1
|
Zhao SQ, Zheng HL, Zhong XT, Wang ZY, Su Y, Shi YY. Effects and mechanisms of Helicobacter pylori infection on the occurrence of extra-gastric tumors. World J Gastroenterol 2024; 30:4090-4103. [DOI: 10.3748/wjg.v30.i37.4090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 08/23/2024] [Accepted: 09/12/2024] [Indexed: 09/26/2024] Open
Abstract
Helicobacter pylori (H. pylori) colonizes the human stomach and many studies have discussed the mechanisms of H. pylori infection leading to gastric diseases, including gastric cancer. Additionally, increasing data have shown that the infection of H. pylori may contribute to the development of extra-gastric diseases and tumors. Inflammation, systemic immune responses, microbiome disorders, and hypergastrinemia caused by H. pylori infection are associated with many extra-gastric malignancies. This review highlights recent discoveries; discusses the relationship between H. pylori and various extra-gastric tumors, such as colorectal cancer, lung cancer, cholangiocarcinoma, and gallbladder carcinoma; and explores the mechanisms of extra-gastric carcinogenesis by H. pylori. Overall, these findings refine our understanding of the pathogenic processes of H. pylori, provide guidance for the clinical treatment and management of H. pylori-related extra-gastric tumors, and help improve prognosis.
Collapse
Affiliation(s)
- Shi-Qing Zhao
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing 100191, China
- Health Science Center, Peking University, Beijing 100191, China
| | - Hui-Ling Zheng
- Department of Gastroenterology, Peking University Third Hospital, Beijing 100191, China
| | - Xiao-Tian Zhong
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing 100191, China
- Health Science Center, Peking University, Beijing 100191, China
| | - Zi-Ye Wang
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing 100191, China
- Health Science Center, Peking University, Beijing 100191, China
| | - Yi Su
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing 100191, China
- Health Science Center, Peking University, Beijing 100191, China
| | - Yan-Yan Shi
- Research Center of Clinical Epidemiology, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
2
|
Alkeridy W, Alanezi K, Alshehri FK, Alkhedr M, Albabtain MA, Alamri M, Jabaan R, Almugren A, Alsahafi M, Alkhowaiter SS. The Prevalence of Gastroesophageal Reflux Disease (GERD) in H. pylori-Positive and -Negative Patients. Cureus 2024; 16:e72059. [PMID: 39569223 PMCID: PMC11578635 DOI: 10.7759/cureus.72059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/18/2024] [Indexed: 11/22/2024] Open
Abstract
INTRODUCTION The presence of Helicobacter pylori (H. pylori) infection can influence gastric acid production. While both gastroesophageal reflux disease (GERD) and H. pylori are highly prevalent in the Saudi Arabian population, the relationship between these two conditions remains uncertain. This study aims to assess the association between GERD and H. pylori. METHODS This was a cross-sectional study which included patients who underwent endoscopy with gastric biopsy to test for H. pylori. H. pylori status was assessed using histopathology. The GERD-Q questionnaire was used to ascertain the presence of GERD. Multivariate analysis was used to assess the relationship between H. pylori and GERD. RESULTS A total of 352 participants were included, with a mean age of 45 years (±14.7) and 58% of them being female. 148 participants (42%) tested positive for H. pylori. 59 (39.9%) had GERD. GERD was identified in 39.9% of the H. pylori-positive group and 37.3% of the H. pylori-negative group. H. pylori was not significantly associated with GERD in the multivariate analysis (P = 0.726, OR = 1.089, 95% CI: 0.675 - 1.759). CONCLUSION There was no association between H. pylori infection and GERD in this Saudi Arabian population.
Collapse
Affiliation(s)
- Walid Alkeridy
- Department of Medicine, College of Medicine, King Saud University, Riyadh, SAU
- Department of Medicine, Geriatric Division, University of British Columbia, Vancouver, CAN
- General Administration of Home Health Care, Therapeutic Affairs Deputyship, Riyadh, SAU
| | - Khalid Alanezi
- Department of Medicine, College of Medicine, King Saud University, Riyadh, SAU
| | - Faisal K Alshehri
- Department of Medicine, College of Medicine, King Saud University, Riyadh, SAU
| | - Mudafr Alkhedr
- Department of Medicine, College of Medicine, King Saud University, Riyadh, SAU
| | | | - Musab Alamri
- Department of Medicine, College of Medicine, King Saud University, Riyadh, SAU
| | - Rayan Jabaan
- Department of Medicine, College of Medicine, King Saud University, Riyadh, SAU
| | | | - Majid Alsahafi
- Division of Gastroenterology, Department of Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, SAU
| | - Saad S Alkhowaiter
- Department of Medicine - Gastroenterology, College of Medicine, King Saud University, Riyadh, SAU
| |
Collapse
|
3
|
Argüero J, Sifrim D. Pathophysiology of gastro-oesophageal reflux disease: implications for diagnosis and management. Nat Rev Gastroenterol Hepatol 2024; 21:282-293. [PMID: 38177402 DOI: 10.1038/s41575-023-00883-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/28/2023] [Indexed: 01/06/2024]
Abstract
Gastro-oesophageal reflux disease (GERD) is a common gastrointestinal disorder in which retrograde flow of gastric content into the oesophagus causes uncomfortable symptoms and/or complications. It has a multifactorial and partially understood pathophysiology. GERD starts in the stomach, where the refluxate material is produced. Following the trajectory of reflux, the failure of the antireflux barrier, primarily the lower oesophageal sphincter and the crural diaphragm, enables the refluxate to reach the oesophageal lumen, triggering oesophageal or extra-oesophageal symptoms. Reflux clearance mechanisms such as primary and secondary peristalsis and the arrival of bicarbonate-rich saliva are critical to prevent mucosal damage. Alterations of the oesophageal mucosal integrity, such as macroscopic oesophagitis or microscopic changes, determine the perception of symptoms. The intensity of the symptoms is affected by peripheral and central neural and psychological mechanisms. In this Review, we describe an updated understanding of the complex and multifactorial pathophysiology of GERD. It is now recognized that different GERD phenotypes have different degrees of reflux, severity of mucosal integrity damage and type, and severity of symptoms. These variations are probably due to the occurrence of a predominant pathophysiological mechanism in each patient. We also describe the main pathophysiological mechanisms of GERD and their implications for personalized diagnosis and management.
Collapse
Affiliation(s)
- Julieta Argüero
- Neurogastroenterology section of Gastroenterology Department, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Daniel Sifrim
- Wingate Institute of Neurogastroenterology, Queen Mary University of London, London, UK.
| |
Collapse
|
4
|
Kim J. Gastroesophageal Reflux Disease. HELICOBACTER PYLORI 2023:389-393. [DOI: 10.1007/978-981-97-0013-4_30] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
5
|
Ma S, Guo X, Wang C, Yin Y, Xu G, Chen H, Qi X. Association of Barrett's esophagus with Helicobacter pylori infection: a meta-analysis. Ther Adv Chronic Dis 2022; 13:20406223221117971. [PMID: 36034104 PMCID: PMC9403448 DOI: 10.1177/20406223221117971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 07/19/2022] [Indexed: 11/25/2022] Open
Abstract
Background and Aims: Barrett’s esophagus (BE) is the only recognized precursor for esophageal
adenocarcinoma. Helicobacter pylori (H.
pylori) infection is a major contributing factor towards upper
gastrointestinal diseases, but its relationship with BE remains
controversial. Some previous studies suggested that H.
pylori infection negatively correlated with BE, while others
did not. This may be attributed to the difference in the selection of
control groups among studies. The present meta-analysis aims to clarify
their association by combining all available data from well-designed
studies. Methods: The PubMed, EMBASE, and Cochrane
Library databases were searched. Odds ratios (ORs) with 95%
confidence intervals (CIs) were pooled by a random-effects model.
Heterogeneity was evaluated using the Cochran’s Q test and
I2 statistics. Meta-regression, subgroup,
and leave-one-out sensitivity analyses were employed to explore the sources
of heterogeneity. Results: Twenty-four studies with 1,354,369 participants were included. Meta-analysis
found that patients with BE had a significantly lower prevalence of
H. pylori infection than those without (OR = 0.53, 95%
CI = 0.45–0.64; p < 0.001). The heterogeneity was
statistically significant (I² = 79%;
p < 0.001). Meta-regression, subgroup, and leave-one-out
sensitivity analyses did not find any source of heterogeneity. Meta-analysis
of 7 studies demonstrated that CagA-positive H. pylori
infection inversely correlated with BE (OR = 0.25, 95% CI = 0.15–0.44;
p = 0.000), but not CagA-negative H.
pylori infection (OR = 1.22, 95% CI = 0.90–1.67;
p = 0.206). Meta-analysis of 4 studies also
demonstrated that H. pylori infection inversely correlated
with LSBE (OR = 0.39, 95% CI = 0.18–0.86; p = 0.019), but
not SSBE (OR = 0.73, 95% CI = 0.30–1.77; p = 0.484). Conclusion: H. pylori infection negatively correlates with BE. More
experimental studies should be necessary to elucidate the potential
mechanisms in future.
Collapse
Affiliation(s)
| | | | | | | | - Guangqin Xu
- Department of Gastroenterology, General
Hospital of Northern Theater Command, Shenyang, China
- Graduate School, Dalian Medical University,
Dalian, China
| | - Hongxin Chen
- Department of Gastroenterology, General
Hospital of Northern Theater Command, Shenyang, China
- Graduate School, Liaoning University of
Traditional Chinese Medicine, Shenyang, China
| | | |
Collapse
|
6
|
Du YL, Duan RQ, Duan LP. Helicobacter pylori infection is associated with reduced risk of Barrett's esophagus: a meta-analysis and systematic review. BMC Gastroenterol 2021; 21:459. [PMID: 34876031 PMCID: PMC8650239 DOI: 10.1186/s12876-021-02036-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 11/16/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Helicobacter pylori (Hp) is a class I carcinogen in gastric carcinogenesis, but its role in Barrett's esophagus (BE) is unknown. Therefore, we aimed to explore the possible relationship. METHODS We reviewed observational studies published in English until October 2019. Summary odds ratios (ORs) and 95% confidence intervals (CIs) were calculated for included studies. RESULTS 46 studies from 1505 potential citations were eligible for inclusion. A significant inverse relationship with considerable heterogeneity was found between Hp (OR = 0.70; 95% CI, 0.51-0.96; P = 0.03) and BE, especially the CagA-positive Hp strain (OR = 0.28; 95% CI, 0.15-0.54; P = 0.0002). However, Hp infection prevalence was not significantly different between patients with BE and the gastroesophageal reflux disease (GERD) control (OR = 0.99; 95% CI, 0.82-1.19; P = 0.92). Hp was negatively correlated with long-segment BE (OR = 0.47; 95% CI, 0.25-0.90; P = 0.02) and associated with a reduced risk of dysplasia. However, Hp had no correlated with short-segment BE (OR = 1.11; 95% CI, 0.78-1.56; P = 0.57). In the present infected subgroup, Hp infection prevalence in BE was significantly lower than that in controls (OR = 0.69; 95% CI, 0.54-0.89; P = 0.005); however, this disappeared in the infection history subgroup (OR = 0.88; 95% CI, 0.43-1.78; P = 0.73). CONCLUSIONS Hp, especially the CagA-positive Hp strain, and BE are inversely related with considerable heterogeneity, which is likely mediated by a decrease in GERD prevalence, although this is not observed in the absence of current Hp infection.
