel
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Richard M Peek
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, United States of America.,Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States of America
| | - Simon Mallal
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, United States of America.,Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States of America.,Institute for Immunology and Infectious Diseases, Murdoch University, Murdoch, Australia
| | - Holly M Scott Algood
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States of America.,Tennessee Valley Healthcare System, Veterans Affairs, Nashville, TN, United States of America
| | - Timothy L Cover
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN, United States of America.,Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, United States of America.,Tennessee Valley Healthcare System, Veterans Affairs, Nashville, TN, United States of America
| |
Collapse
|
71
|
Park JY, Forman D, Waskito LA, Yamaoka Y, Crabtree JE. Epidemiology of Helicobacter pylori and CagA-Positive Infections and Global Variations in Gastric Cancer. Toxins (Basel) 2018; 10:E163. [PMID: 29671784 PMCID: PMC5923329 DOI: 10.3390/toxins10040163] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 04/10/2018] [Accepted: 04/10/2018] [Indexed: 12/11/2022] Open
Abstract
Gastric cancer is a major health burden and is the fifth most common malignancy and the third most common cause of death from cancer worldwide. Development of gastric cancer involves several aspects, including host genetics, environmental factors, and Helicobacter pylori infection. There is increasing evidence from epidemiological studies of the association of H. pylori infection and specific virulence factors with gastric cancer. Studies in animal models indicate H. pylori is a primary factor in the development of gastric cancer. One major virulence factor in H. pylori is the cytotoxin-associated gene A (cagA), which encodes the CagA protein in the cag pathogenicity island (cag PAI). Meta-analysis of studies investigating CagA seropositivity irrespective of H. pylori status identified that CagA seropositivity increases the risk of gastric cancer (OR = 2.87, 95% CI: 1.95⁻4.22) relative to the risk of H. pylori infection alone (OR = 2.31, 95% CI: 1.58⁻3.39). Eradicating H. pylori is a strategy for reducing gastric cancer incidence. A meta-analysis of six randomised controlled trials (RCTs) suggests that searching for and eradicating H. pylori infection reduces the subsequent incidence of gastric cancer with a pooled relative risk of 0.66 (95% CI: 0.46⁻0.95). The introduction in regions of high gastric cancer incidence of population-based H. pylori screening and treatment programmes, with a scientifically valid assessment of programme processes, feasibility, effectiveness and possible adverse consequences, would impact the incidence of H. pylori-induced gastric cancer. Given the recent molecular understanding of the oncogenic role of CagA, targeting H. pylori screening and treatment programmes in populations with a high prevalence of H. pylori CagA-positive strains, particularly the more oncogenic East Asian H. pylori CagA strains, may be worth further investigation to optimise the benefits of such strategies.
Collapse
Affiliation(s)
- Jin Young Park
- International Agency for Research on Cancer, 69372 Lyon, France.
| | - David Forman
- International Agency for Research on Cancer, 69372 Lyon, France.
| | - Langgeng Agung Waskito
- Institute of Tropical Disease, Universitas Airlangga, Surabaya 60113, Indonesia.
- Department of Environmental and Preventive Medicine, Faculty of Medicine, Oita University, Yufu, Oita 879-5503, Japan.
| | - Yoshio Yamaoka
- Department of Environmental and Preventive Medicine, Faculty of Medicine, Oita University, Yufu, Oita 879-5503, Japan.
- Department of Medicine-Gastroenterology, Michael E. DeBakey Veterans Affairs Medical Center and Baylor College of Medicine, Houston, TX 77030, USA.
| | - Jean E Crabtree
- Leeds Institute Biomedical and Clinical Sciences, Wellcome Trust Brenner Building, St. James's University Hospital, University of Leeds, Leeds LS9 7TF, UK.
| |
Collapse
|
72
|
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: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
73
|
Horiguchi N, Tahara T, Yamada H, Yoshida D, Okubo M, Nagasaka M, Nakagawa Y, Shibata T, Tsukamoto T, Kuroda M, Ohmiya N. In vivo diagnosis of early-stage gastric cancer found after Helicobacter pylori eradication using probe-based confocal laser endomicroscopy. Dig Endosc 2018; 30:219-227. [PMID: 28731617 DOI: 10.1111/den.12926] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2017] [Accepted: 07/18/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIM Early-stage gastric cancer (EGC) found after Helicobacter pylori (Hp) eradication often displays non-tumorous regenerative epithelium and/or maturated tumorous epithelium overlying the cancerous tissue, which may confuse endoscopic and histological diagnosis. Probe-based confocal laser endomicroscopy (pCLE) enables in vivo real-time optical biopsy. We compared the diagnostic yields for these EGC cases using conventional white light endoscopy (WL), magnifying endoscopy with narrow-band imaging (ME-NBI), pCLE, and endoscopic biopsy; we also compared the accuracy of the horizontal extent diagnosis between ME-NBI and pCLE. METHODS This study enrolled 30 patients with 36 EGC lesions after successful Hp eradication. Diagnostic yields of WL, ME-NBI, pCLE, and endoscopic biopsy were prospectively compared. Four points of cancerous margins (oral, anal, anterior, and posterior sites) were also prospectively evaluated with M-NBI and pCLE to determine the horizontal extent of the EGC. RESULTS Diagnostic yield was significantly higher with pCLE than with WL and endoscopic biopsy (97 vs 72%, 97 vs 72%, P = 0.0159, 0.0077, respectively), whereas it did not differ from ME-NBI (88.9%, P = 0.371). Height of non-tumorous regenerative epithelium or maturated atypical glands was 104.7 ± 34.2 μm in the pCLE-positive cases, whereas it was 188.3 ± 27.1 μm in a pCLE-negative case (P = 0.0004). Diagnostic accuracy of the horizontal margin of EGC was significantly higher with pCLE than with ME-NBI (92 vs 70%, P = 0.0159). CONCLUSION pCLE may be helpful for the diagnosis of ambiguous ECG found after Hp eradication because it enables real-time scanning throughout the lesion and detection of subsurface microstructure.
