101
|
Abstract
The late 1800s Louis Pasteur and Robert Koch introduced and popularized the germ theory of disease. At that time, gastric cancer was the most common cause of cancer deaths in most countries making the stomach an early site of microbial research with a focus on gastric luminal and mucosal bacteria and the role of Boas-Oppler bacillus (Lactobacillus) in the diagnosis of gastric cancer. In the 1970s, the research focus evolved to studies of the gastric microbiome in the production of nitrosamines and included development of the Correa cascade. Interest in nitrosamine production peaked in the late 1980s and was replaced by studies of the newly described Helicobacter pylori and studies of its role in gastritis, gastric atrophy, and gastric cancer. The last decade has witnessed a rebirth in interest in the gastric microbiota as part of worldwide interest in the human microbiome. Although fungi were prominent in the studies of gastric microbiology in the nineteenth century, their potential role in disease pathogenesis has yet to be addressed using modern techniques. Overall, current studies of the gastric bacterial microbiome do not provide convincing evidence to expand the role of the gastric microbiome in cancer pathogenesis beyond what is directly attributable to the oncogenic potential of H. pylori and its role in persisting acute-on-chronic inflammation.
Collapse
|
102
|
Rajilic-Stojanovic M, Figueiredo C, Smet A, Hansen R, Kupcinskas J, Rokkas T, Andersen L, Machado JC, Ianiro G, Gasbarrini A, Leja M, Gisbert JP, Hold GL. Systematic review: gastric microbiota in health and disease. Aliment Pharmacol Ther 2020; 51:582-602. [PMID: 32056247 DOI: 10.1111/apt.15650] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 01/09/2020] [Accepted: 01/17/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Helicobacter pylori is the most infamous constituent of the gastric microbiota and its presence is the strongest risk factor for gastric cancer and other gastroduodenal diseases. Although historically the healthy stomach was considered a sterile organ, we now know it is colonised with a complex microbiota. However, its role in health and disease is not well understood. AIM To systematically explore the literature on the gastric microbiota in health and disease as well as the gut microbiota after bariatric surgery. METHODS A systematic search of online bibliographic databases MEDLINE/EMBASE was performed between 1966 and February 2019 with screening in accordance with Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. Randomised controlled trials, cohort studies and observational studies were included if they reported next-generation sequencing derived microbiota analysis on gastric aspirate/tissue or stool samples (bariatric surgical outcomes). RESULTS Sixty-five papers were eligible for inclusion. With the exception of H pylori-induced conditions, overarching gastric microbiota signatures of health or disease could not be determined. Gastric carcinogenesis induces a progressively altered microbiota with an enrichment of oral and intestinal taxa as well as significant changes in host gastric mucin expression. Proton pump inhibitors usage increases gastric microbiota richness. Bariatric surgery is associated with an increase in potentially pathogenic proteobacterial species in patient stool samples. CONCLUSION While H pylori remains the single most important risk factor for gastric disease, its capacity to shape the collective gastric microbiota remains to be fully elucidated. Further studies are needed to explore the intricate host/microbial and microbial/microbial interplay.
Collapse
|
103
|
Zhao R, Liu Z, Xu W, Song L, Ren H, Ou Y, Liu Y, Wang S. Helicobacter pylori infection leads to KLF4 inactivation in gastric cancer through a TET1-mediated DNA methylation mechanism. Cancer Med 2020; 9:2551-2563. [PMID: 32017451 PMCID: PMC7131848 DOI: 10.1002/cam4.2892] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 01/04/2020] [Accepted: 01/14/2020] [Indexed: 12/15/2022] Open
Abstract
Krüppel-like factor 4 (KLF4) has a tumor suppressor role in the progression of gastric cancer (GC), and inhibition or loss of KLF4 expression was identified in GC. The aim of this study was to explore the new molecular mechanism of KLF4 inactivation in gastric cancer. Herein, we report that Helicobacter pylori infection or Cag pathogenicity island protein A (CagA) gene transduction resulted in KLF4 expression downregulation and promoted gastric epithelial cell and gastric cancel cell proliferation, migration, and colony formation. Mechanistically, we found that CagA gene transduction led to DNA methylation of the KLF4 promoter, an effect that was relevant to the significant downregulation of TET1 expression. Causally, knockdown of TET1 expression decreased KLF4 expression, whereas overexpression of TET1 had the opposite effect. Clinically, we found that KLF4 expression and the 5-hmC levels were lower in GC cells with H pylori infection than in GC cells without H pylori infection. Thus, our study not only sheds new light on how H pylori infection promotes the progression of GC but also elucidates a novel mechanism of KLF4 inactivation in GC pathogenesis. During pathogenesis, an alteration in the H pylori/CagA-TET1-KLF4 signaling pathway plays a critical role, suggesting that this pathway may be a prospective target for gastric carcinoma intervention and therapy.
Collapse
Affiliation(s)
- Rongrong Zhao
- Department of Physiopathology, Anhui Medical University, Hefei, Anhui, China.,Department of Pathology, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Zhengxia Liu
- Department of Physiopathology, Anhui Medical University, Hefei, Anhui, China
| | - Wenting Xu
- Department of Physiopathology, Anhui Medical University, Hefei, Anhui, China
| | - Le Song
- Department of Physiopathology, Anhui Medical University, Hefei, Anhui, China
| | - Haifeng Ren
- Department of Physiopathology, Anhui Medical University, Hefei, Anhui, China
| | - Yang Ou
- Department of Physiopathology, Anhui Medical University, Hefei, Anhui, China
| | - Yakun Liu
- Department of Physiopathology, Anhui Medical University, Hefei, Anhui, China
| | - Siying Wang
- Department of Physiopathology, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
104
|
Brüggemann H, Al-Zeer MA. Bacterial signatures and their inflammatory potentials associated with prostate cancer. APMIS 2020; 128:80-91. [PMID: 31990107 DOI: 10.1111/apm.13021] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 11/25/2019] [Indexed: 02/06/2023]
Abstract
Chronic inflammation can create a microenvironment that can contribute to the formation of prostate pathologies. Far less well understood is the origin of inflammation in the prostate. One potential source is microbial infections of the prostate. This review summarizes recent findings regarding the presence of bacteria in the prostate and the dysbiosis of bacterial populations in the urinary tract and the gastrointestinal tract related to prostate cancer, thereby focusing on next-generation sequencing (NGS)-generated data. The current limitations regarding NGS-based detection methods and other difficulties in the quest for a microbial etiology for prostate cancer are discussed. We then focus on a few bacterial species, including Cutibacterium acnes and Escherichia coli that are often NGS-detected in prostatic tissue specimens, and discuss their possible contribution as initiator or enhancer of prostate inflammation and prostate carcinogenesis.
Collapse
Affiliation(s)
| | - Munir A Al-Zeer
- Institute of Biotechnology, Department of Applied Biochemistry, Technical University of Berlin, Berlin, Germany
| |
Collapse
|
105
|
Abstract
UNLABELLED Owing to its strong acid production, the stomach was known to be a bacteria-free organ for many years. On the other hand, the presence of Helicobacter pylori (H. pylori) and other acid-resistant microbiota that are to persist in the stomach challenged this. It is now recognized that the existence of H. pylori and non-H. pylori species have been linked to the improvement of gastric disease; despite this, there is little published data on the interaction of gastric bacterial flora and the resultant effect on gastric health. The stomach has a unique microbiota including five major phyla, such as Firmicutes, Proteobacteria, Actinobacteria, Fusobacteria and Bacteroidetes. These phyla are identified in both H. pylori-infected and uninfected persons. The resident gastric microflora may mediate the role of H. pylori in the gastric diseases. This article aims to review previous studies that examine the impact of H. pylori infection and the effect of resident gastric microbiota on gut health and disease conditions. HOW TO CITE THIS ARTICLE Ozbey G, Sproston E, Hanafiah A. Helicobacter pylori Infection and Gastric Microbiota. Euroasian J Hepato-Gastroenterol 2020;10(1):36-41.