Collapse
Affiliation(s)
- Yan-Lin Du
- Department of Gastroenterology, Peking University Third Hospital, No. 49 North Garden Rd., Haidian District, Beijing, 100191, China
| | - Ru-Qiao Duan
- Department of Gastroenterology, Peking University Third Hospital, No. 49 North Garden Rd., Haidian District, Beijing, 100191, China
| | - Li-Ping Duan
- Department of Gastroenterology, Peking University Third Hospital, No. 49 North Garden Rd., Haidian District, Beijing, 100191, China.
| |
Collapse
|
7
|
Georgiou K, Marinov B, Farooqi AA, Gazouli M. Gut Microbiota in Lung Cancer: Where Do We Stand? Int J Mol Sci 2021; 22:10429. [PMID: 34638770 PMCID: PMC8508914 DOI: 10.3390/ijms221910429] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 09/19/2021] [Accepted: 09/26/2021] [Indexed: 02/06/2023] Open
Abstract
The gut microbiota (GM) is considered to constitute a powerful "organ" capable of influencing the majority of the metabolic, nutritional, physiological, and immunological processes of the human body. To date, five microbial-mediated mechanisms have been revealed that either endorse or inhibit tumorigenesis. Although the gastrointestinal and respiratory tracts are distant physically, they have common embryonic origin and similarity in structure. The lung microbiota is far less understood, and it is suggested that the crosslink between the human microbiome and lung cancer is a complex, multifactorial relationship. Several pathways linking their respective microbiota have reinforced the existence of a gut-lung axis (GLA). Regarding implications of specific GM in lung cancer therapy, a few studies showed that the GM considerably affects immune checkpoint inhibitor (ICI) therapy by altering the differentiation of regulatory T cells and thus resulting in changes in immunomodulation mechanisms, as discovered by assessing drug metabolism directly and by assessing the host immune modulation response. Additionally, the GM may increase the efficacy of chemotherapeutic treatment in lung cancer. The mechanism underlying the role of the GLA in the pathogenesis and progression of lung cancer and its capability for diagnosis, manipulation, and treatment need to be further explored.
Collapse
Affiliation(s)
- Konstantinos Georgiou
- 1st Department of Propaedeutic Surgery, Hippokration General Hospital of Athens, Athens Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Blagoi Marinov
- Medical Simulation Training Center at Research Institute of Medical University of Plovdiv, Tsentar, 4002 Plovdiv, Bulgaria;
| | - Ammad Ahmad Farooqi
- Institute of Biomedical and Genetic Engineering (IBGE), 24 Mauve Area, Sector G-9/1, Islamabad 54000, Pakistan;
| | - Maria Gazouli
- Laboratory of Biology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
8
|
Lynch KL, Falk GW. Helicobacter pylori and GERD. THE ESOPHAGUS 2021:419-427. [DOI: 10.1002/9781119599692.ch24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
9
|
Varga MG, Wood CR, Butt J, Ryan ME, You WC, Pan K, Waterboer T, Epplein M, Shaffer CL. Immunostimulatory membrane proteins potentiate H. pylori-induced carcinogenesis by enabling CagA translocation. Gut Microbes 2021; 13:1-13. [PMID: 33382363 PMCID: PMC7781638 DOI: 10.1080/19490976.2020.1862613] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 11/04/2020] [Accepted: 11/26/2020] [Indexed: 02/07/2023] Open
Abstract
Infection with Helicobacter pylori is the single greatest risk factor for developing gastric adenocarcinoma. In prospective, population-based studies, seropositivity to the uncharacterized H. pylori proteins Hp0305 and Hp1564 was significantly associated with cancer risk in East Asia. However, the mechanism underlying this observation has not been elucidated. Here, we show that Hp0305 and Hp1564 act in concert with previously ascribed H. pylori virulence mechanisms to orchestrate cellular alterations that promote gastric carcinogenesis. In samples from 546 patients exhibiting premalignant gastric lesions, seropositivity to Hp0305 and Hp1564 was significantly associated with increased gastric atrophy across all stomach conditions. In vitro, depletion of Hp0305 and Hp1564 significantly reduced levels of gastric cell-associated bacteria and markedly impaired the ability of H. pylori to stimulate pro-inflammatory cytokine production. Remarkably, our studies revealed that Hp1564 is required for translocation of the oncoprotein CagA into gastric epithelial cells. Our data provide experimental insight into the molecular mechanisms governing novel H. pylori pathogenicity factors that are strongly associated with gastric disease and highlight the potential of Hp0305 and Hp1564 as robust molecular tools that can improve identification of individuals that are highly susceptible to gastric cancer. We demonstrate that Hp0305 and Hp1564 augment H. pylori-mediated inflammation and gastric cancer risk by promoting key bacteria-gastric cell interactions that facilitate delivery of oncogenic microbial cargo to target cells. Thus, therapeutically targeting microbial interactions driven by Hp0305/Hp1564 may enable focused H. pylori eradication strategies to prevent development of gastric malignancies in high-risk populations.
Collapse
Affiliation(s)
- Matthew G. Varga
- Department of Epidemiology, Lineberger Comprehensive Cancer Center and Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA
| | - Cecily R. Wood
- Department of Veterinary Science, University of Kentucky, Lexington, KY, USA
| | - Julia Butt
- Infections and Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | - Mackenzie E. Ryan
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY, USA
| | - Wei-Cheng You
- Department of Cancer Epidemiology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Kaifeng Pan
- Department of Cancer Epidemiology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Tim Waterboer
- Infections and Cancer Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | - Meira Epplein
- Department of Population Health Sciences and Duke Cancer Institute, Cancer Control and Population Sciences Program, Duke University, Durham, NC, USA
| | - Carrie L. Shaffer
- Department of Veterinary Science, University of Kentucky, Lexington, KY, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky, Lexington, KY, USA
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
10
|
Fisher L, Fisher A, Smith PN. Helicobacter pylori Related Diseases and Osteoporotic Fractures (Narrative Review). J Clin Med 2020; 9:E3253. [PMID: 33053671 PMCID: PMC7600664 DOI: 10.3390/jcm9103253] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/28/2020] [Accepted: 10/07/2020] [Indexed: 02/06/2023] Open
Abstract
Osteoporosis (OP) and osteoporotic fractures (OFs) are common multifactorial and heterogenic disorders of increasing incidence. Helicobacter pylori (H.p.) colonizes the stomach approximately in half of the world's population, causes gastroduodenal diseases and is prevalent in numerous extra-digestive diseases known to be associated with OP/OF. The studies regarding relationship between H.p. infection (HPI) and OP/OFs are inconsistent. The current review summarizes the relevant literature on the potential role of HPI in OP, falls and OFs and highlights the reasons for controversies in the publications. In the first section, after a brief overview of HPI biological features, we analyze the studies evaluating the association of HPI and bone status. The second part includes data on the prevalence of OP/OFs in HPI-induced gastroduodenal diseases (peptic ulcer, chronic/atrophic gastritis and cancer) and the effects of acid-suppressive drugs. In the next section, we discuss the possible contribution of HPI-associated extra-digestive diseases and medications to OP/OF, focusing on conditions affecting both bone homeostasis and predisposing to falls. In the last section, we describe clinical implications of accumulated data on HPI as a co-factor of OP/OF and present a feasible five-step algorithm for OP/OF risk assessment and management in regard to HPI, emphasizing the importance of an integrative (but differentiated) holistic approach. Increased awareness about the consequences of HPI linked to OP/OF can aid early detection and management. Further research on the HPI-OP/OF relationship is needed to close current knowledge gaps and improve clinical management of both OP/OF and HPI-related disorders.
Collapse
Affiliation(s)
- Leon Fisher
- Department of Gastroenterology, Frankston Hospital, Peninsula Health, Melbourne 3199, Australia
| | - Alexander Fisher
- Department of Geriatric Medicine, The Canberra Hospital, ACT Health, Canberra 2605, Australia;
- Department of Orthopedic Surgery, The Canberra Hospital, ACT Health, Canberra 2605, Australia;
- Australian National University Medical School, Canberra 2605, Australia
| | - Paul N Smith
- Department of Orthopedic Surgery, The Canberra Hospital, ACT Health, Canberra 2605, Australia;
- Australian National University Medical School, Canberra 2605, Australia
| |
Collapse
|
11
|
Association Between Helicobacter pylori Infection and Short-segment/Long-segment Barrett's Esophagus in a Japanese Population: A Large Cross-Sectional Study. J Clin Gastroenterol 2020; 54:439-444. [PMID: 31524650 DOI: 10.1097/mcg.0000000000001264] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
GOAL The goal of this study was to investigate the relationship between Helicobacter pylori (H. pylori) infection and short-segment and long-segment Barrett's esophagus (SSBE and LSBE). BACKGROUND H. pylori infection is reported to be inversely associated with Barrett's esophagus (BE) in western countries. However, the impact of BE segment length on the association between BE and H. pylori infection has scarcely been investigated. MATERIALS AND METHODS The study subjects were 41,065 asymptomatic Japanese individuals who took medical surveys between October 2010 and September 2017. Using this large database of healthy Japanese subjects, we investigated the association between H. pylori infection and SSBE/LSBE. We used multivariable logistic regression analysis to estimate odds ratios (ORs) and 95% confidence intervals (CIs). RESULTS Among the study subjects, 36,615 were eligible for the analysis. H. pylori seropositivity was significantly associated with a lower rate of LSBE (OR: 0.42; 95% CI: 0.16-0.91) and a higher rate of SSBE (OR: 1.66; 95% CI: 1.56-1.78) after multivariate adjustment. In the subgroup analysis, H. pylori seropositivity was significantly associated with a high rate of SSBE in subjects without reflux esophagitis (RE) (OR: 1.73; 95% CI: 1.61-1.85). However, H. pylori seropositivity was not associated with SSBE in subjects with RE (OR: 1.07; 95% CI: 0.84-1.37). CONCLUSION In a Japanese population, H. pylori infection was inversely associated with LSBE but significantly associated with SSBE only in subjects without RE. H. pylori may be a risk factor for SSBE, especially in individuals without RE.
Collapse
|
12
|
Aghayeva S, Mara KC, Katzka DA. The impact of Helicobacter pylori on the presence of Barrett's esophagus in Azerbaijan, a high-prevalence area of infection. Dis Esophagus 2019; 32:5512615. [PMID: 31175364 DOI: 10.1093/dote/doz053] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 04/09/2019] [Accepted: 04/30/2019] [Indexed: 12/11/2022]
Abstract
There is a strong evidence that Helicobacter pylori infection is inversely associated with Barrett's esophagus. In a high-prevalence region of H. pylori, low rates of esophageal cancer and its precursor BE may indicate its preventive effect. The aim of this study is to determine the impact of H. pylori on characteristics of Barrett's esophagus. A total of 3317 outpatient upper endoscopy reports from 2013 to 2015 from an urban center in Azerbaijan from all patients with dyspepsia were retrospectively analyzed for patients with Barrett's esophagus. This was matched in a 1:2 ratio to age and gender matched control patients without Barrett's esophagus. The prevalence of H. pylori on Barrett's esophagus and the randomly selected control group were compared. There were 83 patients with BE and 167 control group cases. Biopsy-proven BE was diagnosed in 83 patients: 39 (47%) females, with mean age 43.1 ± 13.3 years. Of these, 13 (15.7%) had long segment and 70 (84.3%) had short segment Barrett's esophagus. A control group included 167 patients: 78 (46.7%) females, with mean age (45.8 ± 13.9). All patients were Caucasians. The rates of gastric inflammation, the presence of atrophy, and intestinal metaplasia in gastric specimens did not differ in patients versus controls. The prevalence of H. pylori was determined as 63.2% in male and 61.5 in female groups (odd ratio (OR) = 0.99 95%CI 0.97, 1.01; P = 0.22). Inflammation of gastric mucosa was strongly associated with the infection (67% vs. 33%; OR = 4.46 95% CI: 2.01, 9.92, P < 0.001). Atrophy was noted in majority of H. pylori-positive cases (OR = 1.43, 95% CI: 0.36, 5.65; P = 0.61). Gastric intestinal metaplasia was observed in 55.6% of H. pylori-positive patients and in 44.4% of negative individuals (OR = 0.74, 95% CI: 0.28, 1.94; P = 0.54). There was not a significant difference in the prevalence of HP in BE and control groups; 63.9% were positive for infection in BE cases and 61.7% of controls (OR = 1.10, 95% CI: 0.64, 1.90; P = 0.74). We found that neither presence of erosive esophagitis, length of BE nor dysplasia (45.5% of H. pylori-positive group, whereas 54.5%) was associated with the presence of the H. pylori infection (Table 1). In a predominantly Caucasian nation with a high prevalence of H. pylori gastritis, the presence of H. pylori was not inversely associated with the presence of Barrett's esophagus. These data challenge the mechanistic implications of this association.