Collapse
Affiliation(s)
- Noriyuki Horiguchi
- Department of Gastroenterology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Tomomitsu Tahara
- Department of Gastroenterology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Hyuga Yamada
- Department of Gastroenterology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Dai Yoshida
- Department of Gastroenterology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Masaaki Okubo
- Department of Gastroenterology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Mitsuo Nagasaka
- Department of Gastroenterology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Yoshihito Nakagawa
- Department of Gastroenterology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Tomoyuki Shibata
- Department of Gastroenterology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Tetsuya Tsukamoto
- Department of Diagnostic Pathology I, Fujita Health University School of Medicine, Toyoake, Japan
| | - Makoto Kuroda
- Department of Diagnostic Pathology I, Fujita Health University School of Medicine, Toyoake, Japan
| | - Naoki Ohmiya
- Department of Gastroenterology, Fujita Health University School of Medicine, Toyoake, Japan
| |
Collapse
|
74
|
Togo R, Ishihara K, Mabe K, Oizumi H, Ogawa T, Kato M, Sakamoto N, Nakajima S, Asaka M, Haseyama M. Preliminary study of automatic gastric cancer risk classification from photofluorography. World J Gastrointest Oncol 2018; 10:62-70. [PMID: 29467917 PMCID: PMC5807881 DOI: 10.4251/wjgo.v10.i2.62] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 12/05/2017] [Accepted: 12/13/2017] [Indexed: 02/05/2023] Open
Abstract
AIM To perform automatic gastric cancer risk classification using photofluorography for realizing effective mass screening as a preliminary study. METHODS We used data for 2100 subjects including X-ray images, pepsinogen I and II levels, PGI/PGII ratio, Helicobacter pylori (H. pylori) antibody, H. pylori eradication history and interview sheets. We performed two-stage classification with our system. In the first stage, H. pylori infection status classification was performed, and H. pylori-infected subjects were automatically detected. In the second stage, we performed atrophic level classification to validate the effectiveness of our system. RESULTS Sensitivity, specificity and Youden index (YI) of H. pylori infection status classification were 0.884, 0.895 and 0.779, respectively, in the first stage. In the second stage, sensitivity, specificity and YI of atrophic level classification for H. pylori-infected subjects were 0.777, 0.824 and 0.601, respectively. CONCLUSION Although further improvements of the system are needed, experimental results indicated the effectiveness of machine learning techniques for estimation of gastric cancer risk.
Collapse
Affiliation(s)
- Ren Togo
- Graduate School of Information Science and Technology, Hokkaido University, Hokkaido 060-0814, Japan
| | - Kenta Ishihara
- Graduate School of Information Science and Technology, Hokkaido University, Hokkaido 060-0814, Japan
| | - Katsuhiro Mabe
- Department of Gastroenterology, National Hospital Organization Hakodate Hospital, Hokkaido 041-8512, Japan
| | - Harufumi Oizumi
- Medical Examination Center of the Yamagata City Medical Association, Yamagata 990-2473, Japan
| | - Takahiro Ogawa
- Graduate School of Information Science and Technology, Hokkaido University, Hokkaido 060-0814, Japan
| | - Mototsugu Kato
- Department of Gastroenterology, National Hospital Organization Hakodate Hospital, Hokkaido 041-8512, Japan
| | - Naoya Sakamoto
- Department of Gastroenterology, Hokkaido University Graduate School of Medicine, Hokkaido 060-8648, Japan
| | - Shigemi Nakajima
- Department of General Medicine, Japan Community Healthcare Organization Shiga Hospital, Shiga 520-0846, Japan
| | - Masahiro Asaka
- Health Sciences University of Hokkaido, Hokkaido 061-0293, Japan
| | - Miki Haseyama
- Graduate School of Information Science and Technology, Hokkaido University, Hokkaido 060-0814, Japan
| |
Collapse
|
75
|
Simultaneous detection of human CYP2C19 polymorphisms and antibiotic resistance of Helicobacter pylori using a personalised diagnosis kit. J Glob Antimicrob Resist 2018; 13:174-179. [PMID: 29444465 DOI: 10.1016/j.jgar.2017.12.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 12/22/2017] [Accepted: 12/30/2017] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVES A personalised diagnosis kit for Helicobacter pylori that employs visual gene chip technology for the simultaneous detection of CYP2C19 polymorphisms and clarithromycin/levofloxacin antibiotic resistance was evaluated. METHODS Gastric antrum mucosa biopsy specimens of 394 patients were tested using the kit. DNA sequencing and antibiotic susceptibility testing of the H. pylori were also performed. RESULTS In total, 267 (67.8%) of the 394 specimens were positive for H. pylori using the kit and DNA sequencing, and 136 (34.5%) were positive by culturing. For human CYP2C19 and the bacterial 23S rRNA and gyrA genes, the concordance rates were 92.4% (364/394), 96.6% (258/267) and 97.0% (259/267) between the kit and DNA sequencing results, respectively. For clarithromycin and levofloxacin resistance, the concordance rates were 90.4% (123/136) and 81.6% (111/136) between the kit and antibiotic susceptibility testing results. CONCLUSIONS The personalised diagnosis kit for H. pylori provides useful information for the choice of proton pump inhibitor and antibiotic in combination therapy.
Collapse
|
76
|
Lee Y, Hung S, Wang H, Lin C, Wang H, Chang M, Sung J, Chiou Y, Lin S. Is there different risk of cancer among end-stage renal disease patients undergoing hemodialysis and peritoneal dialysis? Cancer Med 2018; 7:485-498. [PMID: 29356425 PMCID: PMC5806101 DOI: 10.1002/cam4.1289] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Revised: 11/20/2017] [Accepted: 11/21/2017] [Indexed: 01/02/2023] Open
Abstract
Cancer is a global issue in recent decade. Despite this alarming increase in the incidence of cancer, to date, whether the risk of developing cancer differs among peritoneal dialysis (PD) and hemodialysis (HD) patients is still uncertain. In this retrospective cohort study, data were obtained from the National Health Insurance Research Database of Taiwan, which provides coverage to almost 99% of the nation's population. After matching, a total of 4491 (or 3369) incident PD patients and 8982 (or 6738) incident HD patients between 2000 and 2009 were enrolled from the database. In addition, 22,455 (or 16,845) nondialysis patients were selected as a control group. The patients were monitored for the occurrence of cancer until 2010, and their data were analyzed using several different models. In general, the results showed that the risks of hepatocellular, kidney, bladder, extra kidney/bladder urinary tract, and thyroid cancers were higher in dialysis patients. We also compared the risk of cancer between two dialysis groups by using the HD patients as the reference group. The result showed that there is no significant different for each cancer risk between two dialysis groups. In conclusion, dialysis patients had a higher risk of certain types of cancer than those in the nonuremia group. However, there was no significant difference in the cancer risk between the two dialysis groups when compared directly.