Collapse
Affiliation(s)
- Gokben Ozbey
- Department of Medical Services and Techniques, Vocational School of Health Services, Firat University, Elazig, Turkey
| | - Emma Sproston
- Department of Biology and Biochemistry, School of Biological Sciences, University of Aberdeen, Aberdeen, United Kingdom
| | - Alfizah Hanafiah
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
106
|
Curty G, de Carvalho PS, Soares MA. The Role of the Cervicovaginal Microbiome on the Genesis and as a Biomarker of Premalignant Cervical Intraepithelial Neoplasia and Invasive Cervical Cancer. Int J Mol Sci 2019; 21:ijms21010222. [PMID: 31905652 PMCID: PMC6981542 DOI: 10.3390/ijms21010222] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 12/24/2019] [Accepted: 12/25/2019] [Indexed: 12/24/2022] Open
Abstract
The microbiome is able to modulate immune responses, alter the physiology of the human organism, and increase the risk of viral infections and development of diseases such as cancer. In this review, we address changes in the cervical microbiota as potential biomarkers to identify the risk of cervical intraepithelial neoplasia (CIN) development and invasive cervical cancer in the context of human papillomavirus (HPV) infection. Current approaches for clinical diagnostics and the manipulation of microbiota with the use of probiotics and through microbiota transplantation are also discussed.
Collapse
|
107
|
Min J, Vega PN, Engevik AC, Williams JA, Yang Q, Patterson LM, Simmons AJ, Bliton RJ, Betts JW, Lau KS, Magness ST, Goldenring JR, Choi E. Heterogeneity and dynamics of active Kras-induced dysplastic lineages from mouse corpus stomach. Nat Commun 2019; 10:5549. [PMID: 31804471 PMCID: PMC6895174 DOI: 10.1038/s41467-019-13479-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 10/31/2019] [Indexed: 02/07/2023] Open
Abstract
Dysplasia is considered a key transition state between pre-cancer and cancer in gastric carcinogenesis. However, the cellular or phenotypic heterogeneity and mechanisms of dysplasia progression have not been elucidated. We have established metaplastic and dysplastic organoid lines, derived from Mist1-Kras(G12D) mouse stomach corpus and studied distinct cellular behaviors and characteristics of metaplastic and dysplastic organoids. We also examined functional roles for Kras activation in dysplasia progression using Selumetinib, a MEK inhibitor, which is a downstream mediator of Kras signaling. Here, we report that dysplastic organoids die or show altered cellular behaviors and diminished aggressive behavior in response to MEK inhibition. However, the organoids surviving after MEK inhibition maintain cellular heterogeneity. Two dysplastic stem cell (DSC) populations are also identified in dysplastic cells, which exhibited different clonogenic potentials. Therefore, Kras activation controls cellular dynamics and progression to dysplasia, and DSCs might contribute to cellular heterogeneity in dysplastic cell lineages.
Collapse
Affiliation(s)
- Jimin Min
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Paige N Vega
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Amy C Engevik
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Janice A Williams
- Cell Imaging Share Resource, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Qing Yang
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
- Institute of Pathogen Biology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Loraine M Patterson
- Center for GI Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Alan J Simmons
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - R Jarrett Bliton
- UNC Departments of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Joshua W Betts
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Ken S Lau
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Scott T Magness
- Center for GI Biology and Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- UNC Departments of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
- University of North Carolina Chapel Hill/ North Carolina State University joint Departments of Biomedical Engineering, Chapel Hill, NC, 27599, USA
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - James R Goldenring
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
- Nashville VA Medical Center, Nashville, TN, 37232, USA
| | - Eunyoung Choi
- Department of Surgery, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA.
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA.
| |
Collapse
|
108
|
Vergara D, Simeone P, Damato M, Maffia M, Lanuti P, Trerotola M. The Cancer Microbiota: EMT and Inflammation as Shared Molecular Mechanisms Associated with Plasticity and Progression. JOURNAL OF ONCOLOGY 2019; 2019:1253727. [PMID: 31772577 PMCID: PMC6854237 DOI: 10.1155/2019/1253727] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 08/31/2019] [Indexed: 02/07/2023]
Abstract
With the advent of novel molecular platforms for high-throughput/next-generation sequencing, the communities of commensal and pathogenic microorganisms that inhabit the human body have been defined in depth. In the last decade, the role of microbiota-host interactions in driving human cancer plasticity and malignant progression has been well documented. Germ-free preclinical models provided an invaluable tool to demonstrate that the human microbiota can confer susceptibility to various types of cancer and can also modulate the host response to therapeutic treatments. Of interest, besides the detrimental effects of dysbiosis on cancer etiopathogenesis, specific microorganisms have been shown to exert protective activities against cancer growth. This has strong clinical implications, as restoration of the physiologic microbiota is being rapidly implemented as a novel anticancer therapeutic strategy. Here, we reviewed past and recent literature depicting the role of microbiota-host interactions in modulating key molecular mechanisms that drive human cancer plasticity and lead to malignant progression. We analyzed microbiota-host interactions occurring in the gut as well as in other anatomic sites, such as oral and nasal cavities, lungs, breast, esophagus, stomach, reproductive tract, and skin. We revealed a common ground of biological alterations and pathways modulated by a dysbiotic microbiota and potentially involved in the control of cancer progression. The molecular mechanisms most frequently affected by the pathogenic microorganisms to induce malignant progression involve epithelial-mesenchymal transition- (EMT-) dependent barrier alterations and tumor-promoting inflammation. This evidence may pave the way to better stratify high-risk cancer patients based on unique microenvironmental/microbial signatures and to develop novel, personalized, biological therapies.
Collapse
Affiliation(s)
- Daniele Vergara
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
- Laboratory of Clinical Proteomic, “Giovanni Paolo II” Hospital, ASL-Lecce, Italy
| | - Pasquale Simeone
- Department of Medicine and Aging Sciences, “G.d'Annunzio” University of Chieti-Pescara, Chieti, Italy
- Laboratory of Cytomorphology, Center for Advanced Studies and Technology (CAST), “G.d'Annunzio” University of Chieti-Pescara, Chieti, Italy
| | - Marina Damato
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
- Laboratory of Clinical Proteomic, “Giovanni Paolo II” Hospital, ASL-Lecce, Italy
| | - Michele Maffia
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
- Laboratory of Clinical Proteomic, “Giovanni Paolo II” Hospital, ASL-Lecce, Italy
| | - Paola Lanuti
- Department of Medicine and Aging Sciences, “G.d'Annunzio” University of Chieti-Pescara, Chieti, Italy
- Laboratory of Cytomorphology, Center for Advanced Studies and Technology (CAST), “G.d'Annunzio” University of Chieti-Pescara, Chieti, Italy
| | - Marco Trerotola
- Laboratory of Cancer Pathology, Center for Advanced Studies and Technology (CAST), “G.d'Annunzio” University of Chieti-Pescara, Chieti, Italy
- Department of Medical, Oral and Biotechnological Sciences, “G.d'Annunzio” University of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
109
|
Gunathilake MN, Lee J, Choi IJ, Kim YI, Ahn Y, Park C, Kim J. Association between the relative abundance of gastric microbiota and the risk of gastric cancer: a case-control study. Sci Rep 2019; 9:13589. [PMID: 31537876 PMCID: PMC6753194 DOI: 10.1038/s41598-019-50054-x] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 08/30/2019] [Indexed: 12/24/2022] Open
Abstract
The human gut hosts a diverse community of bacteria referred to as the gut microbiome. We investigated the association between the relative abundance of gastric microbiota and gastric cancer (GC) risk in a Korean population. The study participants included 268 GC patients and 288 controls. DNA was extracted from gastric biopsies, and 16S rRNA gene analysis was performed. Unconditional logistic regression models were used to observe the associations. Of the participants, those who had the highest level (highest tertile) of relative Helicobacter pylori and Propionibacterium acnes abundances showed a significantly higher risk for GC after adjusting for potential confounding variables (odds ratio (OR) = 1.86, 95% confidence interval (CI) = 1.17–2.97, p for trend = 0.017 and OR = 4.77, 95% CI = 2.94–7.74, p for trend <0.001, respectively). Subjects who carried Prevotella copri had a significantly higher risk of GC than noncarriers (OR = 2.54, 95% CI = 1.42–4.55, p for trend = 0.002). There was a lower risk of GC in subjects carrying Lactococcus lactis than in noncarriers (OR = 0.21, 95% CI = 0.10–0.44, p for trend <0.001). H. pylori, P. acnes and P. copri are strong risk factors, whereas L. lactis is a protective factor, for GC development in Koreans. Further microbiome studies are warranted to verify the findings of the current study.