Collapse
Affiliation(s)
| | - K C Mara
- Biomedical statistics and Informatics
| | - D A Katzka
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, USA
| |
Collapse
|
13
|
Degiovani M, Ribas CAPM, Czeczko NG, Parada AA, Fronchetti JDA, Malafaia O. IS THERE A RELATION BETWEEN HELYBACTER PYLORI AND INTESTINAL METAPLASIA IN SHORT COLUMN EPITELIZATION UP TO 10 MM IN THE DISTAL ESOPHAGUS? ACTA ACUST UNITED AC 2019; 32:e1480. [PMID: 31859933 PMCID: PMC6918731 DOI: 10.1590/0102-672020190001e1480] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 05/08/2019] [Indexed: 12/16/2022]
Abstract
Background: The presence of intestinal metaplasia in the distal esophagus (Barrett’s esophagus) is an important precursor of adenocarcinoma. Knowledge of the risk factors and the process by which the Barrett develops is very important and Helicobacter pylori (HP) can contribute to this development. Aim: To analyze the impact of HP in the gastric mucosa with intestinal metaplasia in the distal esophagus in areas of columnar epithelialization smaller than 10 mm in length and epidemiological data on prevalence Method: A retrospective study in which were included 373 consecutive patients diagnosed with columnar epithelium in the distal esophagus was done. In all, HP was investigated by urease and histology, exclusion and inclusion factors were applied and patients were divided into two groups: the first grouping the ones without histological diagnosis of Barrett’s esophagus (235-63%) and the second with it (138-37%). Results: There was no significant difference between HP and non-HP patients in relation to the probability of having intestinal metaplasia (p=0.587). When related to the general group, there was an inverse association between the bacterium and the columnar epithelia in the distal esophagus. Age (p=0.031), gender (p=0.013) and HP (p=0.613) when related together to intestinal metaplasia showed no significant relation. In isolation, when related to age and gender, regardless of HP, results confirmed that patients in more advanced age and women present a higher incidence of intestinal metaplasia. Conclusion: There is an inverse relation between HP and the areas of columnar epithelization in the distal esophagus, regardless of the presence or absence of intestinal metaplasia. Age and gender, regardless of HP, showed higher prevalence in women and in older the number of cases with intestinal metaplasia in the distal esophagus.
Collapse
Affiliation(s)
- Matheus Degiovani
- Postgraduate Program in Principles of Surgery, Mackenzie Evangelical School of Medicine - Paraná, Curitiba, PR, Brazil.,Digestive Endoscopy Service, 9 de Julho Hospital, São Paulo, SP, Brazil
| | | | - Nicolau Gregori Czeczko
- Postgraduate Program in Principles of Surgery, Mackenzie Evangelical School of Medicine - Paraná, Curitiba, PR, Brazil
| | - Artur Adolfo Parada
- Postgraduate Program in Principles of Surgery, Mackenzie Evangelical School of Medicine - Paraná, Curitiba, PR, Brazil.,Digestive Endoscopy Service, 9 de Julho Hospital, São Paulo, SP, Brazil
| | - Juliana de Andrade Fronchetti
- Postgraduate Program in Principles of Surgery, Mackenzie Evangelical School of Medicine - Paraná, Curitiba, PR, Brazil
| | - Osvaldo Malafaia
- Postgraduate Program in Principles of Surgery, Mackenzie Evangelical School of Medicine - Paraná, Curitiba, PR, Brazil
| |
Collapse
|
14
|
Abstract
Helicobacter pylori is a Gram-negative bacterium that infects the gastric epithelia of its human host. Everyone who is colonized with these pathogenic bacteria can develop gastric inflammation, termed gastritis. Additionally, a small proportion of colonized people develop more adverse outcomes, including gastric ulcer disease, gastric adenocarcinoma, or gastric mucosa-associated lymphoid tissue lymphoma. The development of these adverse outcomes is dependent on the establishment of a chronic inflammatory response. The development and control of this chronic inflammatory response are significantly impacted by CD4+ T helper cell activity. Noteworthy, T helper 17 (Th17) cells, a proinflammatory subset of CD4+ T cells, produce several proinflammatory cytokines that activate innate immune cell antimicrobial activity, drive a pathogenic immune response, regulate B cell responses, and participate in wound healing. Therefore, this review was written to take an intricate look at the involvement of Th17 cells and their affiliated cytokines (interleukin-17A [IL-17A], IL-17F, IL-21, IL-22, and IL-26) in regulating the immune response to H. pylori colonization and carcinogenesis.
Collapse
|
15
|
Cheng M, Cao L, Ning K. Microbiome Big-Data Mining and Applications Using Single-Cell Technologies and Metagenomics Approaches Toward Precision Medicine. Front Genet 2019; 10:972. [PMID: 31649735 PMCID: PMC6794611 DOI: 10.3389/fgene.2019.00972] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 09/12/2019] [Indexed: 12/20/2022] Open
Abstract
With the development of high-throughput sequencing technologies as well as various bioinformatics analytic tools, microbiome is not a “microbial dark matter” anymore. In this review, we first summarized the current analytical strategies used for big-data mining such as single-cell sequencing and metagenomics. We then provided insights into the integration of these strategies, showing significant advantages in fully describing microbiome from multiple aspects. Moreover, we discussed the correlation between gut microbiome with host organs and diseases, confirming the importance of big-data mining in clinical practices. We finally proposed new ideas about the trend of big-data mining in microbiome using multi-omics approaches and single-cell sequencing. The integration of multi-omics approaches and single-cell sequencing can provide full understanding of microbiome at both macroscopic level and microscopic level, thus contributing to precision medicine.
Collapse
Affiliation(s)
- Mingyue Cheng
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Le Cao
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Kang Ning
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
16
|
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
|
17
|
Westerik N, Reid G, Sybesma W, Kort R. The Probiotic Lactobacillus rhamnosus for Alleviation of Helicobacter pylori-Associated Gastric Pathology in East Africa. Front Microbiol 2018; 9:1873. [PMID: 30154777 PMCID: PMC6102400 DOI: 10.3389/fmicb.2018.01873] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 07/25/2018] [Indexed: 12/16/2022] Open
Abstract
The probiotic Lactobacillus rhamnosus GG (LGG) can play a role in establishing a harmless relationship with Helicobacter pylori and reduce gastric pathology in East African populations. H. pylori has the ability to inhabit the surface of the mucous layer of the human stomach and duodenum. In the developing world, an estimated 51% of the population is carrier of H. pylori, while in some Western countries these numbers dropped below 20%, which is probably associated with improved sanitation and smaller family sizes. Colonization by H. pylori can be followed by inflammation of the gastric mucus layer, and is a risk factor in the development of atrophic gastritis, peptic ulcers and gastric cancer. Notwithstanding the higher prevalence of H. pylori carriers in developing countries, no equal overall increase in gastric pathology is found. This has been attributed to a less pro-inflammatory immune response to H. pylori in African compared to Caucasian populations. In addition, a relatively low exposure to other risk factors in certain African populations may play a role, including the use of non-steroidal anti-inflammatory drugs, smoking, and diets without certain protective factors. A novel approach to the reduction of H. pylori associated gastric pathology is found in the administration of the probiotic bacterium Lactobacillus rhamnosus yoba 2012 (LRY), the generic variant of LGG. This gastro-intestinal isolate inhibits H. pylori by competition for substrate and binding sites as well as production of antimicrobial compounds such as lactic acid. In addition, it attenuates the host's H. pylori-induced apoptosis and inflammation responses and stimulates angiogenesis in the gastric and duodenal epithelium. The probiotic LRY is not able to eradicate H. pylori completely, but its co-supplementation in antibiotic eradication therapy has been shown to relieve side effects of this therapy. In Uganda, unlike other African countries, gastric pathology is relatively common, presumably resulting from the lack of dietary protective factors in the traditional diet. Supplementation with LRY through local production of probiotic yogurt, could be a solution to establish a harmless relationship with H. pylori and reduce gastric pathology and subsequent eradication therapy treatment.
Collapse
Affiliation(s)
- Nieke Westerik
- Department of Molecular Cell Biology, VU University Amsterdam, Amsterdam, Netherlands
- Yoba for Life Foundation, Amsterdam, Netherlands
| | - Gregor Reid
- Canadian R&D Centre for Human Microbiome and Probiotics, Lawson Health Research Institute, London, ON, Canada
- Department of Microbiology and Immunology, and Surgery, Western University, London, ON, Canada
| | | | - Remco Kort
- Department of Molecular Cell Biology, VU University Amsterdam, Amsterdam, Netherlands
- Yoba for Life Foundation, Amsterdam, Netherlands
- Department of Microbiology and Systems Biology, Netherlands Organization for Applied Scientific Research, Zeist, Netherlands
- ARTIS-Micropia, Amsterdam, Netherlands
| |
Collapse
|
18
|
Erőss B, Farkas N, Vincze Á, Tinusz B, Szapáry L, Garami A, Balaskó M, Sarlós P, Czopf L, Alizadeh H, Rakonczay Z, Habon T, Hegyi P. Helicobacter pylori infection reduces the risk of Barrett's esophagus: A meta-analysis and systematic review. Helicobacter 2018; 23:e12504. [PMID: 29938864 PMCID: PMC6055671 DOI: 10.1111/hel.12504] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION The prevalence of Helicobacter pylori infection (HPI) has been decreasing in developed countries, with an increasing prevalence of Barrett's esophagus (BE) and esophageal adenocarcinoma (EAC) at the same time. The aim of our meta-analysis was to quantify the risk of BE in the context of HPI. METHODS A systematic search was conducted in 3 databases for studies on BE with data on prevalence of HPI from inception until December 2016. Odds ratios for BE in HPI were calculated by the random effects model with subgroup analyses for geographical location, presence of dysplasia in BE, and length of the BE segment. RESULTS Seventy-two studies were included in the meta-analysis, including 84 717 BE cases and 390 749 controls. The overall analysis showed that HPI reduces the risk of BE; OR = 0.68 (95% CI: 0.58-0.79, P < .001). Subgroup analyses revealed risk reduction in Asia OR = 0.53 (95% CI: 0.33-0.84, P = .007), Australia OR = 0.56 (95% CI: 0.39-0.80, P = .002), Europe OR = 0.77 (95% CI: 0.60-0.98, P = .035), and North-America OR = 0.59 (95% CI: 0.47-0.74, P < .001). The risk was significantly reduced for dysplastic BE, OR = 0.37 (95% CI: 0.26-0.51, P < .001) for non-dysplastic BE, OR = 0.51 (95% CI: 0.35-0.75, P = .001), and for long segment BE, OR = 0.25 (95% CI: 0.11-0.59, P = .001) in case of HPI. CONCLUSIONS This extensive meta-analysis provides additional evidence that HPI is associated with reduced risk of BE. Subgroup analyses confirmed that this risk reduction is independent of geographical location. HPI is associated with significantly lower risk of dysplastic, non-dysplastic, and long segment BE.