Collapse
Affiliation(s)
- Yi‐Che Lee
- Division of NephrologyDepartment of Internal MedicineE‐DA Dachang Hospital/ I‐Shou UniversityKaohsiungTaiwan
| | - Shih‐Yuan Hung
- Division of NephrologyDepartment of Internal MedicineE‐DA Dachang Hospital/ I‐Shou UniversityKaohsiungTaiwan
- School of Medicine for International StudentsE‐DA Hospital/ I‐Shou UniversityKaohsiungTaiwan
| | - Hao‐Kuang Wang
- School of Medicine for International StudentsE‐DA Hospital/ I‐Shou UniversityKaohsiungTaiwan
- Department of NeurosurgeryE‐DA Hospital/ I‐Shou UniversityKaohsiungTaiwan
| | - Chi‐Wei Lin
- School of Medicine for International StudentsE‐DA Hospital/ I‐Shou UniversityKaohsiungTaiwan
- Department of Medical EducationE‐DA Hospital/ I‐Shou UniversityKaohsiungTaiwan
| | - Hsi‐Hao Wang
- Division of NephrologyDepartment of Internal MedicineE‐DA Dachang Hospital/ I‐Shou UniversityKaohsiungTaiwan
| | - Min‐Yu Chang
- Division of NephrologyDepartment of Internal MedicineE‐DA Dachang Hospital/ I‐Shou UniversityKaohsiungTaiwan
| | - Junne‐Ming Sung
- Division of NephrologyDepartment of Internal MedicineNational Cheng Kung University HospitalTainanTaiwan
| | - Yuan‐Yow Chiou
- Department of PediatricsNational Cheng Kung University HospitalTainanTaiwan
- Institute of Clinical MedicineCollege of MedicineNational Cheng Kung UniversityTainanTaiwan
| | - Sheng‐Hsiang Lin
- Institute of Clinical MedicineCollege of MedicineNational Cheng Kung UniversityTainanTaiwan
- Biostatistics Consulting CenterNational Cheng Kung University HospitalTainanTaiwan
- Department of Public HealthCollege of MedicineNational Cheng‐Kung UniversityTainanTaiwan
| |
Collapse
|
77
|
Sepulveda AR, J. Del Portillo A. Molecular Basis of Diseases of the Gastrointestinal Tract. MOLECULAR PATHOLOGY 2018:387-415. [DOI: 10.1016/b978-0-12-802761-5.00019-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
78
|
Ma F, Li CC, Zhang CY. Development of quantum dot-based biosensors: principles and applications. J Mater Chem B 2018; 6:6173-6190. [DOI: 10.1039/c8tb01869c] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
We review the recent advances in quantum dot-based biosensors and focus on quantum dot-based fluorescent, bioluminescent, chemiluminescent, and photoelectrochemical biosensors.
Collapse
Affiliation(s)
- Fei Ma
- College of Chemistry
- Chemical Engineering and Materials Science
- Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong
- Key Laboratory of Molecular and Nano Probes
- Ministry of Education
| | - Chen-chen Li
- College of Chemistry
- Chemical Engineering and Materials Science
- Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong
- Key Laboratory of Molecular and Nano Probes
- Ministry of Education
| | - Chun-yang Zhang
- College of Chemistry
- Chemical Engineering and Materials Science
- Collaborative Innovation Center of Functionalized Probes for Chemical Imaging in Universities of Shandong
- Key Laboratory of Molecular and Nano Probes
- Ministry of Education
| |
Collapse
|
79
|
Noto JM, Romero-Gallo J, Piazuelo MB, Peek RM. The Mongolian Gerbil: A Robust Model of Helicobacter pylori-Induced Gastric Inflammation and Cancer. Methods Mol Biol 2017; 1422:263-80. [PMID: 27246040 DOI: 10.1007/978-1-4939-3603-8_24] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The Mongolian gerbil is an efficient, robust, and cost-effective rodent model that recapitulates many features of H. pylori-induced gastric inflammation and carcinogenesis in humans, allowing for targeted investigation of the bacterial determinants and environmental factors and, to a lesser degree, host constituents that govern H. pylori-mediated disease. This chapter discusses means through which the Mongolian gerbil model has been used to define mechanisms of H. pylori-inflammation and cancer as well as the current materials and methods for utilizing this model of microbially induced disease.
Collapse
Affiliation(s)
- Jennifer M Noto
- Division of Gastroenterology, Department of Medicine, Vanderbilt University Medical Center, 2215 Garland Avenue MRB IV 1030C, Nashville, TN, 37232-0252, USA
| | - Judith Romero-Gallo
- Division of Gastroenterology, Department of Medicine, Vanderbilt University Medical Center, 2215 Garland Avenue MRB IV 1030C, Nashville, TN, 37232-0252, USA
| | - M Blanca Piazuelo
- Division of Gastroenterology, Department of Medicine, Vanderbilt University Medical Center, 2215 Garland Avenue MRB IV 1030C, Nashville, TN, 37232-0252, USA
| | - Richard M Peek
- Division of Gastroenterology, Department of Medicine, Vanderbilt University Medical Center, 2215 Garland Avenue MRB IV 1030C, Nashville, TN, 37232-0252, USA.
| |
Collapse
|
80
|
Lan KH, Lee WP, Wang YS, Liao SX, Lan KH. Helicobacter pylori CagA protein activates Akt and attenuates chemotherapeutics-induced apoptosis in gastric cancer cells. Oncotarget 2017; 8:113460-113471. [PMID: 29371922 PMCID: PMC5768339 DOI: 10.18632/oncotarget.23050] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Accepted: 11/13/2017] [Indexed: 12/13/2022] Open
Abstract
Infection with cagA-positive Helicobacter pylori is associated with a higher risk of gastric cancer. The cagA gene product, CagA, is translocated into gastric epithelial cells and perturbs host cellular biological functions. Etoposide, a topoisomerase II inhibitor widely used to couple DNA damage to apoptosis, is a common cytotoxic agent used for advanced gastric cancer. We investigate the effect of CagA on etoposide-induced apoptosis in gastric cancer cells to elucidate whether CagA play a role in gastric carcinogenesis via impairing DNA damage-dependent apoptosis. AGS cell lines stably expressing CagA isolated from H. pylori 26695 strain were established. In the presence of etoposide, viability of parental AGS cells was decreased in a time-and dose-dependent manner, whereas CagA-expressing AGS cells were less susceptible to etoposide induced cell-killing effect. Suppression of etoposide-induced apoptosis was shown in CagA-expressing but not in parental AGS cells by DNA fragmentation, cell cycle, and annexin-V assays. This inhibitory effect of etoposide-induced apoptosis conferred by CagA was also demonstrated in SCM1 and MKN45 gastric cancer cell lines, with two additional chemotherapeutics, 5-FU and cisplatin. The effect of Akt activation on inhibition of etoposide-induced cytotoxicity by CagA was also evaluated. CagA expression and etoposide administration activate Akt in a dose-dependent manner. Enhancement of etoposide cytotoxicity by a PI-3-kinase inhibitor, LY294002, was evident in parental but was attenuated in CagA-expressing AGS cells. CagA may activate Akt, either in the absence or presence of etoposide, potentially contributing to gastric carcinogenesis associated with H. pylori infection and therapeutic resistance by impairing DNA damage-dependent apoptosis.