Collapse
Affiliation(s)
- Madhawa Neranjan Gunathilake
- Department of Cancer Control and Population Health, Graduate School of Cancer Science and Policy, Goyang-si, 10408, Gyeonggi-do, South Korea
| | - Jeonghee Lee
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, Goyang-si, 10408, Gyeonggi-do, South Korea
| | - Il Ju Choi
- Center for Gastric Cancer, National Cancer Center Hospital, National Cancer Center, Goyang-si, 10408, Gyeonggi-do, South Korea
| | - Young-Il Kim
- Center for Gastric Cancer, National Cancer Center Hospital, National Cancer Center, Goyang-si, 10408, Gyeonggi-do, South Korea
| | - Yongju Ahn
- Microbiome Division, Theragen Etex, 145 Gwanggyo-ro, Gyeongtong-gu, Suwon-si, Gyeonggi-do, 16229, South Korea
| | - Chanhyeok Park
- Microbiome Division, Theragen Etex, 145 Gwanggyo-ro, Gyeongtong-gu, Suwon-si, Gyeonggi-do, 16229, South Korea
| | - Jeongseon Kim
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, Goyang-si, 10408, Gyeonggi-do, South Korea.
| |
Collapse
|
110
|
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
|
111
|
He C, Peng C, Wang H, Ouyang Y, Zhu Z, Shu X, Zhu Y, Lu N. The eradication of Helicobacter pylori restores rather than disturbs the gastrointestinal microbiota in asymptomatic young adults. Helicobacter 2019; 24:e12590. [PMID: 31124220 DOI: 10.1111/hel.12590] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 04/04/2019] [Accepted: 04/08/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND The eradication of Helicobacter pylori (H pylori) has been suggested to reduce the risk of gastric cancer, but its impact on the gut microbiota has attracted public attention. This study aimed to investigate the short-term and long-term effects of bismuth quadruple therapy on both gastric and fecal microbiota. METHODS Ten asymptomatic young adults with H pylori-related gastritis were treated with bismuth quadruple therapy for 14 days, and 7 age-matched adults without H pylori infection were enrolled as healthy controls. Both fecal and gastric mucosa samples were collected from H pylori-positive patients at weeks 0, 6, and 26, while fecal samples were collected from healthy controls. The gastric and gut microbiota were analyzed by 16S rRNA gene sequencing. RESULTS The structure of the gastric microbiota was significantly changed after the eradication of H pylori with increased alpha diversity over time. The relative abundance of H pylori sharply decreased from more than 70% to nearly 0% after treatment, while some beneficial bacteria, such as Lactobacillus and Bifidobacterium, were increased. The microbial diversity of gut microbiota was higher in H pylori-infected patients than in healthy controls, which tended to decrease after eradication. The potentially beneficial gut bacteria Blautia and Lachnoclostridium were enriched at week 26 compared to week 0, while the pathogenic Alistipes were depleted to a level close to that of the healthy controls. CONCLUSIONS Bismuth quadruple therapy for H pylori eradication can restore the diversity of gastric microbiota with enrichment of beneficial bacteria. The composition of gut microbiota after H pylori eradication trends toward healthy status instead of becoming dysbiotic.
Collapse
Affiliation(s)
- Cong He
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Chao Peng
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Huan Wang
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yaobin Ouyang
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhenhua Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xu Shu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yin Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Nonghua Lu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
112
|
Impact of the Gastrointestinal Microbiome in Health and Disease: Co-evolution with the Host Immune System. Curr Top Microbiol Immunol 2019; 421:303-318. [PMID: 31123894 DOI: 10.1007/978-3-030-15138-6_12] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Microbes within the gastrointestinal tract communicate with each other and with the host, which has profound effects on health and disease development. Only now, it is becoming apparent that how and when we acquire our own unique collection of "gut microbes" and also how we choose to maintain them is fundamental to our health. Helicobacter pylori is the most common bacterial infection worldwide, colonizing around half of the world's population, and is the major risk factor for gastric adenocarcinoma. More recently, it has also been shown to have some beneficial effects in terms of protecting against the development of other diseases. Here, we review the current knowledge on how H. pylori has shaped gastrointestinal microbiota colonization and the host immune system with specific focus on the impact of H. pylori on the various microbiome niches of the gastrointestinal tract. We discuss how the presence of H. pylori influences the physiology of three major regions within the gastrointestinal tract-specifically the oesophagus, stomach and colon. We pay particular attention to the role of H. pylori under chronic inflammatory conditions including the development of cancer. With increased incidence of diseases such as eosinophilic oesophagitis, oesophageal adenocarcinoma and squamous cell carcinoma being attributed to the decline in H. pylori, their disease pathogenesis in light of changing H. pylori colonization is also discussed.
Collapse
|
113
|
Dong Z, Chen B, Pan H, Wang D, Liu M, Yang Y, Zou M, Yang J, Xiao K, Zhao R, Zheng X, Zhang L, Zhang Y. Detection of Microbial 16S rRNA Gene in the Serum of Patients With Gastric Cancer. Front Oncol 2019; 9:608. [PMID: 31338330 PMCID: PMC6629868 DOI: 10.3389/fonc.2019.00608] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 06/20/2019] [Indexed: 01/03/2023] Open
Abstract
Aberrance in the blood bacterial microbiome has been identified and validated in several non-infectious diseases, including cancer. The occurrence and progression of gastric cancer has been found to be associated with alterations in the microbiome composition. However, the composition of the blood microbiome in patients with gastric cancer is not well-characterized. To test this hypothesis, we conducted a case-control study to investigate the microbiota compositions in the serum of patients with gastric cancer. The serum microbiome was investigated in patients with gastric cancer, atypical hyperplasia, chronic gastritis, and in healthy controls using 16S rRNA gene sequencing targeting the V1-V2 region. Our results revealed that the structure of the serum microbiome in gastric cancer was significantly different from all other groups, and alpha diversity decreased from the healthy control to patients with gastric cancer. The serum microbiome correlated significantly with tumor-node-metastasis (TNM) stage, lymphatic metastasis, tumor diameter, and invasion depth in gastric cancer. Three genera or species, namely, Acinetobacter, Bacteroides, Haemophilus parainfluenzae, were enriched in patients with gastric cancer, whereas Sphingomonas, Comamonas, and Pseudomonas stutzeri were enriched in the healthy control. Furthermore, the structure of serum microbiota differed between gastric cancer lymphatic metastasis and non-lymphatic metastasis. As a pilot investigation to characterizing the serum microbiome in gastric cancer, our study provided a foundation for improving our understanding of the role of microbiota in the pathogenesis of gastric cancer.