Collapse
Affiliation(s)
- Bálint Erőss
- Institute for Translational MedicineMedical SchoolUniversity of PécsPécsHungary
| | - Nelli Farkas
- Institute of BioanalysisMedical SchoolUniversity of PécsPécsHungary
| | - Áron Vincze
- Department of GastroenterologyFirst Department of MedicineMedical SchoolUniversity of PécsPécsHungary
| | - Benedek Tinusz
- Institute for Translational MedicineMedical SchoolUniversity of PécsPécsHungary
| | - László Szapáry
- Institute for Translational MedicineMedical SchoolUniversity of PécsPécsHungary
| | - András Garami
- Institute for Translational MedicineMedical SchoolUniversity of PécsPécsHungary
| | - Márta Balaskó
- Institute for Translational MedicineMedical SchoolUniversity of PécsPécsHungary
| | - Patrícia Sarlós
- Department of GastroenterologyFirst Department of MedicineMedical SchoolUniversity of PécsPécsHungary
| | - László Czopf
- Department of CardiologyFirst Department of MedicineMedical SchoolUniversity of PécsPécsHungary
| | - Hussain Alizadeh
- Department of HematologyFirst Department of MedicineMedical SchoolUniversity of PécsPécsHungary
| | - Zoltán Rakonczay
- Department of PathophysiologyMedical SchoolUniversity of SzegedSzegedHungary
| | - Tamás Habon
- Department of CardiologyFirst Department of MedicineMedical SchoolUniversity of PécsPécsHungary
| | - Péter Hegyi
- Institute for Translational MedicineMedical SchoolUniversity of PécsPécsHungary
| |
Collapse
|
19
|
Manabe N, Haruma K. Pathophysiology and Acid Production Different in Asians: Different from the Western People? THE RISE OF ACID REFLUX IN ASIA 2018:37-49. [DOI: 10.1007/978-81-322-0846-4_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
20
|
Abstract
Answer questions and earn CME/CNE The human body harbors enormous numbers of microbiota that influence cancer susceptibility, in part through their prodigious metabolic capacity and their profound influence on immune cell function. Microbial pathogens drive tumorigenesis in 15% to 20% of cancer cases. Even larger numbers of malignancies are associated with an altered composition of commensal microbiota (dysbiosis) based on microbiome studies using metagenomic sequencing. Although association studies cannot distinguish whether changes in microbiota are causes or effects of cancer, a causative role is supported by rigorously controlled preclinical studies using gnotobiotic mouse models colonized with one or more specific bacteria. These studies demonstrate that microbiota can alter cancer susceptibility and progression by diverse mechanisms, such as modulating inflammation, inducing DNA damage, and producing metabolites involved in oncogenesis or tumor suppression. Evidence is emerging that microbiota can be manipulated for improving cancer treatment. By incorporating probiotics as adjuvants for checkpoint immunotherapy or by designing small molecules that target microbial enzymes, microbiota can be harnessed to improve cancer care. CA Cancer J Clin 2017;67:326-344. © 2017 American Cancer Society.
Collapse
Affiliation(s)
- Aadra P. Bhatt
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Matthew R. Redinbo
- Department of Chemistry, University of North Carolina, Chapel Hill, NC 27599, USA
- Department of Biochemistry & Biophysics, Microbiology & Immunology, and the Integrated Program for Biological and Genome Sciences, University of North Carolina, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Scott J. Bultman
- Department of Genetics, University of North Carolina, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA
| |
Collapse
|
21
|
Lupu VV, Ignat A, Ciubotariu G, Ciubară A, Moscalu M, Burlea M. Helicobacter pylori infection and gastroesophageal reflux in children. Dis Esophagus 2016; 29:1007-1012. [PMID: 26455913 DOI: 10.1111/dote.12429] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Some studies suggest that Helicobacter pylori (H. pylori) infection would be a protective factor for the gastroesophageal reflux. The aim of this study was to explore this fact. A group of 72 children, admitted in a pediatric gastroenterology regional center in Northeast Romania, diagnosed with gastroesophageal reflux by 24-hour continuous esophageal pH monitoring (results were interpreted using the Boix-Ochoa score), underwent upper endoscopy with gastric biopsy to detect the presence of H. pylori by the rapid urease testing and for bacteriological and histologic examination. 19 children (26.39%) had H. pylori infection, while 53 (73.61%) did not. The grade of esophagitis was classified according to the Los Angeles classification system. Out of 47 children with esophagitis A, 16 (34.04%) had H. pylori infection, while out of the 25 children with esophagitis B, only 3 (12%) had H. pylori infection, with statistic significance (χ2 = 54.69, P << 0.05, 95% confidence interval [CI]). Regarding the value of the Boix-Ochoa score, it appears that the presence of the H. pylori determines lower pH-metry scores (F = 8.13, P = 0.0015, 95% CI). The presence of the H. pylori was not an important factor in the gastroesophageal reflux. On the other hand its relationship with esophagitis appears to be inverse ratio. The fact that the H. pylori presence is statistically greater in the grade A esophagitis could confirm the hypothesis that the bacteria would slow down the development of the esophagitis.
Collapse
Affiliation(s)
- V V Lupu
- Pediatrics Department, University of Medicine and Pharmacy 'Gr. T. Popa', Iasi, Romania
| | - A Ignat
- Pediatrics Department, University of Medicine and Pharmacy 'Gr. T. Popa', Iasi, Romania
| | - G Ciubotariu
- Pediatrics Department, University of Medicine and Pharmacy 'Gr. T. Popa', Iasi, Romania
| | - A Ciubară
- Psychiatry Department, University of Medicine and Pharmacy 'Gr. T. Popa', Iasi, Romania
| | - M Moscalu
- Medical Informatics and Biostatistics Department, University of Medicine and Pharmacy 'Gr. T. Popa', Iasi, Romania
| | - M Burlea
- Pediatrics Department, University of Medicine and Pharmacy 'Gr. T. Popa', Iasi, Romania
| |
Collapse
|
22
|
Ribeiro ICS, Kubrusly LF, Nassif PAN, Ribeiro PFS, Veras RDO, Neppel A. RELATIONSHIP BETWEEN THE PRESENCE OF HELICOBACTER PYLORI WITH INFLAMMATORY ENDOSCOPIC CHANGES IN GASTRODUODENAL MUCOSA. ABCD-ARQUIVOS BRASILEIROS DE CIRURGIA DIGESTIVA 2016; 29:142-145. [PMID: 27759774 PMCID: PMC5074662 DOI: 10.1590/0102-6720201600030004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 04/14/2016] [Indexed: 01/16/2023]
Abstract
Background: The influence of Helicobacter pylori (HP) in inflammatory disorders of the
digestive mucosa has been the subject of several studies since socioeconomic,
personal and environmental factors were implicated in the bacteria transmission.
Aim: To correlate the inflammatory endoscopic findings with HP infection and the onset
of mucosal diseases mucous of the upper digestive tract. Method: Comparative observational study, in which were collected data from 2247 patients
who underwent upper endoscopy and biopsies for HP with urease test. The patients
were divided into two groups: HP+ and HP- (control) in which endoscopic findings
were observed for the following changes: esophagitis, esophageal ulcer, gastritis,
erosive gastritis, gastric ulcer, bulboduodenitis, bulbar ulcer and without
disease. Results: As for esophagitis, there was little disparity in the distribution favorable to
HP+ group (HP+ =67.11% and HP- =69.89%) and esophageal ulcer (HP+ =0% and HP- =0,
21%). Gastritis was favorable to HP- group (HP+ =78.34% and HP- =73.63%), as well
as erosive gastritis (HP+ = 67,11% and HP- = 64,55%), in bulboduodenitis (HP+
=1,87% and HP- 1,23%), in gastric ulcer (HP+ =2,14% and HP- =2,03%) and in the
absence of alterations in the HP+ group (4.81%) with the HP- control group
(6,30%), in which there was little disproportion in favor of HP- group, but
without statistical significance. As for the bulbar ulcer (HP +=10.16% and HP-
=4.48%), there was statistically significant (p=0.00001). Conclusion: There is no difference between HP+ and HP- groups in inflammatory changes in
endoscopic gastroduodenal mucosa, except for the relationship between HP and
bulbar ulcer.
Collapse
Affiliation(s)
- Irma Cláudia Saboya Ribeiro
- Postgraduate Program in Principles of Surgery, Evangelic Faculty of Paraná/University Evangelic Hospital of Curitiba/Medical Research Institute, Curitiba, PR, Brazil.,Gastrointestinal Endoscopy Service, 9 of July Hospital, São Paulo, SP, Brazil
| | - Luiz Fernandao Kubrusly
- Postgraduate Program in Principles of Surgery, Evangelic Faculty of Paraná/University Evangelic Hospital of Curitiba/Medical Research Institute, Curitiba, PR, Brazil
| | - Paulo Afonso Nunes Nassif
- Postgraduate Program in Principles of Surgery, Evangelic Faculty of Paraná/University Evangelic Hospital of Curitiba/Medical Research Institute, Curitiba, PR, Brazil
| | | | - Rodrigo de Oliveira Veras
- Postgraduate Program in Principles of Surgery, Evangelic Faculty of Paraná/University Evangelic Hospital of Curitiba/Medical Research Institute, Curitiba, PR, Brazil
| | - Aline Neppel
- Postgraduate Program in Principles of Surgery, Evangelic Faculty of Paraná/University Evangelic Hospital of Curitiba/Medical Research Institute, Curitiba, PR, Brazil
| |
Collapse
|
23
|
Abstract
BACKGROUND Infection with Helicobacter pylori is associated with severe digestive diseases including chronic gastritis, peptic ulcer disease, and gastric cancer. Successful eradication of this common gastric pathogen in individual patients is known to prevent the occurrence of peptic ulcer disease and gastric cancer. DISCUSSION With half of the world's population being infected with H, pylori and only few antibiotics result in an effective eradication, a successful antibiotic driven worldwide eradication program seems unlikely. In addition, H. pylori eradication is not always beneficial as it has been described that eradication can be associated with an increased frequency of other disorders such as pediatric asthma, inflammatory bowel diseases and Barrett's Esophagus. We have to accept that eradication of this infection is a two-edged sword that is both useful and harmful and we should therefore focus our H. pylori eradication policy toward selectively identify and destroy only the virulent strains. CONCLUSION In order to still be able to effectively treat H. pylori infections in the future we need an alternative diagnostic/treatment algorithm. This would involve a shift towards more precise and enhanced disease predicting diagnosis that tries to identify patients with chance of developing severe diseases such as gastric cancer, rather than the current regime that is geared towards find and destroy all H. pylori.
Collapse
Affiliation(s)
| | - Johannes G Kusters
- Department of Medical Microbiology, University Medical Center Utrecht, Heidelberglaan 100, Utrecht, 3584 CX, The Netherlands.
| |
Collapse
|
24
|
The socioeconomic profile of a Barrett's oesophagus cohort assessed by the 2010 Index of Multiple Deprivation. Eur J Gastroenterol Hepatol 2016; 28:199-204. [PMID: 26571328 DOI: 10.1097/meg.0000000000000523] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
BACKGROUND Several reports have described the relationship between socioeconomic status and oesophageal adenocarcinoma but only one with its precursor condition, Barrett's oesophagus. We therefore investigated such an association. PATIENTS The majority (88%) of patients diagnosed with Barrett's at Rotherham District General Hospital between 28 April 1978 and 31 August 2012 consented to inclusion in the UK Barrett's Oesophagus Registry. Those residing within Rotherham form the basis of this study. METHODS We assessed socioeconomic status using the Index of Multiple Deprivation 2010 scores which can be assigned to every English postcode. The scores for the whole of England were divided into five equal groups; those of the 6257 postcodes within Rotherham (including those of Barrett's patients) were compared against the national quintile relevant to their score. We examined the ratio of observed against expected numbers of Barrett's in each quintile before and since 2001, the median year of diagnosis. RESULTS The study group comprised 1076 patients with Barrett's oesophagus. Before 2001 their distribution across the deprivation quintiles was similar to that expected. Since then it has changed significantly, with 37% more Barrett's patients than expected among the two least deprived quintiles, but 11% fewer than expected in the larger population comprising the two most deprived quintiles (P=0.0001). There was no significant difference in the distribution of sex (P=0.27), nor the mean age at diagnosis between the two time periods (P=0.92). CONCLUSION Since 2001, there has been a major change in the distribution of Barrett's in relation to socioeconomic status, measured by the Index of Multiple Deprivation.