Collapse
Affiliation(s)
- Keng-Hsueh Lan
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital National Taiwan University Cancer Center, Taipei, Taiwan
| | - Wei-Ping Lee
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan.,Department and Institute of Biochemistry, National Yang-Ming University, Taipei, Taiwan
| | - Yu-Shan Wang
- Institute of Molecular Medicine and Bioengineering, National Chiao Tung University, Hsinchu, Taiwan
| | - Shi-Xian Liao
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Keng-Hsin Lan
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Department and Institute of Pharmacology, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
81
|
Evaluation of the visibility of early gastric cancer using linked color imaging and blue laser imaging. BMC Gastroenterol 2017; 17:150. [PMID: 29216843 PMCID: PMC5721593 DOI: 10.1186/s12876-017-0707-5] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 11/24/2017] [Indexed: 01/06/2023] Open
Abstract
Background Blue laser imaging (BLI) and linked color imaging (LCI) are the color enhancement features of the LASEREO endoscopic system, which provide a narrow band light observation function and expansion and reduction of the color information, respectively. Methods We examined 82 patients with early gastric cancer (EGC) diagnosed between April 2014 and August 2015. Five expert and 5 non-expert endoscopists retrospectively compared images obtained on non-magnifying BLI bright mode (BLI-BRT) and LCI with those obtained via conventional white light imaging (WLI). Interobserver agreement was also assessed. Results In experts’ evaluation of the images, an improvement in visibility was observed in 73% (60/82) and 20% (16/82) of cases under LCI and BLI-BRT, respectively. In non-experts’ evaluation of the images, an improvement in visibility was observed in 76.8% (63/82) and 24.3% (20/82) of cases under LCI and BLI-BRT, respectively. There were no significant differences between experts and non-experts in the evaluation of the images. The improvement in visibility was significantly higher with LCI than with BLI-BRT in experts and non-experts (p < 0.01). With regard to tumor color on WLI, the improvement in the visibility of reddish and whitish tumors was significantly higher than that of isochromatic tumors when LCI was used. The improvement in visibility with LCI was observed in 71% (12/17) and 74% (48/65) of patients with and without Helicobacter pylori (Hp) eradication, respectively; no significant difference in improvement was observed between these groups. The interobserver agreement was good to satisfactory at ≥ 0.62. Conclusions In conclusion, our study showed that LCI improved the visibility of EGC, regardless of the level of endoscopists’ experience or Hp eradication in patients, particularly for EGCs with a reddish or whitish color. The improvement in visibility was significantly higher with LCI than that with BLI.
Collapse
|
82
|
Burkitt MD, Duckworth CA, Williams JM, Pritchard DM. Helicobacter pylori-induced gastric pathology: insights from in vivo and ex vivo models. Dis Model Mech 2017; 10:89-104. [PMID: 28151409 PMCID: PMC5312008 DOI: 10.1242/dmm.027649] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Gastric colonization with Helicobacter pylori induces diverse human pathological conditions, including superficial gastritis, peptic ulcer disease, mucosa-associated lymphoid tissue (MALT) lymphoma, and gastric adenocarcinoma and its precursors. The treatment of these conditions often relies on the eradication of H. pylori, an intervention that is increasingly difficult to achieve and that does not prevent disease progression in some contexts. There is, therefore, a pressing need to develop new experimental models of H. pylori-associated gastric pathology to support novel drug development in this field. Here, we review the current status of in vivo and ex vivo models of gastric H. pylori colonization, and of Helicobacter-induced gastric pathology, focusing on models of gastric pathology induced by H. pylori, Helicobacter felis and Helicobacter suis in rodents and large animals. We also discuss the more recent development of gastric organoid cultures from murine and human gastric tissue, as well as from human pluripotent stem cells, and the outcomes of H. pylori infection in these systems.
Collapse
Affiliation(s)
- Michael D Burkitt
- Gastroenterology Research Unit, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool L69 3GE, UK
| | - Carrie A Duckworth
- Gastroenterology Research Unit, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool L69 3GE, UK
| | - Jonathan M Williams
- Pathology and Pathogen Biology, Royal Veterinary College, North Mymms AL9 7TA, UK
| | - D Mark Pritchard
- Gastroenterology Research Unit, Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool L69 3GE, UK
| |
Collapse
|
83
|
|
84
|
Magnifying NBI Patterns of Gastric Mucosa After Helicobacter pylori Eradication and Its Potential Link to the Gastric Cancer Risk. Dig Dis Sci 2017; 62:2421-2427. [PMID: 28702753 DOI: 10.1007/s10620-017-4676-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Accepted: 07/06/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND Gastric cancer develops after successful H. pylori eradication in patients with severe atrophic gastritis. We classified atrophic and non-atrophic mucosa of gastric body using magnifying NBI endoscopy in patients after successful H. pylori eradication. MATERIALS AND METHODS One hundred and twenty-five patients after successful H. pylori eradication (median period after eradication: 36 months) were enrolled. Magnifying NBI patterns in the uninvolved gastric body were divided into the following: restored-small, round pits, accompanied with honeycomb-like subepithelial capillary networks; atrophic-well-demarcated oval or tubulovillous pits with clearly visible coiled or wavy vessels. The subjects were also classified into the three types: Grade 0-restored pattern is shown in all or almost the entire area of gastric body; Grade 1-mixture of restored and atrophic pattern, there is a considerable portion of the atrophic area in the lesser curvature; Grade 2-atrophic pattern is shown in all or almost the entire area of the gastric body. RESULTS Sensitivity and specificity for atrophic type for detection of histological intestinal metaplasia were 95.9 and 98.3%, respectively. No association was observed between the prevalence of Grades 0, 1 and 2 and duration after eradication, while grades 1 and 2 were significantly frequent in gastric cancer patients diagnosed both before (27/35: 77%) and after (23/31: 74%) eradication, compared to the cancer-free subjects (15/59: 25%) (P < 0.001). The grades 1 and 2 were also common in patients who underwent H. pylori eradication for gastric ulcer. CONCLUSIONS Magnifying the NBI pattern well correlates with pathological status of gastric mucosa after H. pylori eradication and may predict gastric cancer occurrence.
Collapse
|
85
|
Insulin-like growth factor binding protein-1 (IGFBP-1) upregulated by Helicobacter pylori and is associated with gastric cancer cells migration. Pathol Res Pract 2017; 213:1029-1036. [PMID: 28864349 DOI: 10.1016/j.prp.2017.08.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 07/26/2017] [Accepted: 08/20/2017] [Indexed: 12/24/2022]
Abstract
Insulin-like growth factor binding protein-1 (IGFBP-1), a secreted protein, implicated of various cells in mediating the proliferation, migration, invasion, adhesion, survival and so on. In this study, we assessed the expression and release of IGFBP-1 from gastric cancer cells with H. pylori 26695 infection and the biological functions of IGFBP-1 in gastric cancer cells. The results showed that the expression and release of IGFBP-1 were increased in gastric cancer cells (MGC-803, BGC-823, SGC-7901) infected with H. pylori 26695. In addition, the upregulation of IGFBP-1 was dose-dependent in BGC-823 cells infected with H. pylori 26695 but not time-dependent. The upregulation of IGFBP-1 got to peak at 12h after H. pylori 26695 infection and then decreased over time. Subsequently, we measured its functions by silencing and overexpressing IGFBP1 which suggested that overexpression of IGFBP-1 could inhibit the migration of BGC-823 and SGC-7901 cells. However, knocking down the IGFBP-1 could increase the migration of BGC-823 and SGC-7901 cells. Functional findings illustrated that IGFBP-1 was implicated in H. pylori 26695-induced MMP-9 expression in BGC-823 cells. In addition, overexpressing IGFBP1 reduce the promoting effect of MMP-9 on the BGC-823 cells migration. In summary, we demonstrated that IGFBP-1 suppress the migration of BGC-823 cells and play a protective role in the process of H. pylori-induced gastric cancer.