Collapse
Affiliation(s)
- Zhaogang Dong
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, China.,Shandong Province Key Laboratories of Medicine and Health (Tumor Marker Translational Medicine Laboratory), Qilu Hospital of Shandong University, Jinan, China
| | - Bin Chen
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing, China
| | - Hongwei Pan
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, China.,Shandong Province Key Laboratories of Medicine and Health (Tumor Marker Translational Medicine Laboratory), Qilu Hospital of Shandong University, Jinan, China
| | - Ding Wang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, China.,Shandong Province Key Laboratories of Medicine and Health (Tumor Marker Translational Medicine Laboratory), Qilu Hospital of Shandong University, Jinan, China
| | - Min Liu
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, China.,Shandong Province Key Laboratories of Medicine and Health (Tumor Marker Translational Medicine Laboratory), Qilu Hospital of Shandong University, Jinan, China
| | - Yongmei Yang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, China.,Shandong Province Key Laboratories of Medicine and Health (Tumor Marker Translational Medicine Laboratory), Qilu Hospital of Shandong University, Jinan, China
| | - Mingjin Zou
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, China.,Shandong Province Key Laboratories of Medicine and Health (Tumor Marker Translational Medicine Laboratory), Qilu Hospital of Shandong University, Jinan, China
| | - Junjie Yang
- College of Life Science, Qilu Normal University, Jinan, China
| | - Ke Xiao
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, China.,Shandong Province Key Laboratories of Medicine and Health (Tumor Marker Translational Medicine Laboratory), Qilu Hospital of Shandong University, Jinan, China
| | - Rui Zhao
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, China.,Shandong Province Key Laboratories of Medicine and Health (Tumor Marker Translational Medicine Laboratory), Qilu Hospital of Shandong University, Jinan, China
| | - Xin Zheng
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, China.,Shandong Province Key Laboratories of Medicine and Health (Tumor Marker Translational Medicine Laboratory), Qilu Hospital of Shandong University, Jinan, China
| | - Lei Zhang
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing, China.,Shandong Children's Microbiome Center, Qilu Children's Hospital of Shandong University, Jinan, China.,Qingdao Human Microbiome Center, The Affiliated Central Hospital of Qingdao University, Qingdao, China
| | - Yi Zhang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan, China.,Shandong Province Key Laboratories of Medicine and Health (Tumor Marker Translational Medicine Laboratory), Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
114
|
Mentis AFA, Boziki M, Grigoriadis N, Papavassiliou AG. Helicobacter pylori infection and gastric cancer biology: tempering a double-edged sword. Cell Mol Life Sci 2019; 76:2477-2486. [PMID: 30783683 PMCID: PMC11105440 DOI: 10.1007/s00018-019-03044-1] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Accepted: 02/08/2019] [Indexed: 12/15/2022]
Abstract
Helicobacter pylori (H. pylori) infection affects an estimated 4.4 billion people globally. Moreover, H. pylori presents the most significant risk factor for gastric cancer and low-grade mucosa-associated lymphoid tissue (MALT) lymphoma, and it is the first example of bacterial infection linked to carcinogenesis. Here, we contend that H. pylori research, which focuses on a cancer-causing pathogen resident in a relatively accessible organ, the stomach, could constitute an exemplar for microbial-related carcinogenesis in less tractable organs, such as the pancreas and lung. In this context, molecular biological approaches that could reap rewards are reviewed, including: (1) gastric cancer dynamics, particularly the role of stem cells and the heterogeneity of neoplastic cells, which are currently being investigated at the single-cell sequencing level; (2) mechanobiology, and the role of three-dimensional organoids and matrix metalloproteases; and (3) the connection between H. pylori and host pathophysiology and the gut microbiome. In the context of H. pylori's contribution to gastric cancer, several important conundrums remain to be fully elucidated. From among them, this article discusses (1) why H. pylori infection, which causes both gastric and duodenal inflammation, is only linked to gastric cancer; (2) whether a "precision oncomicrobiology" approach could enable a fine-tuning of the expression of only cancer-implicated H. pylori genes while maintaining beneficial H. pylori-mediated factors in extra-gastric tissues; and (3) the feasibility of using antibiotics targeting the microbial DNA damage system, which shares commonalities with mechanisms for human cell replication, as chemopreventives. Additional therapeutic perspectives are also discussed.
Collapse
Affiliation(s)
- Alexios-Fotios A Mentis
- Department of Medical Microbiology, Public Health Laboratories, Hellenic Pasteur Institute, Athens, Greece
- Department of Microbiology, University Hospital of Larissa, University of Thessaly, Larissa, Greece
| | - Marina Boziki
- Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nikolaos Grigoriadis
- Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Athanasios G Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 75 Mikras Asias Street, 11527, Athens, Greece.
| |
Collapse
|
115
|
Pero R, Brancaccio M, Laneri S, Biasi MGD, Lombardo B, Scudiero O. A Novel View of Human Helicobacter pylori Infections: Interplay between Microbiota and Beta-Defensins. Biomolecules 2019; 9:biom9060237. [PMID: 31216758 PMCID: PMC6627275 DOI: 10.3390/biom9060237] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 06/11/2019] [Accepted: 06/13/2019] [Indexed: 02/07/2023] Open
Abstract
The gut microbiota is significantly involved in the preservation of the immune system of the host, protecting it against the pathogenic bacteria of the stomach. The correlation between gut microbiota and the host response supports human gastric homeostasis. Gut microbes may be shifted in Helicobacter pylori (Hp)-infected individuals to advance gastric inflammation and distinguished diseases. Particularly interesting is the establishment of cooperation between gut microbiota and antimicrobial peptides (AMPs) of the host in the gastrointestinal tract. AMPs have great importance in the innate immune reactions to Hp and participate in conservative co-evolution with an intricate microbiome. β-Defensins, a class of short, cationic, arginine-rich proteins belonging to the AMP group, are produced by epithelial and immunological cells. Their expression is enhanced during Hp infection. In this review, we discuss the impact of the gut microbiome on the host response, with particular regard to β-defensins in Hp-associated infections. In microbial infections, mostly in precancerous lesions induced by Hp infection, these modifications could lead to different outcomes.
Collapse
Affiliation(s)
- Raffaela Pero
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II", 80131 Napoli, Italy.
- Task Force sugli Studi del Microbioma, Università degli Studi di Napoli "Federico II", 80131 Napoli, Italy.
| | - Mariarita Brancaccio
- Department of Biology and Evolution of Marine Organisms, Stazione Zoologica Anton Dohrn, 80121 Napoli, Italy.
| | - Sonia Laneri
- Dipartimento di Farmacia, Università degli Studi di Napoli "Federico II", Via Montesano 49, 80131 Napoli, Italy.
| | | | - Barbara Lombardo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II", 80131 Napoli, Italy.
- CEINGE-Biotecnologie Avanzate Scarl, Via G. Salvatore 486, 80145 Napoli, Italy.
| | - Olga Scudiero
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli "Federico II", 80131 Napoli, Italy.
- Task Force sugli Studi del Microbioma, Università degli Studi di Napoli "Federico II", 80131 Napoli, Italy.
- CEINGE-Biotecnologie Avanzate Scarl, Via G. Salvatore 486, 80145 Napoli, Italy.
| |
Collapse
|
116
|
Panebianco C, Pazienza V. Body site-dependent variations of microbiota in pancreatic cancer pathophysiology. Crit Rev Clin Lab Sci 2019; 56:260-273. [PMID: 31060399 DOI: 10.1080/10408363.2019.1615407] [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] [Indexed: 12/24/2022]
Abstract
Lack of specific symptoms and reliable biomarkers, along with aggressive nature and resistance to therapies makes pancreatic cancer (PC) one of the leading causes of death from cancer worldwide. The search for new diagnostic, prognostic, predictive, and therapeutic tools that could improve clinical outcomes of patients has led, in recent years, to the investigation of potential roles for the microbiota in the pathogenesis of this disease. The human microbiota encompasses trillions of microorganisms residing within several body tissues and organs, where they provide beneficial functions for host homeostasis and health. Derangements of the microbial ecology in different anatomic districts have been described in PC, as in many other diseases, both in patients and in animal models. In detail, infection from the gastric pathogen Helicobacter pylori and changes in composition and diversity of oral, intestinal, and pancreatic microbiota have been found to associate with PC. Future research should assess how to potentially exploit such differences in microbiota composition as diagnostic, prognostic, or predictive biomarkers, and as targets for therapeutic interventions, in the hope of improving the dismal prognosis of this insidious cancer.
