48 results on '"Mejías-Luque R"'
Search Results
2. Expression duktaler und azinärer immunhistochemischer Marker in präneoplastischen Läsionen verschiedener muriner Pankreatitis- und Pankreaskarzinom-Modelle
- Author
-
Metzler, T., Ma, Q., Ruland, J., Brutau-Abia, A., Mejías-Luque, R., Regel, I., Mahajan, U. M., Löprich, J., Matiasek, K., Weichert, W., and Steiger, K.
- Published
- 2023
- Full Text
- View/download PDF
3. MUC5AC (mucin 5AC, oligomeric mucus/gel-forming)
- Author
-
Mejías-Luque, R, primary, Cobler, L, additional, and de, Bolós C, additional
- Published
- 2011
- Full Text
- View/download PDF
4. Gastric cancer prevention by community eradication of Helicobacter pylori: a cluster-randomized controlled trial.
- Author
-
Pan KF, Li WQ, Zhang L, Liu WD, Ma JL, Zhang Y, Ulm K, Wang JX, Zhang L, Bajbouj M, Zhang LF, Li M, Vieth M, Quante M, Wang LH, Suchanek S, Mejías-Luque R, Xu HM, Fan XH, Han X, Liu ZC, Zhou T, Guan WX, Schmid RM, Gerhard M, Classen M, and You WC
- Subjects
- Humans, Male, Female, Middle Aged, China epidemiology, Adult, Anti-Bacterial Agents therapeutic use, Anti-Bacterial Agents administration & dosage, Aged, Metronidazole therapeutic use, Metronidazole administration & dosage, Tetracycline therapeutic use, Tetracycline administration & dosage, Organometallic Compounds therapeutic use, Organometallic Compounds administration & dosage, Stomach Neoplasms prevention & control, Stomach Neoplasms epidemiology, Stomach Neoplasms microbiology, Helicobacter Infections drug therapy, Helicobacter Infections epidemiology, Helicobacter Infections microbiology, Helicobacter pylori drug effects, Omeprazole therapeutic use, Omeprazole administration & dosage
- Abstract
Gastric cancer is a leading cause of cancer-related deaths in China. Affecting more than 40% of the world's population, Helicobacter pylori is a major risk factor for gastric cancer. While previous clinical trials indicated that eradication of H. pylori could reduce gastric cancer risk, this remains to be shown using a population-based approach. We conducted a community-based, cluster-randomized, controlled, superiority intervention trial in Linqu County, China, with individuals who tested positive for H. pylori using a
13 C-urea breath test randomly assigned to receiving either (1) a 10-day, quadruple anti-H. pylori treatment (comprising 20 mg of omeprazole, 750 mg of tetracycline, 400 mg of metronidazole and 300 mg of bismuth citrate) or (2) symptom alleviation treatment with a single daily dosage of omeprazole and bismuth citrate. H. pylori-negative individuals did not receive any treatment. We examined the incidence of gastric cancer as the primary outcome. A total of 180,284 eligible participants from 980 villages were enrolled over 11.8 years of follow-up, and a total of 1,035 cases of incident gastric cancer were documented. Individuals receiving anti-H. pylori therapy showed a modest reduction in gastric cancer incidence in intention-to-treat analyses (hazard ratio 0.86, 95% confidence interval 0.74-0.99), with a stronger effect observed for those having successful H. pylori eradication (hazard ratio 0.81, 95% confidence interval 0.69-0.96) than for those who failed treatment. Moderate adverse effects were reported in 1,345 participants during the 10-day treatment. We observed no severe intolerable adverse events during either treatment or follow-up. The findings suggest the potential for H. pylori mass screening and eradication as a public health policy for gastric cancer prevention. Chinese Clinical Trial Registry identifier: ChiCTR-TRC-10000979 ., Competing Interests: Competing interests The authors declare no competing interests., (© 2024. The Author(s), under exclusive licence to Springer Nature America, Inc.)- Published
- 2024
- Full Text
- View/download PDF
5. Helicobacter pylori infection alters gut virome by expanding temperate phages linked to increased risk of colorectal cancer.