Collapse
|
86
|
Prevention of Gastric Cancer: Eradication of Helicobacter Pylori and Beyond. Int J Mol Sci 2017; 18:ijms18081699. [PMID: 28771198 PMCID: PMC5578089 DOI: 10.3390/ijms18081699] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 07/31/2017] [Accepted: 07/31/2017] [Indexed: 12/15/2022] Open
Abstract
Although its prevalence is declining, gastric cancer remains a significant public health issue. The bacterium Helicobacter pylori is known to colonize the human stomach and induce chronic atrophic gastritis, intestinal metaplasia, and gastric cancer. Results using a Mongolian gerbil model revealed that H. pylori infection increased the incidence of carcinogen-induced adenocarcinoma, whereas curative treatment of H. pylori significantly lowered cancer incidence. Furthermore, some epidemiological studies have shown that eradication of H. pylori reduces the development of metachronous cancer in humans. However, other reports have warned that human cases of atrophic metaplastic gastritis are already at risk for gastric cancer development, even after eradication of these bacteria. In this article, we discuss the effectiveness of H. pylori eradication and the morphological changes that occur in gastric dysplasia/cancer lesions. We further assess the control of gastric cancer using various chemopreventive agents.
Collapse
|
87
|
MIFTAHUSSURUR MUHAMMAD, YAMAOKA YOSHIO, GRAHAM DAVIDY. Helicobacter pylori as an oncogenic pathogen, revisited. Expert Rev Mol Med 2017; 19:e4. [PMID: 28322182 PMCID: PMC6905048 DOI: 10.1017/erm.2017.4] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Gastric cancer is an inflammation-associated malignancy aetiologically related to infection with the bacterium, Helicobacter pylori, which is considered a necessary but insufficient cause. Unless treated, H. pylori causes life-long acute and chronic gastric inflammation resulting in progressive gastric mucosal damage that may result in gastric cancer. The rate of progression from superficial gastritis, to an atrophic metaplastic mucosa, and ultimately to cancer relates to the virulence of the infecting H. pylori as well as host and environmental factors. H. pylori virulence is a reflection of its propensity to cause severe gastric inflammation. Both mucosal inflammation and H. pylori can cause host genomic instability, including dysregulation of DNA mismatch repair, stimulation of expression of activation-induced cytidine deaminase, abnormal DNA methylation and dysregulation of micro RNAs, which may result in an accumulation of mutations and loss of normal regulation of cell growth. The difference in cancer risk between the most and least virulent H. pylori strain is only approximately 2-fold. Overall, none of the putative virulence factors identified to date have proved to be disease-specific. The presence, severity, extent and duration of inflammation appear to be the most important factors and current evidence suggests that any host, environmental or bacterial factor that reliably enhances the inflammatory response to the H. pylori infection increases the risk of gastric cancer.
Collapse
Affiliation(s)
- MUHAMMAD MIFTAHUSSURUR
- Department of Medicine, Gastroenterology and Hepatology Section, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Environmental and Preventive Medicine, Oita University Faculty of Medicine, Yufu 879-5593, Japan
- Gastroentero-Hepatology Division, Department of Internal Medicine, Faculty of Medicine – Dr Soetomo Teaching Hospital – Institute of Tropical Disease, Universitas Airlangga, Surabaya 60115, Indonesia
| | - YOSHIO YAMAOKA
- Department of Medicine, Gastroenterology and Hepatology Section, Baylor College of Medicine, Houston, Texas 77030, USA
- Department of Environmental and Preventive Medicine, Oita University Faculty of Medicine, Yufu 879-5593, Japan
| | - DAVID Y. GRAHAM
- Department of Medicine, Gastroenterology and Hepatology Section, Baylor College of Medicine, Houston, Texas 77030, USA
| |
Collapse
|
88
|
Suarez G, Romero-Gallo J, Sierra JC, Piazuelo MB, Krishna US, Gomez MA, Wilson KT, Peek RM. Genetic Manipulation of Helicobacter pylori Virulence Function by Host Carcinogenic Phenotypes. Cancer Res 2017; 77:2401-2412. [PMID: 28209611 DOI: 10.1158/0008-5472.can-16-2922] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 11/23/2016] [Accepted: 01/31/2017] [Indexed: 02/06/2023]
Abstract
Helicobacter pylori is the strongest risk factor for gastric adenocarcinoma, yet only a minority of infected persons ever develop this malignancy. One cancer-linked locus is the cag type 4 secretion system (cagT4SS), which translocates an oncoprotein into host cells. A structural component of the cagT4SS is CagY, which becomes rapidly altered during in vivo adaptation in mice and rhesus monkeys, rendering the cagT4SS nonfunctional; however, these models rarely develop gastric cancer. We previously demonstrated that the H. pylori cag+ strain 7.13 rapidly induces gastric cancer in Mongolian gerbils. We now use this model, in conjunction with samples from patients with premalignant lesions, to define the effects of a carcinogenic host environment on the virulence phenotype of H. pylori to understand how only a subset of infected individuals develop cancer. H. pylori cagY sequence differences and cagT4SS function were directly related to the severity of inflammation in human gastric mucosa in either a synchronous or metachronous manner. Serial infections of Mongolian gerbils with H. pylori strain 7.13 identified an oscillating pattern of cagT4SS function. The development of dysplasia or cancer selected for attenuated virulence phenotypes, but robust cagT4SS function could be restored upon infection of new hosts. Changes in the genetic composition of cagY mirrored cagT4SS function, although the mechanisms of cagY alterations differed in human isolates (mutations) versus gerbil isolates (addition/deletion of motifs). These results indicate that host carcinogenic phenotypes modify cagT4SS function via altering cagY, allowing the bacteria to persist and induce carcinogenic consequences in the gastric niche. Cancer Res; 77(9); 2401-12. ©2017 AACR.