Collapse
Affiliation(s)
- Concetta Panebianco
- a Division of Gastroenterology , Fondazione IRCCS Casa Sollievo della Sofferenza , San Giovanni Rotondo , Italy
| | - Valerio Pazienza
- a Division of Gastroenterology , Fondazione IRCCS Casa Sollievo della Sofferenza , San Giovanni Rotondo , Italy
| |
Collapse
|
117
|
Modification of the Gastric Mucosal Microbiota by a Strain-Specific Helicobacter pylori Oncoprotein and Carcinogenic Histologic Phenotype. mBio 2019; 10:mBio.00955-19. [PMID: 31138752 PMCID: PMC6538789 DOI: 10.1128/mbio.00955-19] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Microbial communities are essential for the maintenance of human health, and when these communities are altered, hosts can become susceptible to inflammation and disease. Dysbiosis contributes to gastrointestinal cancers, and specific bacterial species are associated with this phenotype. This study uses a robust and reproducible animal model to demonstrate that H. pylori infection induces gastric dysbiosis in a cagA-dependent manner and further that dysbiosis and altered microbial community structure parallel the severity of H. pylori-induced gastric injury. Ultimately, such models of H. pylori infection and cancer that can effectively evaluate multiple determinants simultaneously may yield effective strategies for manipulating the gastric microbiota to prevent the development of gastric cancer. Helicobacter pylori is the strongest risk factor for gastric adenocarcinoma; however, most infected individuals never develop this malignancy. Strain-specific microbial factors, such as the oncoprotein CagA, as well as environmental conditions, such as iron deficiency, augment cancer risk. Importantly, dysbiosis of the gastric microbiota is also associated with gastric cancer. To investigate the combinatorial effects of these determinants in an in vivo model of gastric cancer, Mongolian gerbils were infected with the carcinogenic cag+H. pylori strain 7.13 or a 7.13 cagA isogenic mutant, and microbial DNA extracted from gastric tissue was analyzed by 16S rRNA sequencing. Infection with H. pylori significantly increased gastric inflammation and injury, decreased α-diversity, and altered microbial community structure in a cagA-dependent manner. The effect of iron deficiency on gastric microbial communities was also investigated within the context of infection. H. pylori-induced injury was augmented under conditions of iron deficiency, but despite differences in gastric pathology, there were no significant differences in α- or β-diversity, phyla, or operational taxonomic unit (OTU) abundance among infected gerbils maintained on iron-replete or iron-depleted diets. However, when microbial composition was stratified based solely on the severity of histologic injury, significant differences in α- and β-diversity were present among gerbils harboring premalignant or malignant lesions compared to gerbils with gastritis alone. This study demonstrates that H. pylori decreases gastric microbial diversity and community structure in a cagA-dependent manner and that as carcinogenesis progresses, there are corresponding alterations in community structure that parallel the severity of disease.
Collapse
|
118
|
Suicide journey of H. pylori through gastric carcinogenesis: the role of non-H. pylori microbiome and potential consequences for clinical practice. Eur J Clin Microbiol Infect Dis 2019; 38:1591-1597. [PMID: 31114971 DOI: 10.1007/s10096-019-03564-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 04/09/2019] [Indexed: 12/24/2022]
|
119
|
Gantuya B, El-Serag HB, Matsumoto T, Ajami NJ, Oyuntsetseg K, Azzaya D, Uchida T, Yamaoka Y. Gastric Microbiota in Helicobacter pylori-Negative and -Positive Gastritis Among High Incidence of Gastric Cancer Area. Cancers (Basel) 2019; 11:E504. [PMID: 30974798 PMCID: PMC6520852 DOI: 10.3390/cancers11040504] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 04/02/2019] [Accepted: 04/04/2019] [Indexed: 02/07/2023] Open
Abstract
Helicobacter pylori (H. pylori) related chronic gastritis is a well-known major etiological factor for gastric cancer development. However, H. pylori-negative gastritis (HpN) is not well described. We aimed to examine gastric mucosal microbiota in HpN compared to H. pylori-positive gastritis (HpP) and H. pylori-negative non-gastritis group (control). Here, we studied 11 subjects with HpN, 40 with HpP and 24 controls. We performed endoscopy with six gastric biopsies. Comparison groups were defined based on strict histological criteria for the disease and H. pylori diagnosis. We used 16S rRNA gene amplicon sequencing to profile the gastric microbiota according to comparison groups. These results demonstrate that the HpP group had significantly lower bacterial richness by the operational taxonomic unit (OTU) counts, and Shannon and Simpson indices as compared to HpN or controls. The linear discriminant analysis effect size analysis showed the enrichment of Firmicutes, Fusobacteria, Bacteroidetes and Actinobacteria at phylum level in the HpN group. In the age-adjusted multivariate analysis, Streptococcus sp. and Haemophilus parainfluenzae were at a significantly increased risk for HpN (odds ratio 18.9 and 12.3, respectively) based on abundance. Treponema sp. was uniquely found in HpN based on occurrence. In this paper, we conclude that Streptococcus sp., Haemophilus parainfluenzae and Treponema sp. are candidate pathogenic bacterial species for HpN. These results if confirmed may have important clinical implications.
Collapse
Affiliation(s)
- Boldbaatar Gantuya
- Department of Environmental and Preventive Medicine, Oita University Faculty of Medicine, 1-1 Idaigaoka, Hasama-machi, Yufu-City, Oita 879-5593, Japan.
- Department of Internal Medicine, Gastroenterology Unit, Mongolian National University of Medical Sciences, Zorig Street, Ulaanbaatar-14210, Mongolia.
| | - Hashem B El-Serag
- Department of Medicine, Gastroenterology and Hepatology Section, Baylor College of Medicine, 7200 Cambridge Street, Houston, TX 77030, USA.
| | - Takashi Matsumoto
- Department of Environmental and Preventive Medicine, Oita University Faculty of Medicine, 1-1 Idaigaoka, Hasama-machi, Yufu-City, Oita 879-5593, Japan.
| | - Nadim J Ajami
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Khasag Oyuntsetseg
- Department of Internal Medicine, Gastroenterology Unit, Mongolian National University of Medical Sciences, Zorig Street, Ulaanbaatar-14210, Mongolia.
| | - Dashdorj Azzaya
- Department of Environmental and Preventive Medicine, Oita University Faculty of Medicine, 1-1 Idaigaoka, Hasama-machi, Yufu-City, Oita 879-5593, Japan.
- Department of Internal Medicine, Gastroenterology Unit, Mongolian National University of Medical Sciences, Zorig Street, Ulaanbaatar-14210, Mongolia.
| | - Tomohisa Uchida
- Department of Molecular Pathology, Oita University Faculty of Medicine, 1-1 Idaigaoka, Hasama-machi, Yufu-City, Oita 879-5593, Japan.
| | - Yoshio Yamaoka
- Department of Environmental and Preventive Medicine, Oita University Faculty of Medicine, 1-1 Idaigaoka, Hasama-machi, Yufu-City, Oita 879-5593, Japan.
- Department of Medicine, Gastroenterology and Hepatology Section, Baylor College of Medicine, 7200 Cambridge Street, Houston, TX 77030, USA.
- Global Oita Medical Advanced Research Center for Health, 1-1 Idaigaoka, Hasama-machi, Yufu-City, Oita 879-5593, Japan.
| |
Collapse
|
120
|
Endoplasmic reticulum proteostasis control and gastric cancer. Cancer Lett 2019; 449:263-271. [PMID: 30776479 DOI: 10.1016/j.canlet.2019.01.034] [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: 11/14/2018] [Revised: 01/16/2019] [Accepted: 01/26/2019] [Indexed: 02/07/2023]
Abstract
The endoplasmic reticulum (ER) is the primary organelle responsible for the synthesis, modification, folding and secretion of proteins, especially in specialized secretory cells. It also contributes to the maintenance of cellular functions, such as Ca2+ storage, lipogenesis, gluconeogenesis, and organelle biogenesis. Cellular stress conditions, such as glucose deprivation, hypoxia and disturbance of Ca2+ homeostasis, may increase the risk of protein misfolding and perturb proteostasis. This activates ER stress and triggers the unfolded protein response (UPR), leading to either the restoration of homeostasis or cell death. ER stress and UPR have been shown to play crucial roles in the pathogenesis, progression and treatment response of various cancers. In gastric cancer (GC), one of the most aggressive cancer types, critical functions of ER stress signaling have also started to emerge. Herein, we summarize the current knowledge linking ER stress and UPR to GC; we also discuss the possible nodes of therapeutic intervention and propose directions of future research.