- Author
-
Luo S, Ru J, Khan Mirzaei M, Xue J, Peng X, Ralser A, Hadi JL, Mejías-Luque R, Gerhard M, and Deng L
- Subjects
- Humans, Colorectal Neoplasms virology, Helicobacter Infections complications, Helicobacter pylori, Bacteriophages, Virome, Gastrointestinal Microbiome
- Abstract
Competing Interests: Competing interests: None declared.
- Published
- 2024
- Full Text
- View/download PDF
6. Effects of Helicobacter pylori infection on intestinal microbiota, immunity and colorectal cancer risk.
- Author
-
Engelsberger V, Gerhard M, and Mejías-Luque R
- Subjects
- Humans, Stomach microbiology, Anti-Bacterial Agents therapeutic use, Anti-Bacterial Agents pharmacology, Helicobacter Infections complications, Helicobacter Infections microbiology, Gastrointestinal Microbiome, Helicobacter pylori physiology, Colorectal Neoplasms epidemiology, Colorectal Neoplasms etiology
- Abstract
Infecting about half of the world´s population, Helicobacter pylori is one of the most prevalent bacterial infections worldwide and the strongest known risk factor for gastric cancer. Although H. pylori colonizes exclusively the gastric epithelium, the infection has also been associated with various extragastric diseases, including colorectal cancer (CRC). Epidemiological studies reported an almost two-fold increased risk for infected individuals to develop CRC, but only recently, direct causal and functional links between the chronic infection and CRC have been revealed. Besides modulating the host intestinal immune response, H. pylori is thought to increase CRC risk by inducing gut microbiota alterations. It is known that H. pylori infection not only impacts the gastric microbiota at the site of infection but also leads to changes in bacterial colonization in the distal large intestine. Considering that the gut microbiome plays a driving role in CRC, H. pylori infection emerges as a key factor responsible for promoting changes in microbiome signatures that could contribute to tumor development. Within this review, we want to focus on the interplay between H. pylori infection, changes in the intestinal microbiota, and intestinal immunity. In addition, the effects of H. pylori antibiotic eradication therapy will be discussed., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision., (Copyright © 2024 Engelsberger, Gerhard and Mejías-Luque.)
- Published
- 2024
- Full Text
- View/download PDF
7. A multiserological line assay to potentially discriminate current from past Helicobacter pylori infection.
- Author
-
Li ZX, Bronny K, Formichella L, Mejías-Luque R, Burrell T, Macke L, Lang U, Vasapolli R, Hysenaj O, Stallforth I, Vieth M, You WC, Zhang Y, Suerbaum S, Schulz C, Pan KF, and Gerhard M
- Subjects
- Humans, Antigens, Bacterial, Bacterial Proteins genetics, Antibodies, Bacterial, Cytotoxins, Helicobacter pylori genetics, Helicobacter Infections diagnosis, Helicobacter Infections microbiology, Gastritis microbiology
- Abstract
Objectives: Early diagnosis is important in controlling Helicobacter pylori-induced gastritis and progression to gastric malignancy. Serological testing is an efficient non-invasive diagnostic method, but currently does not allow differentiation between active and past infections. To fill this diagnostic gap we investigated the diagnostic value of a panel of ten H. pylori-specific antibodies in individuals with different H. pylori infection status within a German population., Methods: We used the recomLine Helicobacter IgG 2.0 immunoblotting assay to analyse ten H. pylori-specific antibodies in serum samples collected from 1108 volunteers. From these, 788 samples were used to build exposure and infection status models and 320 samples for model validation. H. pylori infection status was verified by histological examination. We applied logistic regression to select antibodies correlated to infection status and developed, with independent validation, discriminating models and risk scores. Receiving operating characteristic analysis was performed to assess the accuracy of the discriminating models., Results: Antibody reactivity against cytotoxin-associated gene A (CagA), H. pylori chaperone (GroEL), and hook-associated protein 2 homologue (FliD) was independently associated with the risk of H. pylori exposure with ORs and 95% CIs of 99.24 (46.50-211.80), 46.17 (17.45-122.17), and 22.16 (8.46-55.04), respectively. A risk score comprising these three selected antibodies differentiated currently H. pylori infected or eradicated participants from negatives with an area under the curve of 0.976 (95% CI: 0.965-0.987) (Model 1). Seropositivity for vacuolating cytotoxin A (VacA), GroEL, FliD, H. pylori adhesin A (HpaA), and γ-glutamyl transpeptidase (gGT) was associated with a current infection with an area under the curve of 0.870 (95% CI: 0.837-0.903), which may help discriminate currently infected patients from eradicated ones (Model 2)., Discussion: The recomLine assay is sensitive and specific in determining H. pylori infection and eradication status and thus represents a valuable tool in the management of H. pylori infection., (Copyright © 2023 European Society of Clinical Microbiology and Infectious Diseases. Published by Elsevier Ltd. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
8. Gut virome profiling identifies an association between temperate phages and colorectal cancer promoted by Helicobacter pylori infection.