Collapse
|
25
|
|
26
|
Kim J. Gastroesophageal Reflux Disease. HELICOBACTER PYLORI 2016:317-321. [DOI: 10.1007/978-981-287-706-2_29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
27
|
Kronsteiner B, Bassaganya-Riera J, Philipson C, Viladomiu M, Carbo A, Abedi V, Hontecillas R. Systems-wide analyses of mucosal immune responses to Helicobacter pylori at the interface between pathogenicity and symbiosis. Gut Microbes 2016; 7:3-21. [PMID: 26939848 PMCID: PMC4856448 DOI: 10.1080/19490976.2015.1116673] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 10/29/2015] [Accepted: 10/31/2015] [Indexed: 02/08/2023] Open
Abstract
Helicobacter pylori is the dominant member of the gastric microbiota in over half of the human population of which 5-15% develop gastritis or gastric malignancies. Immune responses to H. pylori are characterized by mixed T helper cell, cytotoxic T cell and NK cell responses. The presence of Tregs is essential for the control of gastritis and together with regulatory CX3CR1+ mononuclear phagocytes and immune-evasion strategies they enable life-long persistence of H. pylori. This H. pylori-induced regulatory environment might contribute to its cross-protective effect in inflammatory bowel disease and obesity. Here we review host-microbe interactions, the development of pro- and anti-inflammatory immune responses and how the latter contribute to H. pylori's role as beneficial member of the gut microbiota. Furthermore, we present the integration of existing and new data into a computational/mathematical model and its use for the investigation of immunological mechanisms underlying initiation, progression and outcomes of H. pylori infection.
Collapse
Affiliation(s)
- Barbara Kronsteiner
- Nutritional Immunology and Molecular Medicine Laboratory and Center for Modeling Immunity to Enteric Pathogens; Virginia Bioinformatics Institute; Virginia Tech; Blacksburg, VA, USA
| | - Josep Bassaganya-Riera
- Nutritional Immunology and Molecular Medicine Laboratory and Center for Modeling Immunity to Enteric Pathogens; Virginia Bioinformatics Institute; Virginia Tech; Blacksburg, VA, USA
| | | | - Monica Viladomiu
- Nutritional Immunology and Molecular Medicine Laboratory and Center for Modeling Immunity to Enteric Pathogens; Virginia Bioinformatics Institute; Virginia Tech; Blacksburg, VA, USA
| | | | - Vida Abedi
- Nutritional Immunology and Molecular Medicine Laboratory and Center for Modeling Immunity to Enteric Pathogens; Virginia Bioinformatics Institute; Virginia Tech; Blacksburg, VA, USA
| | - Raquel Hontecillas
- Nutritional Immunology and Molecular Medicine Laboratory and Center for Modeling Immunity to Enteric Pathogens; Virginia Bioinformatics Institute; Virginia Tech; Blacksburg, VA, USA
| |
Collapse
|
28
|
Abdel Jalil AA, Katzka DA, Castell DO. Approach to the patient with dysphagia. Am J Med 2015; 128:1138.e17-23. [PMID: 26007674 DOI: 10.1016/j.amjmed.2015.04.026] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 04/15/2015] [Accepted: 04/16/2015] [Indexed: 12/17/2022]
Abstract
Dysphagia is a fascinating symptom. It is ostensibly simple when defined by trouble swallowing, yet its subtleties in deciphering and its variations in pathophysiology almost mandate a thorough knowledge of medicine itself. With patience and careful questioning, a multitude of various disorders may be suggested before an objective test is performed. Indeed, the ability to diligently and comprehensively explore the symptom of dysphagia is not only rewarding but also a real test for a physician who prides himself or herself on good history taking.
Collapse
Affiliation(s)
- Ala' A Abdel Jalil
- Esophageal Disorders Program, Division of Gastroenterology and Hepatology, Medical University of South Carolina, Charleston.
| | - David A Katzka
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minn
| | - Donald O Castell
- Esophageal Disorders Program, Division of Gastroenterology and Hepatology, Medical University of South Carolina, Charleston
| |
Collapse
|
29
|
Philipson CW, Bassaganya-Riera J, Viladomiu M, Kronsteiner B, Abedi V, Hoops S, Michalak P, Kang L, Girardin SE, Hontecillas R. Modeling the Regulatory Mechanisms by Which NLRX1 Modulates Innate Immune Responses to Helicobacter pylori Infection. PLoS One 2015; 10:e0137839. [PMID: 26367386 PMCID: PMC4569576 DOI: 10.1371/journal.pone.0137839] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 08/22/2015] [Indexed: 12/15/2022] Open
Abstract
Helicobacter pylori colonizes half of the world’s population as the dominant member of the gastric microbiota resulting in a lifelong chronic infection. Host responses toward the bacterium can result in asymptomatic, pathogenic or even favorable health outcomes; however, mechanisms underlying the dual role of H. pylori as a commensal versus pathogenic organism are not well characterized. Recent evidence suggests mononuclear phagocytes are largely involved in shaping dominant immunity during infection mediating the balance between host tolerance and succumbing to overt disease. We combined computational modeling, bioinformatics and experimental validation in order to investigate interactions between macrophages and intracellular H. pylori. Global transcriptomic analysis on bone marrow-derived macrophages (BMDM) in a gentamycin protection assay at six time points unveiled the presence of three sequential host response waves: an early transient regulatory gene module followed by sustained and late effector responses. Kinetic behaviors of pattern recognition receptors (PRRs) are linked to differential expression of spatiotemporal response waves and function to induce effector immunity through extracellular and intracellular detection of H. pylori. We report that bacterial interaction with the host intracellular environment caused significant suppression of regulatory NLRC3 and NLRX1 in a pattern inverse to early regulatory responses. To further delineate complex immune responses and pathway crosstalk between effector and regulatory PRRs, we built a computational model calibrated using time-series RNAseq data. Our validated computational hypotheses are that: 1) NLRX1 expression regulates bacterial burden in macrophages; and 2) early host response cytokines down-regulate NLRX1 expression through a negative feedback circuit. This paper applies modeling approaches to characterize the regulatory role of NLRX1 in mechanisms of host tolerance employed by macrophages to respond to and/or to co-exist with intracellular H. pylori.
Collapse
Affiliation(s)
- Casandra W. Philipson
- Center for Modeling Immunity to Enteric Pathogens, Virginia Bioinformatics Institute at Virginia Tech, Blacksburg, VA, United States of America
- Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute at Virginia Tech, Blacksburg, VA, United States of America
| | - Josep Bassaganya-Riera
- Center for Modeling Immunity to Enteric Pathogens, Virginia Bioinformatics Institute at Virginia Tech, Blacksburg, VA, United States of America
- Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute at Virginia Tech, Blacksburg, VA, United States of America
- Virginia-Maryland Regional College of Veterinary Medicine, Blacksburg, VA, United States of America
| | - Monica Viladomiu
- Center for Modeling Immunity to Enteric Pathogens, Virginia Bioinformatics Institute at Virginia Tech, Blacksburg, VA, United States of America
- Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute at Virginia Tech, Blacksburg, VA, United States of America
| | - Barbara Kronsteiner
- Center for Modeling Immunity to Enteric Pathogens, Virginia Bioinformatics Institute at Virginia Tech, Blacksburg, VA, United States of America
- Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute at Virginia Tech, Blacksburg, VA, United States of America
| | - Vida Abedi
- Center for Modeling Immunity to Enteric Pathogens, Virginia Bioinformatics Institute at Virginia Tech, Blacksburg, VA, United States of America
- Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute at Virginia Tech, Blacksburg, VA, United States of America
| | - Stefan Hoops
- Center for Modeling Immunity to Enteric Pathogens, Virginia Bioinformatics Institute at Virginia Tech, Blacksburg, VA, United States of America
- Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute at Virginia Tech, Blacksburg, VA, United States of America
| | - Pawel Michalak
- Center for Modeling Immunity to Enteric Pathogens, Virginia Bioinformatics Institute at Virginia Tech, Blacksburg, VA, United States of America
| | - Lin Kang
- Center for Modeling Immunity to Enteric Pathogens, Virginia Bioinformatics Institute at Virginia Tech, Blacksburg, VA, United States of America
| | - Stephen E. Girardin
- Laboratory of Medicine & Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Raquel Hontecillas
- Center for Modeling Immunity to Enteric Pathogens, Virginia Bioinformatics Institute at Virginia Tech, Blacksburg, VA, United States of America
- Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute at Virginia Tech, Blacksburg, VA, United States of America
- * E-mail:
| |
Collapse
|
30
|
Runge TM, Abrams JA, Shaheen NJ. Epidemiology of Barrett's Esophagus and Esophageal Adenocarcinoma. Gastroenterol Clin North Am 2015; 44:203-31. [PMID: 26021191 PMCID: PMC4449458 DOI: 10.1016/j.gtc.2015.02.001] [Citation(s) in RCA: 154] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Barrett's esophagus (BE) is the precursor to esophageal adenocarcinoma (EAC), a disease with increasing burden in the Western world, especially in white men. Risk factors for BE include obesity, tobacco smoking, and gastroesophageal reflux disease (GERD). EAC is the most common form of esophageal cancer in the United States. Risk factors include GERD, tobacco smoking, and obesity, whereas nonsteroidal antiinflammatory drugs and statins may be protective. Factors predicting progression from nondysplastic BE to EAC include dysplastic changes on esophageal histology and length of the involved BE segment. Biomarkers have shown promise, but none are approved for clinical use.
Collapse
Affiliation(s)
- Thomas M. Runge
- University of North Carolina at Chapel Hill, Center for Esophageal Diseases and Swallowing, Division of Gastroenterology and Hepatology, Chapel Hill, NC
| | - Julian A. Abrams
- Division of Digestive and Liver Diseases, Columbia University Medical Center, New York, NY
| | - Nicholas J. Shaheen
- University of North Carolina at Chapel Hill, Center for Esophageal Diseases and Swallowing, Division of Gastroenterology and Hepatology, Chapel Hill, NC
| |
Collapse
|
31
|
Mukaisho KI, Nakayama T, Hagiwara T, Hattori T, Sugihara H. Two distinct etiologies of gastric cardia adenocarcinoma: interactions among pH, Helicobacter pylori, and bile acids. Front Microbiol 2015; 6:412. [PMID: 26029176 PMCID: PMC4426758 DOI: 10.3389/fmicb.2015.00412] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 04/20/2015] [Indexed: 12/12/2022] Open
Abstract
Gastric cancer can be classified as cardia and non-cardia subtypes according to the anatomic site. Although the gastric cancer incidence has decreased steadily in several countries over the past 50 years, the incidence of cardia cancers and esophageal adenocarcinoma (EAC) continue to increase. The etiological factors involved in the development of both cardia cancers and EACs are associated with high animal fat intake, which causes severe obesity. Central obesity plays roles in cardiac-type mucosa lengthening and partial hiatus hernia development. There are two distinct etiologies of cardia cancer subtypes: one associated with gastroesophageal reflux (GER), which predominantly occurs in patients without Helicobacter pylori (H. pylori) infection and resembles EAC, and the other associated with H. pylori atrophic gastritis, which resembles non-cardia cancer. The former can be developed in the environment of high volume duodenal content reflux, including bile acids and a higher acid production in H. pylori-negative patients. N-nitroso compounds, which are generated from the refluxate that includes a large volume of bile acids and are stabilized in the stomach (which has high levels of gastric acid), play a pivotal role in this carcinogenesis. The latter can be associated with the changing colonization of H. pylori from the distal to the proximal stomach with atrophic gastritis because a high concentration of soluble bile acids in an environment of low acid production is likely to act as a bactericide or chemorepellent for H. pylori in the distal stomach. The manuscript introduces new insights in causative factors of adenocarcinoma of the cardia about the role of bile acids in gastro-esophageal refluxate based upon robust evidences supporting interactions among pH, H. pylori, and bile acids.