Collapse
|
121
|
Dam B, Misra A, Banerjee S. Role of Gut Microbiota in Combating Oxidative Stress. OXIDATIVE STRESS IN MICROBIAL DISEASES 2019:43-82. [DOI: 10.1007/978-981-13-8763-0_4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
122
|
Butt J, Varga MG, Blot WJ, Teras L, Visvanathan K, Le Marchand L, Haiman C, Chen Y, Bao Y, Sesso HD, Wassertheil-Smoller S, Ho GY, Tinker LE, Peek RM, Potter JD, Cover TL, Hendrix LH, Huang LC, Hyslop T, Um C, Grodstein F, Song M, Zeleniuch-Jacquotte A, Berndt S, Hildesheim A, Waterboer T, Pawlita M, Epplein M. Serologic Response to Helicobacter pylori Proteins Associated With Risk of Colorectal Cancer Among Diverse Populations in the United States. Gastroenterology 2019; 156:175-186.e2. [PMID: 30296434 PMCID: PMC6309494 DOI: 10.1053/j.gastro.2018.09.054] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 09/12/2018] [Accepted: 09/27/2018] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Previous studies reported an association of the bacteria Helicobacter pylori, the primary cause of gastric cancer, and risk of colorectal cancer (CRC). However, these findings have been inconsistent, appear to vary with population characteristics, and may be specific for virulence factor VacA. To more thoroughly evaluate the potential association of H pylori antibodies with CRC risk, we assembled a large consortium of cohorts representing diverse populations in the United States. METHODS We used H pylori multiplex serologic assays to analyze serum samples from 4063 incident cases of CRC, collected before diagnosis, and 4063 matched individuals without CRC (controls) from 10 prospective cohorts for antibody responses to 13 H pylori proteins, including virulence factors VacA and CagA. The association of seropositivity to H pylori proteins, as well as protein-specific antibody level, with odds of CRC was determined by conditional logistic regression. RESULTS Overall, 40% of controls and 41% of cases were H pylori-seropositive (odds ratio [OR], 1.09; 95% CI, 0.99-1.20). H pylori VacA-specific seropositivity was associated with an 11% increased odds of CRC (OR, 1.11; 95% CI, 1.01-1.22), and this association was particularly strong among African Americans (OR, 1.45; 95% CI, 1.08-1.95). Additionally, odds of CRC increased with level of VacA antibody in the overall cohort (P = .008) and specifically among African Americans (P = .007). CONCLUSIONS In an analysis of a large consortium of cohorts representing diverse populations, we found serologic responses to H pylori VacA to associate with increased risk of CRC risk, particularly for African Americans. Future studies should seek to understand whether this marker is related to virulent H pylori strains carried in these populations.
Collapse
Affiliation(s)
- Julia Butt
- Infection and Cancer Epidemiology, Division of Molecular Diagnostics of Oncogenic Infections, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany (; ; )
- Cancer Control and Population Sciences Program, Duke Cancer Institute, and Department of Population Health Sciences, Duke University, 2424 Erwin Road, Suite 602, Durham, NC 27705, USA ()
| | - Matthew G. Varga
- University of North Carolina at Chapel Hill, Department of Epidemiology, Gillings School for Global Public Health and Lineberger Comprehensive Cancer Center, 2102E McGavran Greenberg Hall, Chapel Hill, NC 27599, USA ()
| | - William J. Blot
- Division of Epidemiology, Vanderbilt University Medical Center, 2525 West End Avenue Nashville, TN 37203, USA ()
| | - Lauren Teras
- Behavioral and Epidemiology Research Group, American Cancer Society, 250 Williams St, Atlanta, GA 30303, USA (; )
| | - Kala Visvanathan
- Department of Epidemiology, Johns Hopkins School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205 USA ()
| | - Loïc Le Marchand
- Epidemiology Program, University of Hawai’i Cancer Center, 701 Ilalo Street, Honolulu, HI 96813 USA ()
| | - Christopher Haiman
- University of Southern California and USC Norris Comprehensive Cancer Center, Los Angeles, CA, USA ()
| | - Yu Chen
- Department of Population Health, New York University School of Medicine, 650 First Avenue, New York, NY 10016 USA (; )
| | - Ying Bao
- Brigham and Women’s Hospital, 75 Francis Street, Boston, MA 02115 USA (; )
| | - Howard D. Sesso
- Brigham and Women’s Hospital, 75 Francis Street, Boston, MA 02115 USA (; )
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 667 Huntington Avenue, Boston, MA 02115 USA (; )
| | - Sylvia Wassertheil-Smoller
- Department of Epidemiology & Population Health, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461 USA ()
| | - Gloria Y.F. Ho
- Department of Occupational Medicine, Epidemiology and Prevention, Feinstein Institute for Medical Research, Northwell Health; Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Great Neck, NY 11021 USA ()
| | - Lesley E. Tinker
- Cancer Prevention Program, Division of Public Health Sciences at Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue, Seattle, WA 98109 USA ()
| | - Richard M. Peek
- Division of Gastroenterology, Department of Medicine, Vanderbilt University Medical Center, 2215 Garland Avenue, 1030C MRB IV (1025C), Nashville, TN 37232 USA, ()
| | - John D. Potter
- Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue, Seattle WA 98109 USA ()
| | - Timothy L. Cover
- Department of Medicine and Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232 USA; Veterans Affairs Tennessee Valley Healthcare System, Nashville, TN USA ()
| | - Laura H. Hendrix
- Department of Biostatistics and Bioinformatics, Duke University, 2424 Erwin Road, Durham, NC 27705 USA (; )
| | - Li-Ching Huang
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN 37203 USA ()
| | - Terry Hyslop
- Cancer Control and Population Sciences Program, Duke Cancer Institute, and Department of Population Health Sciences, Duke University, 2424 Erwin Road, Suite 602, Durham, NC 27705, USA ()
- Department of Biostatistics and Bioinformatics, Duke University, 2424 Erwin Road, Durham, NC 27705 USA (; )
| | - Caroline Um
- Behavioral and Epidemiology Research Group, American Cancer Society, 250 Williams St, Atlanta, GA 30303, USA (; )
| | - Francine Grodstein
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 667 Huntington Avenue, Boston, MA 02115 USA (; )
| | - Mingyang Song
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, 667 Huntington Avenue, Boston, MA 02115 USA (; )
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA 02115 USA ()
- Clinical and Translational Epidemiology Unit and Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02115 USA, ()
| | - Anne Zeleniuch-Jacquotte
- Department of Population Health, New York University School of Medicine, 650 First Avenue, New York, NY 10016 USA (; )
| | - Sonja Berndt
- Infections and Immunoepidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Room SG/6E102, Rockville, MD 20850 USA (; )
| | - Allan Hildesheim
- Infections and Immunoepidemiology Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, 9609 Medical Center Drive, Room SG/6E102, Rockville, MD 20850 USA (; )
| | - Tim Waterboer
- Infection and Cancer Epidemiology, Division of Molecular Diagnostics of Oncogenic Infections, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany (; ; )
| | - Michael Pawlita
- Infection and Cancer Epidemiology, Division of Molecular Diagnostics of Oncogenic Infections, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany (; ; )
| | - Meira Epplein
- Cancer Control and Population Sciences Program, Duke Cancer Institute, and Department of Population Health Sciences, Duke University, 2424 Erwin Road, Suite 602, Durham, NC 27705, USA ()
| |
Collapse
|
123
|
Tavera G, Morgan DR, Williams SM. Tipping the Scale Toward Gastric Disease: A Host-Pathogen Genomic Mismatch? CURRENT GENETIC MEDICINE REPORTS 2018; 6:199-207. [PMID: 30775159 PMCID: PMC6373874 DOI: 10.1007/s40142-018-0153-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE OF REVIEW Chronic infection with Helicobacter pylori infection is necessary but not sufficient to initiate development of intestinal-type gastric adenocarcinoma. It is not clear what additional factors tip the scale from commensal bacteria towards a pathogen that facilitates development of gastric cancer. Genetic variants in both the pathogen and host have been implicated, but neither alone explains a substantial portion of disease risk. RECENT FINDINGS In this review, we consider studies that address the important role of human and bacterial genetics, ancestry and their interactions in determining gastric disease risk. We observe gaps in the current literature that should guide future work to confirm the hypothesis of the interacting roles of host and bacterial genetics that will be necessary to translate these findings into clinically relevant information. SUMMARY We summarize genetic risk factors for gastric disease in both H. pylori and human hosts. However, genetic variation of one or the other organism in isolation insufficiently explains gastric disease risk. The most promising models of gastric disease risk simultaneously consider the genetic variation of both the H. pylori and human host, under a co-evolution model.