- Author
-
Luo S, Ru J, Mirzaei MK, Xue J, Peng X, Ralser A, Mejías-Luque R, Gerhard M, and Deng L
- Subjects
- Animals, Mice, Virome, Mice, Inbred C57BL, Carcinogenesis, Bacteriophages genetics, Helicobacter pylori, Helicobacter Infections microbiology, Gastrointestinal Microbiome, Colorectal Neoplasms microbiology
- Abstract
Colorectal cancer (CRC) is one of the most commonly diagnosed cancers worldwide. While a close correlation between chronic Helicobacter pylori infection and CRC has been reported, the role of the virome has been overlooked. Here, we infected Apc -mutant mouse models and C57BL/6 mice with H. pylori and conducted a comprehensive metagenomics analysis of H. pylori -induced changes in lower gastrointestinal tract bacterial and viral communities. We observed an expansion of temperate phages in H. pylori infected Apc
+/1638N mice at the early stage of carcinogenesis. Some of the temperate phages were predicted to infect bacteria associated with CRC, including Enterococcus faecalis . We also observed a high prevalence of virulent genes, such as flgJ , cwlJ , and sleB , encoded by temperate phages. In addition, we identified phages associated with pre-onset and onset of H. pylori -promoted carcinogenesis. Through co-occurrence network analysis, we found strong associations between the viral and bacterial communities in infected mice before the onset of carcinogenesis. These findings suggest that the expansion of temperate phages, possibly caused by prophage induction triggered by H. pylori infection, may have contributed to the development of CRC in mice by interacting with the bacterial community.- Published
- 2023
- Full Text
- View/download PDF
9. Metabolite Alterations and Interactions with Microbiota in Helicobacter pylori-Associated Gastric Lesions.
- Author
-
Peng L, Guo Y, Gerhard M, Gao JJ, Liu ZC, Mejías-Luque R, Zhang L, Vieth M, Ma JL, Liu WD, Li ZX, Zhou T, Li WQ, You WC, Zhang Y, and Pan KF
- Subjects
- Humans, RNA, Ribosomal, 16S genetics, Helicobacter pylori genetics, Stomach Neoplasms microbiology, Stomach Neoplasms pathology, Helicobacter Infections microbiology, Helicobacter Infections pathology, Microbiota, Precancerous Conditions microbiology
- Abstract
Metabolites and their interactions with microbiota may be involved in Helicobacter pylori-associated gastric lesion development. This study aimed to explore metabolite alterations upon H. pylori eradication and possible roles of microbiota-metabolite interactions in progression of precancerous lesions. Targeted metabolomics assays and 16S rRNA gene sequencing were conducted to investigate metabolic and microbial alterations of paired gastric biopsy specimens in 58 subjects with successful and 57 subjects with failed anti-H. pylori treatment. Integrative analyses were performed by combining the metabolomics and microbiome profiles from the same intervention participants. A total of 81 metabolites were significantly altered after successful eradication compared to failed treatment, including acylcarnitines, ceramides, triacylglycerol, cholesterol esters, fatty acid, sphingolipids, glycerophospholipids, and glycosylceramides, with P values of <0.05 for all. The differential metabolites showed significant correlations with microbiota in baseline biopsy specimens, such as negative correlations between Helicobacter and glycerophospholipids, glycosylceramide, and triacylglycerol ( P < 0.05 for all), which were altered by eradication. The characteristic negative correlations between glycosylceramides and Fusobacterium , Streptococcus, and Gemella in H. pylori-positive baseline biopsy specimens were further noticed in active gastritis and intestinal metaplasia ( P < 0.05 for all). A panel including differential metabolites, genera, and their interactions may help to discriminate high-risk subjects who progressed from mild to advanced precancerous lesions in short-term and long-term follow-up periods with areas under the curve (AUC) of 0.914 and 0.801, respectively. Therefore, our findings provide new insights into the metabolites and microbiota interactions in H. pylori-associated gastric lesion progression. IMPORTANCE In this study, a panel was established including differential metabolites, genera, and their interactions, which may help to discriminate high-risk subjects for progression from mild lesions to advanced precancerous lesions in short-term and long-term follow-up., Competing Interests: The authors declare no conflict of interest.