Collapse
Affiliation(s)
- Ken-ichi Mukaisho
- Division of Molecular Diagnostic Pathology, Department of Pathology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | | | | | | | | |
Collapse
|
32
|
Kodaman N, Sobota RS, Mera R, Schneider BG, Williams SM. Disrupted human-pathogen co-evolution: a model for disease. Front Genet 2014; 5:290. [PMID: 25202324 PMCID: PMC4142859 DOI: 10.3389/fgene.2014.00290] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 08/05/2014] [Indexed: 02/06/2023] Open
Abstract
A major goal in infectious disease research is to identify the human and pathogenic genetic variants that explain differences in microbial pathogenesis. However, neither pathogenic strain nor human genetic variation in isolation has proven adequate to explain the heterogeneity of disease pathology. We suggest that disrupted co-evolution between a pathogen and its human host can explain variation in disease outcomes, and that genome-by-genome interactions should therefore be incorporated into genetic models of disease caused by infectious agents. Genetic epidemiological studies that fail to take both the pathogen and host into account can lead to false and misleading conclusions about disease etiology. We discuss our model in the context of three pathogens, Helicobacter pylori, Mycobacterium tuberculosis and human papillomavirus, and generalize the conditions under which it may be applicable.
Collapse
Affiliation(s)
- Nuri Kodaman
- Department of Genetics, Geisel School of Medicine, Dartmouth College Hanover, NH, USA ; Department of Molecular Physiology and Biophysics, Center for Human Genetics Research, Vanderbilt University Medical Center Nashville, TN, USA
| | - Rafal S Sobota
- Department of Genetics, Geisel School of Medicine, Dartmouth College Hanover, NH, USA ; Department of Molecular Physiology and Biophysics, Center for Human Genetics Research, Vanderbilt University Medical Center Nashville, TN, USA
| | - Robertino Mera
- Division of Gastroenterology, Department of Medicine, Vanderbilt University Medical Center Nashville, TN, USA
| | - Barbara G Schneider
- Division of Gastroenterology, Department of Medicine, Vanderbilt University Medical Center Nashville, TN, USA
| | - Scott M Williams
- Department of Genetics, Geisel School of Medicine, Dartmouth College Hanover, NH, USA
| |
Collapse
|
33
|
Carbo A, Olivares-Villagómez D, Hontecillas R, Bassaganya-Riera J, Chaturvedi R, Piazuelo MB, Delgado A, Washington MK, Wilson KT, Algood HMS. Systems modeling of the role of interleukin-21 in the maintenance of effector CD4+ T cell responses during chronic Helicobacter pylori infection. mBio 2014; 5:e01243-14. [PMID: 25053783 PMCID: PMC4120195 DOI: 10.1128/mbio.01243-14] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 06/25/2014] [Indexed: 01/25/2023] Open
Abstract
The development of gastritis during Helicobacter pylori infection is dependent on an activated adaptive immune response orchestrated by T helper (Th) cells. However, the relative contributions of the Th1 and Th17 subsets to gastritis and control of infection are still under investigation. To investigate the role of interleukin-21 (IL-21) in the gastric mucosa during H. pylori infection, we combined mathematical modeling of CD4(+) T cell differentiation with in vivo mechanistic studies. We infected IL-21-deficient and wild-type mice with H. pylori strain SS1 and assessed colonization, gastric inflammation, cellular infiltration, and cytokine profiles. Chronically H. pylori-infected IL-21-deficient mice had higher H. pylori colonization, significantly less gastritis, and reduced expression of proinflammatory cytokines and chemokines compared to these parameters in infected wild-type littermates. These in vivo data were used to calibrate an H. pylori infection-dependent, CD4(+) T cell-specific computational model, which then described the mechanism by which IL-21 activates the production of interferon gamma (IFN-γ) and IL-17 during chronic H. pylori infection. The model predicted activated expression of T-bet and RORγt and the phosphorylation of STAT3 and STAT1 and suggested a potential role of IL-21 in the modulation of IL-10. Driven by our modeling-derived predictions, we found reduced levels of CD4(+) splenocyte-specific tbx21 and rorc expression, reduced phosphorylation of STAT1 and STAT3, and an increase in CD4(+) T cell-specific IL-10 expression in H. pylori-infected IL-21-deficient mice. Our results indicate that IL-21 regulates Th1 and Th17 effector responses during chronic H. pylori infection in a STAT1- and STAT3-dependent manner, therefore playing a major role controlling H. pylori infection and gastritis. Importance: Helicobacter pylori is the dominant member of the gastric microbiota in more than 50% of the world's population. H. pylori colonization has been implicated in gastritis and gastric cancer, as infection with H. pylori is the single most common risk factor for gastric cancer. Current data suggest that, in addition to bacterial virulence factors, the magnitude and types of immune responses influence the outcome of colonization and chronic infection. This study uses a combined computational and experimental approach to investigate how IL-21, a proinflammatory T cell-derived cytokine, maintains the chronic proinflammatory T cell immune response driving chronic gastritis during H. pylori infection. This research will also provide insight into a myriad of other infectious and immune disorders in which IL-21 is increasingly recognized to play a central role. The use of IL-21-related therapies may provide treatment options for individuals chronically colonized with H. pylori as an alternative to aggressive antibiotics.
Collapse
Affiliation(s)
| | - Danyvid Olivares-Villagómez
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | | | | | - Rupesh Chaturvedi
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - M Blanca Piazuelo
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Alberto Delgado
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - M Kay Washington
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | | | | |
Collapse
|
34
|
Kronsteiner B, Bassaganya-Riera J, Philipson N, Hontecillas R. Novel insights on the role of CD8+ T cells and cytotoxic responses during Helicobacter pylori infection. Gut Microbes 2014; 5:357-62. [PMID: 24755940 PMCID: PMC4153774 DOI: 10.4161/gmic.28899] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Helicobacter pylori chronically persists in 50% of the human population and causes serious gastric and duodenal pathologies in 15% of infected people. Research on the immune response to the infection has mainly focused on the induction of CD4+ T cell responses. Human studies emphasize the potential clinical relevance of CD8+ cytotoxic T lymphocytes, however this cell type has barely been reported in studies employing mouse or gerbil models. Traditionally characterized as an extracellular bacterium, H. pylori has been identified inside epithelial and immune cells. Similarly to other intracellular bacteria, H. pylori infection of macrophages can alter autophagy and phagosome processing. A novel animal model of H. pylori infection demonstrates for the first time the induction of cytotoxic CD8+ T cell responses in pigs and localization of intracellular H. pylori within lymphoid aggregates. Here, we discuss novel mechanisms of host-H. pylori interactions that could lead to the induction of cytotoxic responses.
Collapse
|
35
|
He C, Yang Z, Lu NH. Helicobacter pylori infection and diabetes: Is it a myth or fact? World J Gastroenterol 2014; 20:4607-4617. [PMID: 24782613 PMCID: PMC4000497 DOI: 10.3748/wjg.v20.i16.4607] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 02/10/2014] [Accepted: 03/06/2014] [Indexed: 02/06/2023] Open
Abstract
Helicobacter pylori (H. pylori) is one of the most common human bacterial pathogens, and infection causes a wide array of gastric disorders, including simple gastritis, peptic ulcers and gastric malignancies. Gastrointestinal inflammation caused by H. pylori can influence the absorption of glucose and lipids, which are also abnormal in diabetes mellitus. Type 2 diabetes mellitus (T2DM), formerly known as non-insulin-dependent diabetes mellitus or adult-onset diabetes, is a metabolic disorder that is characterized by high levels of blood glucose resulting from insulin resistance and relative insulin deficiency. It is an emerging pandemic and is rapidly becoming a serious threat to public health. Emerging data now indicate a strong relationship between H. pylori infection and the incidence of T2DM. The mechanisms underlying the pathogenesis of diabetes are complex, involving insulin resistance, chronic inflammation, insulin secretion deficiency as a result of pancreas β-cell dysfunction, glucotoxicity, and lipotoxicity. H. pylori infection is known to be involved in the pathogenesis of insulin resistance, and the growing awareness of its role in diabetes is important for the early detection of glucose dysregulation and prevention of T2DM in high-risk communities. This review probes the possible relationship between H. pylori and diabetes according to epidemiological surveys and discusses putative mechanisms underlying this correlation.
Collapse
|
36
|
Human and Helicobacter pylori coevolution shapes the risk of gastric disease. Proc Natl Acad Sci U S A 2014; 111:1455-60. [PMID: 24474772 DOI: 10.1073/pnas.1318093111] [Citation(s) in RCA: 167] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Helicobacter pylori is the principal cause of gastric cancer, the second leading cause of cancer mortality worldwide. However, H. pylori prevalence generally does not predict cancer incidence. To determine whether coevolution between host and pathogen influences disease risk, we examined the association between the severity of gastric lesions and patterns of genomic variation in matched human and H. pylori samples. Patients were recruited from two geographically distinct Colombian populations with significantly different incidences of gastric cancer, but virtually identical prevalence of H. pylori infection. All H. pylori isolates contained the genetic signatures of multiple ancestries, with an ancestral African cluster predominating in a low-risk, coastal population and a European cluster in a high-risk, mountain population. The human ancestry of the biopsied individuals also varied with geography, with mostly African ancestry in the coastal region (58%), and mostly Amerindian ancestry in the mountain region (67%). The interaction between the host and pathogen ancestries completely accounted for the difference in the severity of gastric lesions in the two regions of Colombia. In particular, African H. pylori ancestry was relatively benign in humans of African ancestry but was deleterious in individuals with substantial Amerindian ancestry. Thus, coevolution likely modulated disease risk, and the disruption of coevolved human and H. pylori genomes can explain the high incidence of gastric disease in the mountain population.
Collapse
|
37
|
Gashi Z, Sherifi F, Shabani R. The prevalence of helicobacter pylori infection in patients with reflux esophagitis - our experience. Med Arch 2013; 67:402-4. [PMID: 25568507 PMCID: PMC4272478 DOI: 10.5455/medarh.2013.67.402-404] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2013] [Accepted: 11/18/2013] [Indexed: 01/08/2023] Open
Abstract
Introduction and aim: The role of Helicobacter pylori in esophageal disease has not been clearly defined. To clarify this issue, we analyzed 120 patients with histologically confirmed esophageal disease. Material and methods: In this prospective study, 120 patients who underwent upper endoscopy examination were included; among them 70 patients with clinically, endoscopically and histologically confirmed GERD, and 50 patients with BE. This investigation was performed in the Clinic of Gastrohepatology in Prishtina, during the period: June 2009–December 2011. Each patient was investigated for H. pylori infection, by performing biopsy for HUT test. Results: In BE group, H. pylori infection was present in 16.0% of patients. In GERD group, H. pylori infection was present in 42.9%, and in patients of the control group, in 52.0% of cases. So, in BE group, the prevalence of H. pylori infection showed less significant difference, compared to the control group (P = 0.003) and in GERD group (P = 0.0035). Between GERD group and the control group there was no significant difference (GERD vs. G control. P = 0.421). Conclusion: The prevalence of H. pylori infection in patients with BE (16%) was lower in comparison with patients with GERD (42.9%) and with control group (p <0.01). The prevalence of H. pylori infection in patients with BE, especially those with LSBE (9.1%) was very low, which indicates a possible protective role of this microorganism.