Collapse
Affiliation(s)
- Gloria Tavera
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Douglas R Morgan
- Vanderbilt Ingram Cancer Center, Nashville, Tennessee
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Scott M Williams
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
124
|
Abstract
Advances in our understanding of the metabolism and molecular functions of polyamines and their alterations in cancer have led to resurgence in the interest of targeting polyamine metabolism as an anticancer strategy. Increasing knowledge of the interplay between polyamine metabolism and other cancer-driving pathways, including the PTEN-PI3K-mTOR complex 1 (mTORC1), WNT signalling and RAS pathways, suggests potential combination therapies that will have considerable clinical promise. Additionally, an expanding number of promising clinical trials with agents targeting polyamines for both therapy and prevention are ongoing. New insights into molecular mechanisms linking dysregulated polyamine catabolism and carcinogenesis suggest additional strategies that can be used for cancer prevention in at-risk individuals. In addition, polyamine blocking therapy, a strategy that combines the inhibition of polyamine biosynthesis with the simultaneous blockade of polyamine transport, can be more effective than therapies based on polyamine depletion alone and may involve an antitumour immune response. These findings open up new avenues of research into exploiting aberrant polyamine metabolism for anticancer therapy.
Collapse
Affiliation(s)
- Robert A Casero
- Department of Oncology, Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA.
| | - Tracy Murray Stewart
- Department of Oncology, Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Anthony E Pegg
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, PA, USA
| |
Collapse
|
125
|
Abstract
The current article is a review of the most important, accessible, and relevant literature published between April 2017 and March 2018 on other Helicobacters and the gastric microbiome. The first part of the review focuses on literature describing non-Helicobacter pylori-Helicobacter (NHPH) infections in humans and animals whilst the subsequent section focuses specifically on the human gastric microbiome. Novel diagnostic methods as well as new NHPHs species have been identified in recent studies. Furthermore, our knowledge about the pathogenesis of NHPH infections has been further enhanced by important fundamental studies in cell lines and animal models. Over the last year, additional insights over the prevalence and potential prevention strategies of NHPHs have also been reported. With regard to understanding the gastric microbiome, new information detailing the structure of the gastric microbiota at different stages of H. pylori infection, within different patient geographical locations, was documented. There was also a study detailing the impact of proton-pump inhibitor usage and the effect on the gastric microbiome. Newer analysis approaches including defining the active microbiome through analysis of RNA rather than DNA-based sequencing were also published allowing the first assessments of the functional capabilities of the gastric microbiome.
Collapse
Affiliation(s)
- Juozas Kupcinskas
- Department of Gastroenterology and Institute for Digestive Research, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Georgina L Hold
- Microbiome Research Centre, St George & Sutherland Clinical School, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
126
|
The impact of Helicobacter pylori infection, eradication therapy and probiotic supplementation on gut microenvironment homeostasis: An open-label, randomized clinical trial. EBioMedicine 2018; 35:87-96. [PMID: 30145102 PMCID: PMC6161473 DOI: 10.1016/j.ebiom.2018.08.028] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 08/11/2018] [Accepted: 08/11/2018] [Indexed: 02/06/2023] Open
Abstract
Background Helicobacter pylori (H. pylori) infection is associated with remodeling of gastric microbiota. However, comprehensive analyses of the impact of H. pylori infection, eradication therapy and probiotic supplementation on gut microbiota are still lacking. We aimed to provide evidence for clinical decision making. Methods Seventy H. pylori-positive and 35 H. pylori-negative patients (group C) were enrolled. H. pylori-positive patients were randomly assigned to group A (14-day bismuth-containing quadruple therapy) and group B (quadruple therapy supplemented with Clostridium butyricum). Stool samples of group A and B were collected on day 0, 14 and 56 while stool samples of group C were collected on day 0. Gut microbiota was investigated by 16S rRNA sequencing. Findings The Sobs index (richness estimator) was significantly higher in H. pylori-positive samples than H. pylori-negative samples (p < .05). Several metabolic pathways were more abundant in H. pylori-positive communities while some disease-associated pathways had higher potential in H. pylori-negative community through KEGG pathway analysis. Abundances of most butyrate-producing bacteria significantly decreased, while several detrimental bacteria increased after eradication therapy. Probiotic supplementation was associated with improved gastrointestinal symptoms as well as increased Bacteroidetes:Firmicutes ratio. Interpretation While H. pylori infection may not be necessarily detrimental in all patients, eradication of H. pylori was associated with widespread changes in gut microbial ecology and structure. Probiotic supplementation could relieve more gastrointestinal symptoms by inducing alterations in gut microbiota and host immune responses. As such, the decision to eradicate H. pylori should be based on comprehensive analysis of individual patients.
Collapse
|
127
|
|
128
|
Chen X, Hu Y, Xie Y, Wang Y. High salt diet can down-regulate TFF2 expression level in gastric mucosa of MGs after H. pylori infection. Microb Pathog 2018; 118:316-321. [PMID: 29601867 DOI: 10.1016/j.micpath.2018.03.047] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 03/25/2018] [Accepted: 03/26/2018] [Indexed: 12/27/2022]
Abstract
This study aimed to elucidate the effect of Helicobacter pylori (H. pylori) and high salt diet on Trefoil factor 2 (TFF2) expression level of Mongolian gerbils (MGs) gastric mucosa. The results of H. pylori identification and histopathology showed that H. pylori infected MGs model was built successfully. According to the immunohistochemical staining results, 25% (4/16) of H. pylori infected MGs with high salt diet showed high TFF2 expression, which was significantly lower than H. pylori infection group 61% (11/18)(P = 0.045). The results suggested that High salt diet could down-regulated TFF2 expression level of MGs gastric mucosa induced by H. pylori infection.
Collapse
Affiliation(s)
- Xue Chen
- Department of Pathology, Shenyang Medical College, No.146, Huanghe North Avenue, Shenyang, Liaoning, China
| | - You Hu
- Department of Pathology, Shenyang Medical College, No.146, Huanghe North Avenue, Shenyang, Liaoning, China
| | - Ying Xie
- Department of Pathology, Shenyang Medical College, No.146, Huanghe North Avenue, Shenyang, Liaoning, China
| | - Ying Wang
- Department of Pathology, Shenyang Medical College, No.146, Huanghe North Avenue, Shenyang, Liaoning, China.
| |
Collapse
|
129
|
Pereira V, Abraham P, Nallapeta S, Shetty A. Gastric bacterial Flora in patients Harbouring Helicobacter pylori with or without chronic dyspepsia: analysis with matrix-assisted laser desorption ionization time-of-flight mass spectroscopy. BMC Gastroenterol 2018; 18:20. [PMID: 29373960 PMCID: PMC5787260 DOI: 10.1186/s12876-018-0744-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 01/14/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The gastric microbiota has recently been implicated in the causation of organic/structural gastroduodenal diseases (gastric and duodenal ulcers, gastric cancer) in patients with Helicobacter pylori (H. pylori) infection. We aimed to ascertain, in patients harbouring H. pylori, the role of the gastric microbiota in the causation of symptoms (chronic dyspepsia) in the absence of organic disease. METHODS Seventy-four gastric biopsy samples obtained at endoscopy from patients with (n = 21) or without (n = 53) chronic dyspepsia, and that tested positive by the bedside rapid urease test for H. pylori infection, were cultured for detection of H. pylori and non-H. pylori organisms. The cultured organisms were identified by matrix-assisted laser desorption ionization time-of-flight mass spectroscopy (MALDI-TOF MS). RESULTS A total of 106 non-H. pylori isolates were obtained from 74 patients' samples. This included 33 isolates (median 2, range 1-2 per patient) from dyspeptic and 73 (median 2, range 1-2 per patient) from non-dyspeptic patients. These were identified from the Bruker Biotyper 2 database as Staphylococcus spp., Streptococcus spp., Lactobacillus spp., Micrococcus spp., Enterococcus spp., Pseudomonas spp., Escherichia spp., Klebsiella spp. and Bacillus spp., Staphylococcus and Lactobacillus were identified significantly more commonly in dyspeptics and Streptococcus, Pseudomonas, Escherichia coli and Klebsiella pneumoniae in non-dyspeptics. All identified organisms belonged to the phyla Firmicutes and Proteobacteria. CONCLUSIONS There is a qualitative difference in the gastric microbial spectrum between patients harbouring H. pylori with and without chronic dyspepsia. Whether these organisms have an independent role in the development or prevention of dyspepsia or act in concurrence with H. pylori needs study.