Collapse
Affiliation(s)
- Giovanni Suarez
- Departments of Cancer Biology, Pathology, Microbiology, and Immunology, and Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Judith Romero-Gallo
- Departments of Cancer Biology, Pathology, Microbiology, and Immunology, and Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Johanna C Sierra
- Departments of Cancer Biology, Pathology, Microbiology, and Immunology, and Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - M Blanca Piazuelo
- Departments of Cancer Biology, Pathology, Microbiology, and Immunology, and Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Uma S Krishna
- Departments of Cancer Biology, Pathology, Microbiology, and Immunology, and Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Martin A Gomez
- Department of Medicine, National University of Colombia, Bogota, Colombia.,Hospital El Tunal Unit of Gastroenterology, Bogota, Colombia
| | - Keith T Wilson
- Departments of Cancer Biology, Pathology, Microbiology, and Immunology, and Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Richard M Peek
- Departments of Cancer Biology, Pathology, Microbiology, and Immunology, and Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.
| |
Collapse
|
89
|
Helicobacter pylori, Cancer, and the Gastric Microbiota. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 908:393-408. [PMID: 27573782 DOI: 10.1007/978-3-319-41388-4_19] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Gastric adenocarcinoma is one of the leading causes of cancer-related death worldwide and Helicobacter pylori infection is the strongest known risk factor for this disease. Although the stomach was once thought to be a sterile environment, it is now known to house many bacterial species leading to a complex interplay between H. pylori and other residents of the gastric microbiota. In addition to the role of H. pylori virulence factors, host genetic polymorphisms, and diet, it is now becoming clear that components of the gastrointestinal microbiota may also influence H. pylori-induced pathogenesis. In this chapter, we discuss emerging data regarding the gastric microbiota in humans and animal models and alterations that occur to the composition of the gastric microbiota in the presence of H. pylori infection that may augment the risk of developing gastric cancer.
Collapse
|
90
|
Gagnaire A, Nadel B, Raoult D, Neefjes J, Gorvel JP. Collateral damage: insights into bacterial mechanisms that predispose host cells to cancer. Nat Rev Microbiol 2017; 15:109-128. [DOI: 10.1038/nrmicro.2016.171] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
91
|
Yanaka A. Role of Sulforaphane in Protection of Gastrointestinal Tract Against H. pylori and NSAID-Induced Oxidative Stress. Curr Pharm Des 2017; 23:4066-4075. [PMID: 28176666 PMCID: PMC5759180 DOI: 10.2174/1381612823666170207103943] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 01/26/2017] [Accepted: 02/02/2017] [Indexed: 12/15/2022]
Abstract
BACKGROUND Sulforaphane (SFN), a phytochemical found in abundance in broccoli sprouts, potently induces a variety of antioxidant enzymes, and thereby protects cells from injury induced by various kinds of oxidative stresses. It has been suggested that both H. pylori infection and intake of non-steroidal anti-inflammatory drugs (NSAIDs) induce chronic oxidative stress in gastrointestinal (GI) mucosa, thereby causing mucosal injury in the GI tract. Therefore, it would be a reasonable assumption that SFN protects GI mucosa against oxidative injury induced by H. pylori or NSAIDs. METHODS We examined the effects of SFN on H. pylori viability in vitro, levels of gastritis in H.pylori-infected mice in vivo, and in H.pylori-infected human subjects. We also examined the effects of SFN on NSAID-induced small intestinal injury in mice. RESULTS Our data from the H. pylori infection study clearly demonstrated that SFN inhibited H. pylori viability both in vitro and in vivo, and mitigated H. pylori-induced gastritis in mice and humans. Similarly, our study on NSAID-induced small intestinal injury showed that SFN not only mitigated aspirin-induced injury of small intestinal epithelial cells in vitro, but also ameliorated indomethacin-induced small intestinal injury in mice in vivo. CONCLUSIONS These data strongly suggest that SFN contributes to the protection of GI mucosa against oxidative injury induced by H. pylori or NSAIDs.
Collapse
Affiliation(s)
- Akinori Yanaka
- Division of Gastroenterology, Hitachi Medical Education and Research Center, Faculty of Medicine, University of Tsukuba, 2-1-1, Jonan-cho, Hitachi-shi, Ibaraki-ken, 317-0077, Japan
| |
Collapse
|
92
|
Mégraud F, Musso D, Drancourt M, Lehours P. Curved and Spiral Bacilli. Infect Dis (Lond) 2017. [DOI: 10.1016/b978-0-7020-6285-8.00182-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
93
|
HATAKEYAMA M. Structure and function of Helicobacter pylori CagA, the first-identified bacterial protein involved in human cancer. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2017; 93:196-219. [PMID: 28413197 PMCID: PMC5489429 DOI: 10.2183/pjab.93.013] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/14/2023]
Abstract
Chronic infection with Helicobacter pylori cagA-positive strains is the strongest risk factor of gastric cancer. The cagA gene-encoded CagA protein is delivered into gastric epithelial cells via bacterial type IV secretion, where it undergoes tyrosine phosphorylation at the Glu-Pro-Ile-Tyr-Ala (EPIYA) motifs. Delivered CagA then acts as a non-physiological scaffold/hub protein by interacting with multiple host signaling molecules, most notably the pro-oncogenic phosphatase SHP2 and the polarity-regulating kinase PAR1/MARK, in both tyrosine phosphorylation-dependent and -independent manners. CagA-mediated manipulation of intracellular signaling promotes neoplastic transformation of gastric epithelial cells. Transgenic expression of CagA in experimental animals has confirmed the oncogenic potential of the bacterial protein. Structural polymorphism of CagA influences its scaffold function, which may underlie the geographic difference in the incidence of gastric cancer. Since CagA is no longer required for the maintenance of established gastric cancer cells, studying the role of CagA during neoplastic transformation will provide an excellent opportunity to understand molecular processes underlying "Hit-and-Run" carcinogenesis.
Collapse
Affiliation(s)
- Masanori HATAKEYAMA
- Department of Microbiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Correspondence should be addressed: M. Hatakeyama, Division of Microbiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan (e-mail: )
| |
Collapse
|
94
|
Devi S, Ansari SA, Tenguria S, Kumar N, Ahmed N. Multipronged regulatory functions of a novel endonuclease (TieA) from Helicobacter pylori. Nucleic Acids Res 2016; 44:9393-9412. [PMID: 27550181 PMCID: PMC5100599 DOI: 10.1093/nar/gkw730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 08/11/2016] [Indexed: 12/29/2022] Open
Abstract
Helicobacter pylori portrays a classical paradigm of persistent bacterial infections. A well balanced homeostasis of bacterial effector functions and host responses is purported to be the key in achieving long term colonization in specific hosts. H. pylori nucleases have been shown to assist in natural transformation, but their role in virulence and colonization remains elusive. Therefore, it is imperative to understand the involvement of these nucleases in the pathogenesis of H. pylori. Here, we report the multifaceted role of a TNFR-1 interacting endonuclease A (TieA) from H. pylori. tieA expression is differentially regulated in response to environmental stress and post adherence to gastric epithelial cells. Studies with isogenic knockouts of tieA revealed it to be a secretory protein which translocates into the host gastric epithelial cells independent of a type IV secretion system, gets phosphorylated by DNA-PK kinase and auto-phosphorylates as serine kinase. Furthermore, TieA binds to and cleaves DNA in a non-specific manner and promotes Fas mediated apoptosis in AGS cells. Additionally, TieA induced pro-inflammatory cytokine secretion via activation of transcription factor AP-1 and signaled through MAP kinase pathway. Collectively, TieA with its multipronged and moonlighting functions could facilitate H. pylori in maintaining a balance of bacterial adaptation, and elimination by the host responses.