Collapse
Affiliation(s)
- Zaim Gashi
- Clinic of Gastroenterology, University Clinical Center, Prishtina, Republic of Kosova
| | - Fadil Sherifi
- Clinic of Gastroenterology, University Clinical Center, Prishtina, Republic of Kosova
| | - Ragip Shabani
- Institute of Pathology, University Clinical Center, Prishtina, Republic of Kosova
| |
Collapse
|
38
|
Carbo A, Bassaganya-Riera J, Pedragosa M, Viladomiu M, Marathe M, Eubank S, Wendelsdorf K, Bisset K, Hoops S, Deng X, Alam M, Kronsteiner B, Mei Y, Hontecillas R. Predictive computational modeling of the mucosal immune responses during Helicobacter pylori infection. PLoS One 2013; 8:e73365. [PMID: 24039925 PMCID: PMC3764126 DOI: 10.1371/journal.pone.0073365] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 07/18/2013] [Indexed: 02/06/2023] Open
Abstract
T helper (Th) cells play a major role in the immune response and pathology at the gastric mucosa during Helicobacter pylori infection. There is a limited mechanistic understanding regarding the contributions of CD4+ T cell subsets to gastritis development during H. pylori colonization. We used two computational approaches: ordinary differential equation (ODE)-based and agent-based modeling (ABM) to study the mechanisms underlying cellular immune responses to H. pylori and how CD4+ T cell subsets influenced initiation, progression and outcome of disease. To calibrate the model, in vivo experimentation was performed by infecting C57BL/6 mice intragastrically with H. pylori and assaying immune cell subsets in the stomach and gastric lymph nodes (GLN) on days 0, 7, 14, 30 and 60 post-infection. Our computational model reproduced the dynamics of effector and regulatory pathways in the gastric lamina propria (LP) in silico. Simulation results show the induction of a Th17 response and a dominant Th1 response, together with a regulatory response characterized by high levels of mucosal Treg) cells. We also investigated the potential role of peroxisome proliferator-activated receptor γ (PPARγ) activation on the modulation of host responses to H. pylori by using loss-of-function approaches. Specifically, in silico results showed a predominance of Th1 and Th17 cells in the stomach of the cell-specific PPARγ knockout system when compared to the wild-type simulation. Spatio-temporal, object-oriented ABM approaches suggested similar dynamics in induction of host responses showing analogous T cell distributions to ODE modeling and facilitated tracking lesion formation. In addition, sensitivity analysis predicted a crucial contribution of Th1 and Th17 effector responses as mediators of histopathological changes in the gastric mucosa during chronic stages of infection, which were experimentally validated in mice. These integrated immunoinformatics approaches characterized the induction of mucosal effector and regulatory pathways controlled by PPARγ during H. pylori infection affecting disease outcomes.
Collapse
Affiliation(s)
- Adria Carbo
- Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, United States of America
- Center for Modeling Immunity to Enteric Pathogens Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Josep Bassaganya-Riera
- Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, United States of America
- Center for Modeling Immunity to Enteric Pathogens Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, United States of America
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Mireia Pedragosa
- Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, United States of America
- Center for Modeling Immunity to Enteric Pathogens Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Monica Viladomiu
- Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, United States of America
- Center for Modeling Immunity to Enteric Pathogens Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Madhav Marathe
- Center for Modeling Immunity to Enteric Pathogens Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, United States of America
- Network Dynamics and Simulation Science Laboratory, Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Stephen Eubank
- Center for Modeling Immunity to Enteric Pathogens Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, United States of America
- Network Dynamics and Simulation Science Laboratory, Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Katherine Wendelsdorf
- Center for Modeling Immunity to Enteric Pathogens Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, United States of America
- Network Dynamics and Simulation Science Laboratory, Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Keith Bisset
- Center for Modeling Immunity to Enteric Pathogens Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, United States of America
- Network Dynamics and Simulation Science Laboratory, Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Stefan Hoops
- Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, United States of America
- Center for Modeling Immunity to Enteric Pathogens Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Xinwei Deng
- Center for Modeling Immunity to Enteric Pathogens Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, United States of America
- Department of Statistics, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Maksudul Alam
- Center for Modeling Immunity to Enteric Pathogens Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, United States of America
- Network Dynamics and Simulation Science Laboratory, Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Barbara Kronsteiner
- Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, United States of America
- Center for Modeling Immunity to Enteric Pathogens Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Yongguo Mei
- Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, United States of America
- Center for Modeling Immunity to Enteric Pathogens Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, United States of America
| | - Raquel Hontecillas
- Nutritional Immunology and Molecular Medicine Laboratory, Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, United States of America
- Center for Modeling Immunity to Enteric Pathogens Virginia Bioinformatics Institute, Virginia Tech, Blacksburg, Virginia, United States of America
- * E-mail:
| |
Collapse
|
39
|
Abstract
OBJECTIVE Persistent colonisation by Helicobacter pylori, and especially by cagA-positive strains, has been related to several health outcomes with effects in opposite directions. Thus, it is important to evaluate its influence on total and category-specific mortality. DESIGN We conducted prospective cohort analyses in a nationally representative sample of 9895 participants enrolled in the National Health and Nutrition Examination Survey III to assess the association of H pylori status with all-cause and cause-specific mortality. Analyses for the association of H pylori cagA positivity with mortality were conducted in 7384 subjects with data on H pylori cagA status. RESULTS In older people (> 40.1 years), H pylori was not associated with all-cause mortality (HR 1.00; 95% CI 0.84 to 1.18). There was an inverse association of H pylori status with stroke mortality (HR 0.69; 95% CI 0.44 to 1.08), and the inverse association was stronger for H pylori cagA positivity, with the HR of 0.45 (95% CI 0.27 to 0.76). H pylori was also strongly positively related to gastric cancer mortality. After we adjusted p values using the Benjamini-Hochberg false discovery rate method to account for multiple comparisons, these associations remained, and H pylori status was not related to other outcomes. CONCLUSIONS Our findings suggest that H pylori has a mixed role in human health, but is not a major risk factor for all-cause mortality.
Collapse
Affiliation(s)
- Yu Chen
- Department of Population Health, New York University School of Medicine, , New York, NY 10016, USA.
| | | | | |
Collapse
|
40
|
Arboleda RN, Schneider BG, Bravo LE, Romero-Gallo J, Peek RM, Mera RM, Yepez MC, Campo C, Correa P. Use of the noninvasive entero-test in the detection of Helicobacter pylori in children in an endemic area in Colombia. J Pediatr Gastroenterol Nutr 2013; 57:192-6. [PMID: 23880626 PMCID: PMC3725653 DOI: 10.1097/mpg.0b013e318293e1e1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND AND OBJECTIVE Gastric infection with Helicobacter pylori (H pylori), a strong risk factor for gastric cancer, is highly prevalent in children residing in the Colombian Andes. We aimed to validate the use of the Entero-test to culture and genotype H pylori strains from asymptomatic Colombian children. METHODS Children (ages 10-15 years, n = 110, 80 of which were H pylori positive by the urea breath test [UBT]) were subjected to the Entero-test, and strings were cultured and/or used for DNA extraction for polymerase chain reaction (PCR). These children had been treated for H pylori in 2007. A second population of children (ages 10-15 years, n = 95),which had not been previously treated, was also subjected to the Entero-test. RESULTS Of UBT-positive children in the treated group, 29 of 80 (36%) Entero-test samples were H pylori culture positive; 29 additional string extracts were tested by PCR for the H pylori virulence factors cagA and vacA. PCR from cultures and extracts yielded a sensitivity of 74% and specificity of 87%. In the untreated group, 16 of 94 UBT-positive children (17%) produced Entero-tests that were culture positive. Fifty-eight of 94 (62%) string extracts were PCR positive for cagA and/or vacA. In previously treated children, H pylori strains were more often the less virulent vacA s2 (P = 0.001), m2 (P = 0.006), and i2 genotypes (P = 0.039). CONCLUSIONS The Entero-test may be used as a noninvasive test to detect H pylori in asymptomatic children residing in high-risk areas for gastric cancer. Treatment of H pylori in children was associated with less virulent genotypes.
Collapse
Affiliation(s)
- Richard N Arboleda
- Department of Medicine, Division of Gastroenterology, Vanderbilt University School of Medicine, Nashville, TN 37232 , USA
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Helicobacter pylori infection in a pig model is dominated by Th1 and cytotoxic CD8+ T cell responses. Infect Immun 2013; 81:3803-13. [PMID: 23897614 DOI: 10.1128/iai.00660-13] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Helicobacter pylori infection is the leading cause for peptic ulcer disease and gastric adenocarcinoma. Mucosal T cell responses play an important role in mediating H. pylori-related gastric immunopathology. While induced regulatory T (iTreg) cells are required for chronic colonization without disease, T helper 1 (Th1) effector responses are associated with lower bacterial loads at the expense of gastric pathology. Pigs were inoculated with either H. pylori strain SS1 or J99. Phenotypic and functional changes in peripheral blood mononuclear cell (PBMC) populations were monitored weekly, and mucosal immune responses and bacterial loads were assessed up to 2 months postinfection. Both H. pylori strains elicited a Th1 response characterized by increased percentages of CD4(+)Tbet(+) cells and elevated gamma interferon (IFN-γ) mRNA in PBMCs. A subset of CD8(+) T cells expressing Tbet and CD16 increased following infection. Moreover, a significant increase in perforin and granzyme mRNA expression was observed in PBMCs of infected pigs, indicating a predominant cytotoxic immune response. Infiltration of B cells, myeloid cells, T cells expressing Treg- and Th17-associated transcription factors, and cytotoxic T cells was found in the gastric lamina propria of both infected groups. Interestingly, based on bacterial reisolation data, strain SS1 showed greater capacity to colonize and/or persist in the gastric mucosa than did strain J99. This novel pig model of infection closely mimics human gastric pathology and presents a suitable avenue for studying effector and regulatory responses toward H. pylori described in humans.
Collapse
|
42
|
Oikonomopoulou K, Brinc D, Kyriacou K, Diamandis EP. Infection and cancer: revaluation of the hygiene hypothesis. Clin Cancer Res 2013; 19:2834-41. [PMID: 23536438 DOI: 10.1158/1078-0432.ccr-12-3661] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Several studies have shown that persistent infections and inflammation can favor carcinogenesis. At the same time, certain types of pathogens and antitumor immune responses can decrease the risk of tumorigenesis or lead to cancer regression. Infectious agents and their products can orchestrate a wide range of host immune responses, through which they may positively or negatively modulate cancer development and/or progression. The factors that direct this dichotomous influence of infection-mediated immunity on carcinogenesis are not well understood. Even though not universal, several previous reports have investigated the inverse link of pathogen-induced "benign" inflammation to carcinogenesis and various other pathologies, ranging from autoimmune diseases to allergy and cancer. Several models and ideas are discussed in this review, including the impact of decreased exposure to pathogens, as well as the influence of pathogen load, the timing of infection, and the type of instigated immune response on carcinogenesis. These phenomena should guide future investigations into identifying novel targets within the microbial and host proteome, which will assist in the development of cancer therapeutics and vaccine remedies, analogous to earlier efforts based on helminthic components for the prevention and/or treatment of several pathologies.
Collapse
Affiliation(s)
- Katerina Oikonomopoulou
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
43
|
Abstract
Half of the world's population is infected with Helicobacter pylori and approximately 20% of infected individuals develop overt clinical disease such as ulcers and stomach cancer. Paradoxically, despite its classification as a class I carcinogen, H. pylori has been shown to be protective against development of asthma, allergy, and esophageal disease. Given these conflicting roles for H. pylori, researchers are attempting to define the environmental, host, and pathogen interactions that ultimately result in severe disease in some individuals. From the bacterial perspective, the toxins, CagA and VacA, have each been shown to be polymorphic and to contribute to disease in an allele-dependent manner. Based on the notable advances that have recently been made in the CagA field, herein we review recent studies that have begun to shed light on the role of CagA polymorphism in H. pylori disease. Moreover, we discuss the potential interaction of CagA and VacA as a mediator of gastric disease.
Collapse
|
44
|
Helicobacter pylori colonization ameliorates glucose homeostasis in mice through a PPAR γ-dependent mechanism. PLoS One 2012; 7:e50069. [PMID: 23166823 PMCID: PMC3499487 DOI: 10.1371/journal.pone.0050069] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Accepted: 10/18/2012] [Indexed: 02/06/2023] Open
Abstract
Background There is an inverse secular trend between the incidence of obesity and gastric colonization with Helicobacter pylori, a bacterium that can affect the secretion of gastric hormones that relate to energy homeostasis. H. pylori strains that carry the cag pathogenicity island (PAI) interact more intimately with gastric epithelial cells and trigger more extensive host responses than cag− strains. We hypothesized that gastric colonization with H. pylori strains differing in cag PAI status exert distinct effects on metabolic and inflammatory phenotypes. Methodology/Principal Findings To test this hypothesis, we examined metabolic and inflammatory markers in db/db mice and mice with diet-induced obesity experimentally infected with isogenic forms of H. pylori strain 26695: the cag PAI wild-type and its cag PAI mutant strain 99–305. H. pylori colonization decreased fasting blood glucose levels, increased levels of leptin, improved glucose tolerance, and suppressed weight gain. A response found in both wild-type and mutant H. pylori strain-infected mice included decreased white adipose tissue macrophages (ATM) and increased adipose tissue regulatory T cells (Treg) cells. Gene expression analyses demonstrated upregulation of gastric PPAR γ-responsive genes (i.e., CD36 and FABP4) in H. pylori-infected mice. The loss of PPAR γ in immune and epithelial cells in mice impaired the ability of H. pylori to favorably modulate glucose homeostasis and ATM infiltration during high fat feeding. Conclusions/Significance Gastric infection with some commensal strains of H. pylori ameliorates glucose homeostasis in mice through a PPAR γ-dependent mechanism and modulates macrophage and Treg cell infiltration into the abdominal white adipose tissue.