Collapse
Affiliation(s)
- Verima Pereira
- Division of Gastroenterology, P D Hinduja Hospital, V S Marg, Mahim, Mumbai, 400016 India
| | - Philip Abraham
- Division of Gastroenterology, P D Hinduja Hospital, V S Marg, Mahim, Mumbai, 400016 India
| | | | - Anjali Shetty
- Division of Microbiology, P D Hinduja Hospital, Mumbai, India
| |
Collapse
|
130
|
Gorkiewicz G, Moschen A. Gut microbiome: a new player in gastrointestinal disease. Virchows Arch 2018; 472:159-172. [PMID: 29243124 PMCID: PMC5849673 DOI: 10.1007/s00428-017-2277-x] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 11/28/2017] [Accepted: 11/29/2017] [Indexed: 02/07/2023]
Abstract
The gastrointestinal (GI) tract harbors a diverse and host-specific gut microbial community. Whereas host-microbe interactions are based on homeostasis and mutualism, the microbiome also contributes to disease development. In this review, we summarize recent findings connecting the GI microbiome with GI disease. Starting with a description of biochemical factors shaping microbial compositions in each gut segment along the longitudinal axis, improved histological techniques enabling high resolution visualization of the spatial microbiome structure are highlighted. Subsequently, inflammatory and neoplastic diseases of the esophagus, stomach, and small and large intestines are discussed and the respective changes in microbiome compositions summarized. Finally, approaches aiming to restore disturbed microbiome compositions thereby promoting health are discussed.
Collapse
Affiliation(s)
- Gregor Gorkiewicz
- Institute of Pathology, Medical University of Graz, Neue Stiftingtalstrasse 6, 8010 Graz, Austria
| | - Alexander Moschen
- Christian Doppler Laboratory for Mucosal Immunology & Division of Internal Medicine I, Department of Medicine, Medical University Innsbruck, Peter-Mayr-Strasse 1, 6020 Innsbruck, Austria
| |
Collapse
|
131
|
McGee DJ, Lu XH, Disbrow EA. Stomaching the Possibility of a Pathogenic Role for Helicobacter pylori in Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2018; 8:367-374. [PMID: 29966206 PMCID: PMC6130334 DOI: 10.3233/jpd-181327] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 05/14/2018] [Indexed: 12/20/2022]
Abstract
While a small subset of Parkinson's disease cases have genetic causes, most cases are sporadic and may have an environmental contributor that has largely remained enigmatic. Remarkably, gastrointestinal symptoms in PD patients serve as a prodrome for the eventual motor dysfunctions. Herein, we review studies exploring a possible link between the gastric human pathogen Helicobacter pylori and PD. We provide plausible and testable hypotheses for how this organism might contribute to PD: 1) a toxin(s) produced by the bacteria; 2) disruption of the intestinal microbiome; 3) local inflammation that crosses the gut-brain axis, leading to neuroinflammation; and 4) manipulation of the pharmacokinetics of the PD drug levodopa by H. pylori, even in those not receiving exogenous levodopa. Key findings are: 1) people with PD are 1.5-3-fold more likely to be infected with H. pylori than people without PD; 2) H. pylori-infected PD patients display worse motor functions than H. pylori-negative PD patients; 3) eradication of H. pylori improves motor function in PD patients over PD patients whose H. pylori was not eradicated; and 4) eradication of H. pylori improves levodopa absorption in PD patients compared to that of PD patients whose H. pylori was not eradicated. Evidence is accumulating that H. pylori has a link with PD, but the mechanism is unclear. Future work should explore the effects of H. pylori on development of PD in defined PD animal models, focusing on the roles of H. pylori toxins, inflammation, levodopa absorption, and microbiome dysbiosis.
Collapse
Affiliation(s)
- David J. McGee
- Department of Microbiology and Immunology, LSU Health Sciences Center-Shreveport, Shreveport, LA, USA
| | - Xiao-Hong Lu
- Department of Pharmacology, Toxicology, and Neuroscience, LSU Health Sciences Center-Shreveport, Shreveport, LA, USA
| | - Elizabeth A. Disbrow
- Department of Pharmacology, Toxicology, and Neuroscience, LSU Health Sciences Center-Shreveport, Shreveport, LA, USA
- Department of Neurology, LSU Health Sciences Center-Shreveport, Shreveport, LA, USA
| |
Collapse
|
132
|
Klymiuk I, Bilgilier C, Stadlmann A, Thannesberger J, Kastner MT, Högenauer C, Püspök A, Biowski-Frotz S, Schrutka-Kölbl C, Thallinger GG, Steininger C. The Human Gastric Microbiome Is Predicated upon Infection with Helicobacter pylori. Front Microbiol 2017; 8:2508. [PMID: 29312210 PMCID: PMC5735373 DOI: 10.3389/fmicb.2017.02508] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 12/01/2017] [Indexed: 12/18/2022] Open
Abstract
The human gastric lumen is one of the most hostile environments of the human body suspected to be sterile until the discovery of Helicobacter pylori (H.p.). State of the art next generation sequencing technologies multiply the knowledge on H.p. functional genomics as well as on the colonization of supposed sterile human environments like the gastric habitat. Here we studied in a prospective, multicenter, clinical trial the 16S rRNA gene amplicon based bacterial microbiome in a total of 30 homogenized and frozen gastric biopsy samples from eight geographic locations. The evaluation of the samples for H.p. infection status was done by histopathology and a specific PCR assay. CagA status was determined by a CagA-specific PCR assay. Patients were grouped accordingly as H.p.-negative, H.p.-positive but CagA-negative and H.p.-positive and CagA-positive (n = 10, respectively). Here we show that H.p. infection of the gastric habitat dominates the gastric microbiota in most patients and is associated with a significant decrease of the microbial alpha diversity from H.p. negative to H.p. positive with CagA as a considerable factor. The genera Actinomyces, Granulicatella, Veillonella, Fusobacterium, Neisseria, Helicobacter, Streptococcus, and Prevotella are significantly different between the H.p.-positive and H.p.-negative sample groups. Differences in microbiota found between CagA-positive and CagA-negative patients were not statistically significant and need to be re-evaluated in larger sample cohorts. In conclusion, H.p. infection dominates the gastric microbiome in a multicentre cohort of patients with varying diagnoses.
Collapse
Affiliation(s)
- Ingeborg Klymiuk
- Center for Medical Research, Medical University of Graz, Graz, Austria
| | - Ceren Bilgilier
- Division of Infectious Diseases, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Alexander Stadlmann
- Division of Infectious Diseases, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Jakob Thannesberger
- Division of Infectious Diseases, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Marie-Theres Kastner
- Division of Infectious Diseases, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Christoph Högenauer
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Graz, Austria
| | - Andreas Püspök
- Department of Internal Medicine II, St. John's Hospital Eisenstadt, Eisenstadt, Austria
| | - Susanne Biowski-Frotz
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University Vienna, Vienna, Austria
| | - Christiane Schrutka-Kölbl
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University Vienna, Vienna, Austria
| | - Gerhard G. Thallinger
- Institute of Computational Biotechnology, Graz University of Technology, Graz, Austria
- BioTechMed OMICS Center Graz, Graz, Austria
| | - Christoph Steininger
- Division of Infectious Diseases, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| |
Collapse
|