Collapse
Affiliation(s)
- Savita Devi
- Pathogen Biology Laboratory, Department of Biotechnology and Bioinformatics, University of Hyderabad, Gachibowli, Hyderabad 500046, India
| | - Suhail A Ansari
- Pathogen Biology Laboratory, Department of Biotechnology and Bioinformatics, University of Hyderabad, Gachibowli, Hyderabad 500046, India
| | - Shivendra Tenguria
- Pathogen Biology Laboratory, Department of Biotechnology and Bioinformatics, University of Hyderabad, Gachibowli, Hyderabad 500046, India
| | - Naveen Kumar
- Pathogen Biology Laboratory, Department of Biotechnology and Bioinformatics, University of Hyderabad, Gachibowli, Hyderabad 500046, India
| | - Niyaz Ahmed
- Pathogen Biology Laboratory, Department of Biotechnology and Bioinformatics, University of Hyderabad, Gachibowli, Hyderabad 500046, India
| |
Collapse
|
95
|
Evaluation of nitric oxide production and proliferation activity of recombinant Bacterioferritin of Helicobacter pylori on macrophages. Microb Pathog 2016; 100:149-153. [DOI: 10.1016/j.micpath.2016.07.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Revised: 07/13/2016] [Accepted: 07/13/2016] [Indexed: 02/07/2023]
|
96
|
Tahara T, Shibata T, Horiguchi N, Kawamura T, Okubo M, Ishizuka T, Nagasaka M, Nakagawa Y, Ohmiya N. A Possible Link between Gastric Mucosal Atrophy and Gastric Cancer after Helicobacter pylori Eradication. PLoS One 2016; 11:e0163700. [PMID: 27706195 PMCID: PMC5051933 DOI: 10.1371/journal.pone.0163700] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 09/13/2016] [Indexed: 02/07/2023] Open
Abstract
Background The effect of H. pylori eradication in gastric cancer prevention can be attributed to the improvement of atrophic gastritis, which is a known risk of gastric cancer. However, gastric cancer has also been diagnosed after long-term H. pylori eradication. This study aimed to clarify the association between gastric atrophy and gastric cancer after H. pylori eradication, including its clinicopathological features. Methods A total of 55 consecutive patients with 64 early gastric cancers (EGCs) diagnosed after H. pylori eradication were enrolled. The degree of endoscopic atrophy and the histological degrees of mononuclear cell infiltration, atrophy, and metaplasia in the corpus and adjacent mucosa of the EGCs were determined and scored. Results The majority of EGCs (63/64) were located within the endoscopically assessed atrophic mucosa or along the atrophic border. The adjacent mucosa of the EGCs presented significantly higher degrees of all histological parameters than in the corpus (mononuclear cell infiltration, 0.86+/-0.09 vs. 0.51+/-0.11, P = 0.016; atrophy, 1.77+/-0.13 vs. 0.65+/-0.14, P<0.0001; metaplasia, 1.68+/-0.13 vs. 0.48+/-0.1, P<0.0001). The degree of endoscopic atrophy improved in the patients with longer post-H. pylori eradication periods; however, this trend was not observed for the histological parameters, and high degrees of atrophy and metaplasia were observed in the adjacent mucosa of the EGCs compared with the corpus during all periods (all P<0.05). The histological degrees of atrophy and metaplasia in the adjacent mucosa were particularly higher in the patients who underwent eradication due to gastric ulcers. Conclusions Severe gastric atrophy remained in the adjacent mucosa of the EGCs after H. pylori eradication, which may be linked to gastric carcinogenesis.
Collapse
Affiliation(s)
- Tomomitsu Tahara
- Department of Gastroenterology, Fujita Health University School of Medicine, Toyoake, Japan
- * E-mail:
| | - Tomoyuki Shibata
- Department of Gastroenterology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Noriyuki Horiguchi
- Department of Gastroenterology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Tomohiko Kawamura
- Department of Gastroenterology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Masaaki Okubo
- Department of Gastroenterology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Takamitsu Ishizuka
- Department of Gastroenterology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Mitsuo Nagasaka
- Department of Gastroenterology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Yoshihito Nakagawa
- Department of Gastroenterology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Naoki Ohmiya
- Department of Gastroenterology, Fujita Health University School of Medicine, Toyoake, Japan
| |
Collapse
|
97
|
Sue S, Shibata W, Kameta E, Sato T, Ishii Y, Kaneko H, Miwa H, Sasaki T, Tamura T, Kondo M, Maeda S. Intestine-specific homeobox (ISX) induces intestinal metaplasia and cell proliferation to contribute to gastric carcinogenesis. J Gastroenterol 2016; 51:949-60. [PMID: 26872890 DOI: 10.1007/s00535-016-1176-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Accepted: 01/26/2016] [Indexed: 02/04/2023]
Abstract
BACKGROUND Helicobacter pylori induces chronic inflammation and intestinal metaplasia (IM) through genetic and epigenetic changes and activation of intracellular signaling pathways that contribute to gastric carcinogenesis. However, the precise mechanism of IM in gastric carcinogenesis has not been fully elucidated. We previously found that intestine-specific homeobox (ISX) mRNA expression increased in organoids cultured from Helicobacter-infected mouse mucosa. In this study, we elucidate the role of ISX in the development of IM and gastric carcinogenesis. METHODS ISX expression was assessed in Helicobacter-infected mouse and human gastric mucosa. MKN45 gastric cancer cells were co-cultured with H. pylori to determine whether Helicobacter infection induced ISX expression. We established stable MKN45 transfected cells expressing ISX (Stable-ISX MKN45) and performed a spheroid colony formation assay and a xenograft model. We performed ISX immunohistochemistry in cancer and adjacent gastric tissues. RESULTS ISX expression was increased in mouse and human gastric mucosa infected with Helicobacter. The presence of IM and H. pylori infection in human stomach was correlated with ISX expression. H. pylori induced ISX mRNA and protein expression. CDX1/2, cyclinD1, and MUC2 were upregulated in Stable-ISX MKN45, whereas MUC5AC was downregulated. Stable-ISX MKN45 cells formed more spheroid colonies, and had high tumorigenic ability. ISX expression in gastric cancer and adjacent mucosa were correlated. CONCLUSIONS ISX expression induced by H. pylori infection may lead to IM and hyperproliferation of gastric mucosa through CDX1/2 and cyclinD1 expression, contributing to gastric carcinogenesis.