Collapse
|
45
|
Bohm M, Malik Z, Sebastiano C, Thomas R, Gaughan J, Kelsen S, Richter JE. Mucosal eosinophilia: prevalence and racial/ethnic differences in symptoms and endoscopic findings in adults over 10 years in an urban hospital. J Clin Gastroenterol 2012; 46:567-74. [PMID: 22186744 DOI: 10.1097/mcg.0b013e31823d3305] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Eosinophilic esophagitis is a chronic inflammatory disease with mucosal accumulation of eosinophils. There is a paucity of data among racial/ethnic groups other than white patients. AIM To determine if racial/ethnic differences exist in clinical presentation, endoscopic appearance, and biopsy results in adult patients (age ≥18 y) with mucosal eosinophilia and examine the prevalence of mucosal eosinophilia at an urban hospital over a 10-year period. METHODS Pathology reports searched at Temple University Hospital 2000 to 2009; key words: "eosinophils", "esophagus", and "biopsy". Clinical and endoscopic records reviewed on patients with ≥15 eosinophils/high power field. RESULTS A total of 64 adults (average age, 41 y; 62% male patients; 81% white, 12% black, and 6% Hispanic). White patients were significantly younger (P=0.03). Adult mucosal eosinophilia diagnosis increased by 833% (3 in 2000 to 25 in 2009); black/Hispanic diagnosis increased by 500% (1 in 2000 to 5 in 2009). Solid food dysphagia was more common among white patients (72% vs. 0.33%, P=0.02). Reflux symptoms were more common in black/Hispanic patients (42% vs. 22%, P=0.16). Normal endoscopy (42% vs. 13%, P=0.04) and reflux changes (41% vs. 21%, P=0.16) were more common in black/Hispanic patients. Furrows (42% vs. 8%, P=0.04) and rings (46% vs. 0%, P=0.002) were more common in white patients. Average eosinophil counts did not vary between groups. CONCLUSIONS Mucosal eosinophilia presents with significant differences between racial/ethnic groups in age at onset, symptoms at presentation, and endoscopic features. Differences may reflect different phenotypes of the same disease or separate disease entities.
Collapse
Affiliation(s)
- Matthew Bohm
- Department of Medicine, Temple University School of Medicine, Philadelphia, PA, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Fischbach LA, Nordenstedt H, Kramer JR, Gandhi S, Dick-Onuoha S, Lewis A, El-Serag HB. The association between Barrett's esophagus and Helicobacter pylori infection: a meta-analysis. Helicobacter 2012; 17:163-75. [PMID: 22515353 PMCID: PMC3335759 DOI: 10.1111/j.1523-5378.2011.00931.x] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE The effect of Helicobacter pylori on Barrett's esophagus is poorly understood. We conducted a meta-analysis to summarize the existing literature examining the effect that H. pylori has on Barrett's esophagus. DESIGN We performed a comprehensive search to identify studies pertaining to the association between H. pylori and Barrett's esophagus. We conducted meta-regression analyses to identify sources of variation in the effect of H. pylori on Barrett's esophagus. RESULTS Our analysis included a total of 49 studies that examined the effect of H. pylori on Barrett's esophagus and seven studies that examined the effect of cag A positivity on Barrett's esophagus. Overall, H. pylori, and even more so cag A, tended to be protective for Barrett's esophagus in most studies; however, there was obvious heterogeneity across studies. The effect of H. pylori on Barrett's esophagus varied by geographic location and in the presence of selection and information biases. Only four studies were found without obvious selection and information bias, and these showed a protective effect of H. pylori on Barrett's esophagus (Relative risk = 0.46 [95% CI: 0.35, 0.60]). CONCLUSIONS Estimates for the effect of H. pylori on Barrett's esophagus were heterogeneous across studies. We identified selection and information bias as potential sources of this heterogeneity. Few studies without obvious selection and information bias have been conducted to examine the effect of H. pylori on Barrett's esophagus, but in these, H. pylori infection is associated with a reduced risk of Barrett's esophagus.
Collapse
Affiliation(s)
- Lori A. Fischbach
- Department of Epidemiology, University of North Texas Health Science Center, Fort Worth, TX
| | - Helena Nordenstedt
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, TX,Houston VA Health Services Research & Development Center of Excellence, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX
| | - Jennifer R. Kramer
- Houston VA Health Services Research & Development Center of Excellence, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX,Section of Health Services Research, Department of Medicine, Baylor College of Medicine, Houston, TX
| | - Subi Gandhi
- Department of Epidemiology, University of North Texas Health Science Center, Fort Worth, TX
| | - Sam Dick-Onuoha
- Department of Epidemiology, University of North Texas Health Science Center, Fort Worth, TX
| | - Anthony Lewis
- Department of Epidemiology, University of North Texas Health Science Center, Fort Worth, TX
| | - Hashem B. El-Serag
- Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, TX,Houston VA Health Services Research & Development Center of Excellence, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX,Section of Health Services Research, Department of Medicine, Baylor College of Medicine, Houston, TX
| |
Collapse
|
47
|
Chen Y, Blaser MJ. Association between gastric Helicobacter pylori colonization and glycated hemoglobin levels. J Infect Dis 2012; 205:1195-202. [PMID: 22427676 PMCID: PMC3308905 DOI: 10.1093/infdis/jis106] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Accepted: 11/14/2011] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Few studies have evaluated the potential influence of Helicobacter pylori on biomarkers for diabetes. METHODS We conducted cross-sectional analyses using data from 7417 participants in the National Health and Nutrition Examination Survey (NHANES) III (aged ≥18 years) and 6072 participants in NHANES 1999-2000 (aged ≥3 years) to assess the association between H. pylori and levels of glycosylated hemoglobin (HbA1c). RESULTS There was no association between H. pylori and history of self-reported diabetes. Helicobacter pylori seropositivity, especially H. pylori cagA positivity, was positively associated (P < .01, NHANES III; P = .02, NHANES 1999-2000) with HbA1c levels after excluding individuals with history of diabetes and controlling for potential confounders. There was also a synergistic interaction between H. pylori and higher body mass index (BMI), such that increased levels of HbA1c associated with having both H. pylori and higher BMI were greater than the sum of their individual effects (P for interaction < .01). This interaction was observed consistently in both NHANES III and NHANES 1999-2000 and for H. pylori cagA positivity in NHANES III. CONCLUSIONS The findings indicate a role of H. pylori in impaired glucose tolerance in adults that may be potentiated by higher BMI level.
Collapse
Affiliation(s)
- Yu Chen
- Department of Environmental Medicine, New York University School of Medicine, New York, NY, USA.
| | | |
Collapse
|
48
|
Arnold IC, Hitzler I, Müller A. The immunomodulatory properties of Helicobacter pylori confer protection against allergic and chronic inflammatory disorders. Front Cell Infect Microbiol 2012; 2:10. [PMID: 22919602 PMCID: PMC3417532 DOI: 10.3389/fcimb.2012.00010] [Citation(s) in RCA: 97] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Accepted: 01/30/2012] [Indexed: 12/30/2022] Open
Abstract
Chronic infection with the gastric bacterial pathogen Helicobacter pylori causes gastritis and predisposes carriers to a high risk of developing gastric and duodenal ulcers, gastric cancer, and gastric lymphoma, but has also recently been shown to protect against certain allergic and chronic inflammatory disorders. The immunomodulatory properties that allow the bacteria to persist for decades in infected individuals in the face of a vigorous, yet ultimately non-protective, innate, and adaptive immune response may at the same time confer protection against allergies, asthma, and inflammatory bowel diseases. Experimental evidence from mouse models suggests that H. pylori has evolved to skew the adaptive immune response toward immune tolerance rather than immunity, which promotes persistent infection on the one hand, and inhibits auto-aggressive and allergic T-cell responses on the other. Regulatory T-cells mediating peripheral immune tolerance have emerged as key cellular players in facilitating persistent infection as well as protection from allergies, in both observational studies in humans and experimental work in mice. Recent data suggest that H. pylori actively targets dendritic cells to promote tolerance induction. The findings discussed in this review raise the possibility of harnessing the immunomodulatory properties of H. pylori for the prevention and treatment of allergic and auto-immune diseases, and also provide new insights relevant for H. pylori-specific vaccine development.
Collapse
Affiliation(s)
- Isabelle C Arnold
- Institute of Molecular Cancer Research, University of Zürich Zürich, Switzerland
| | | | | |
Collapse
|
49
|
Cohen D, Shoham O, Orr N, Muhsen K. An inverse and independent association between Helicobacter pylori infection and the incidence of shigellosis and other diarrheal diseases. Clin Infect Dis 2011; 54:e35-42. [PMID: 22157171 DOI: 10.1093/cid/cir916] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES We examined the association between Helicobacter pylori infection and the incidence of diarrheal diseases. METHODS In a nested case-control study participants were sampled from cohorts of male Israeli soldiers aged 18-21 years, serving in field units and followed up for diarrheal diseases. Case patients (n = 177) were subjects who visited the base clinic because diarrhea and were positive for Shigella sonnei (n = 66), enterotoxigenic Escherichia coli (ETEC) (n = 31) or negative for bacterial pathogens (n = 80; diarrhea of unknown etiology). Controls (n = 418) were subjects who did not suffer from a diarrheal disease during the follow-up. They were matched to case patients by training unit and period. Serum samples were obtained from participants at the beginning of their field training and were tested for anti-H. pylori immunoglobulin (Ig) G and preexisting Shigella sonnei lipopolysaccharide IgG and IgA antibodies using enzyme-linked immunosorbent assay. RESULTS The proportion of H. pylori-infected subjects was significantly lower among case patients with infection of unknown etiology (36.3%) than among controls (56.0%) (adjusted odds ratio [OR], 0.43; 95% confidence interval [CI], .24-.77; P = .005). The proportion of H. pylori-infected subjects among case patients with S. sonnei shigellosis was also significantly lower than in the control group: 36.3% versus 56.0%. The association persisted after adjusting for sociodemographic variables and preexisting S. sonnei serum IgA antibodies (adjusted OR, 0.37; 95% CI, .14-.95; P = .03) and IgG antibodies (adjusted OR, 0.38; 95% CI, .14-.99; P = .04). The direction of the association between H. pylori infection and ETEC diarrhea was similar, albeit not statistically significant. CONCLUSIONS Our findings suggest an active role of H. pylori in protection against diarrheal diseases.
Collapse
Affiliation(s)
- Dani Cohen
- Department of Epidemiology and Preventive Medicine, School of Public Health, Sackler Faculty of Medicine, Tel Aviv University, Israel.
| | | | | | | |
Collapse
|
50
|
Sachs G, Marcus EA, Scott DR. The role of the NMDA receptor in Helicobacter pylori-induced gastric damage. Gastroenterology 2011; 141:1967-9. [PMID: 22033182 PMCID: PMC4388038 DOI: 10.1053/j.gastro.2011.10.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Affiliation(s)
- George Sachs
- Department of Medicine, Department of Physiology, David Geffen School of Medicine at UCLA and Veterans Administration Greater Los Angeles, Healthcare Sysytem, Los Angeles, California
| | - Elizabeth A. Marcus
- Department of Pediatrics, David Geffen School of Medicine at UCLA and Veterans Administration Greater Los Angeles, Healthcare Sysytem, Los Angeles, California
| | - David R. Scott
- Department of Physiology, David Geffen School of Medicine at UCLA and Veterans Administration Greater Los Angeles, Healthcare Sysytem, Los Angeles, California
| |
Collapse
|