Collapse
Affiliation(s)
- Soichiro Sue
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Wataru Shibata
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan.,Advanced Medical Research Center, Yokohama City University, Yokohama, Japan
| | - Eri Kameta
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Takeshi Sato
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Yasuaki Ishii
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Hiroaki Kaneko
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Haruo Miwa
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Tomohiko Sasaki
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Toshihide Tamura
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Masaaki Kondo
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Shin Maeda
- Department of Gastroenterology, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan.
| |
Collapse
|
98
|
Lind J, Backert S, Hoffmann R, Eichler J, Yamaoka Y, Perez-Perez GI, Torres J, Sticht H, Tegtmeyer N. Systematic analysis of phosphotyrosine antibodies recognizing single phosphorylated EPIYA-motifs in CagA of East Asian-type Helicobacter pylori strains. BMC Microbiol 2016; 16:201. [PMID: 27590005 PMCID: PMC5009636 DOI: 10.1186/s12866-016-0820-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 05/19/2016] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Highly virulent strains of the gastric pathogen Helicobacter pylori encode a type IV secretion system (T4SS) that delivers the effector protein CagA into gastric epithelial cells. Translocated CagA undergoes tyrosine phosphorylation by members of the oncogenic c-Src and c-Abl host kinases at EPIYA-sequence motifs A, B and D in East Asian-type strains. These phosphorylated EPIYA-motifs serve as recognition sites for various SH2-domains containing human proteins, mediating interactions of CagA with host signaling factors to manipulate signal transduction pathways. Recognition of phospho-CagA is mainly based on the use of commercial pan-phosphotyrosine antibodies that were originally designed to detect phosphotyrosines in mammalian proteins. Specific anti-phospho-EPIYA antibodies for each of the three sites in CagA are not forthcoming. RESULTS This study was designed to systematically analyze the detection preferences of each phosphorylated East Asian CagA EPIYA-motif by pan-phosphotyrosine antibodies and to determine a minimal recognition sequence. We synthesized phospho- and non-phosphopeptides derived from each predominant EPIYA-site, and determined the recognition patterns by seven different pan-phosphotyrosine antibodies using Western blotting, and also investigated representative East Asian H. pylori isolates during infection. The results indicate that a total of only 9-11 amino acids containing the phosphorylated East Asian EPIYA-types are required and sufficient to detect the phosphopeptides with high specificity. However, the sequence recognition by the different antibodies was found to bear high variability. From the seven antibodies used, only four recognized all three phosphorylated EPIYA-motifs A, B and D similarly well. Two of the phosphotyrosine antibodies preferentially bound primarily to the phosphorylated motif A and D, while the seventh antibody failed to react with any of the phosphorylated EPIYA-motifs. Control experiments confirmed that none of the antibodies reacted with non-phospho-CagA peptides and in accordance were able to recognize phosphotyrosine proteins in human cells. CONCLUSIONS The results of this study disclose the various binding preferences of commercial anti-phosphotyrosine antibodies for phospho-EPIYA-motifs, and are valuable in the application for further characterization of CagA phosphorylation events during infection with H. pylori and risk prediction for gastric disease development.
Collapse
Affiliation(s)
- Judith Lind
- Department of Biology, Division of Microbiology, Friedrich Alexander University Erlangen-Nuremberg, Staudtstr. 5, D-91058, Erlangen, Germany
| | - Steffen Backert
- Department of Biology, Division of Microbiology, Friedrich Alexander University Erlangen-Nuremberg, Staudtstr. 5, D-91058, Erlangen, Germany
| | - Rebecca Hoffmann
- Department of Chemistry and Pharmacy, Friedrich Alexander University Erlangen-Nuremberg, Schuhstraße 19, D-91052, Erlangen, Germany
| | - Jutta Eichler
- Department of Chemistry and Pharmacy, Friedrich Alexander University Erlangen-Nuremberg, Schuhstraße 19, D-91052, Erlangen, Germany
| | - Yoshio Yamaoka
- Department of Environmental and Preventive Medicine, Oita University Faculty of Medicine, Yufu, Japan
| | - Guillermo I Perez-Perez
- Department of Medicine and Microbiology, New York University, Langone Medical Centre, New York, USA
| | - Javier Torres
- Unidad de Investigación en Enfermedades Infecciosas, Hospital de Pediatría del Instituto Mexicano del Seguro Social, Mexico City, México
| | - Heinrich Sticht
- Bioinformatics, Institute for Biochemistry, Friedrich Alexander University Erlangen-Nuremberg, Fahrstrasse 17, D-91054, Erlangen, Germany
| | - Nicole Tegtmeyer
- Department of Biology, Division of Microbiology, Friedrich Alexander University Erlangen-Nuremberg, Staudtstr. 5, D-91058, Erlangen, Germany.
| |
Collapse
|
99
|
Helicobacter pylori with high thioredoxin-1 expression promotes stomach carcinogenesis in Mongolian gerbils. Clin Res Hepatol Gastroenterol 2016; 40:480-6. [PMID: 26669590 DOI: 10.1016/j.clinre.2015.11.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Revised: 10/18/2015] [Accepted: 11/02/2015] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Previous studies by this group have shown that Helicobacter pylori with high thioredoxin-1 (Trx1) expression might be involved in stomach carcinogenesis in vitro. To study histopathological changes of the stomach mucosa in vivo, a Mongolian gerbil model infected with H. pylori with high Trx1 expression was established. METHODS Healthy, male Mongolian gerbils (n=75) were randomly divided into 3 groups: controls (n=15), which were not infected with H. pylori, high Trx1 (n=30) which were infected with H. pylori with high Trx1 expression and low Trx1 (n=30) which were infected with low Trx1 expression H. pylori. The animals were sacrificed at 4, 20, 34, 48, 70 and 90 weeks after inoculation. RESULTS The Mongolian gerbil model of H. pylori infection was successfully established. Three animals died during the study, leaving 72 animals (controls, n=14; low Trx1, n=29; high Trx1, n=29) examined on schedule. Histopathological analysis of the stomach mucosa showed gradually increased aggravation over time in the high and low Trx1 groups. Compared with control and low Trx1, the histopathological changes were more serious in the high Trx1 group. At 90 weeks, no abnormal changes were found in the controls, but 62.5% of the high Trx1 group and 33.3% of the low Trx1 showed adenocarcinomas. The H. pylori Trx1 level in gastric cancer tissue was significantly higher than that from gastritis tissue. Within gastric cancer cells, high Trx1 expression in H. pylori significantly upregulated cyclin D1. CONCLUSIONS High Trx1 expression in H. pylori promoted stomach carcinogenesis. More studies are needed to confirm this finding.
Collapse
|
100
|
Talebi Bezmin Abadi A. Helicobacter pylori and Gastric Cancer. Front Med (Lausanne) 2016; 3:36. [PMID: 27597945 PMCID: PMC4992849 DOI: 10.3389/fmed.2016.00036] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 08/05/2016] [Indexed: 12/13/2022] Open
Affiliation(s)
- Amin Talebi Bezmin Abadi
- Department of Bacteriology, Faculty of Medical Sciences, Tarbiat Modares University , Tehran , Iran
| |
Collapse
|