- Published
- 2023
- Full Text
- View/download PDF
10. Isotope tracing reveals bacterial catabolism of host-derived glutathione during Helicobacter pylori infection.
- Author
-
Baskerville MJ, Kovalyova Y, Mejías-Luque R, Gerhard M, and Hatzios SK
- Subjects
- Animals, Stomach, Glutathione metabolism, Antioxidants metabolism, Gastric Mucosa microbiology, Mammals, Helicobacter pylori metabolism, Helicobacter Infections microbiology
- Abstract
Mammalian cells synthesize the antioxidant glutathione (GSH) to shield cellular biomolecules from oxidative damage. Certain bacteria, including the gastric pathogen Helicobacter pylori, can perturb host GSH homeostasis. H. pylori infection significantly decreases GSH levels in host tissues, which has been attributed to the accumulation of reactive oxygen species in infected cells. However, the precise mechanism of H. pylori-induced GSH depletion remains unknown, and tools for studying this process during infection are limited. We developed an isotope-tracing approach to quantitatively monitor host-derived GSH in H. pylori-infected cells by mass spectrometry. Using this method, we determined that H. pylori catabolizes reduced GSH from gastric cells using γ-glutamyl transpeptidase (gGT), an enzyme that hydrolyzes GSH to glutamate and cysteinylglycine (Cys-Gly). gGT is an established virulence factor with immunomodulatory properties that is required for H. pylori colonization in vivo. We found that H. pylori internalizes Cys-Gly in a gGT-dependent manner and that Cys-Gly production during H. pylori infection is coupled to the depletion of intracellular GSH from infected cells. Consistent with bacterial catabolism of host GSH, levels of oxidized GSH did not increase during H. pylori infection, and exogenous antioxidants were unable to restore the GSH content of infected cells. Altogether, our results indicate that H. pylori-induced GSH depletion proceeds via an oxidation-independent mechanism driven by the bacterial enzyme gGT, which fortifies bacterial acquisition of nutrients from the host. Additionally, our work establishes a method for tracking the metabolic fate of host-derived GSH during infection., Competing Interests: The authors have declared that no competing interests exist., (Copyright: © 2023 Baskerville et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.)
- Published
- 2023
- Full Text
- View/download PDF
11. Helicobacter pylori promotes colorectal carcinogenesis by deregulating intestinal immunity and inducing a mucus-degrading microbiota signature.
- Author
-
Ralser A, Dietl A, Jarosch S, Engelsberger V, Wanisch A, Janssen KP, Middelhoff M, Vieth M, Quante M, Haller D, Busch DH, Deng L, Mejías-Luque R, and Gerhard M
- Subjects
- Humans, Mice, Animals, Mice, Inbred C57BL, Carcinogenesis pathology, Mucus, Gastric Mucosa pathology, Helicobacter pylori genetics, Helicobacter Infections complications, Helicobacter Infections microbiology, Stomach Neoplasms pathology, Colonic Neoplasms pathology, Microbiota
- Abstract
Objective: Helicobacter pylori infection is the most prevalent bacterial infection worldwide. Besides being the most important risk factor for gastric cancer development, epidemiological data show that infected individuals harbour a nearly twofold increased risk to develop colorectal cancer (CRC). However, a direct causal and functional connection between H. pylori infection and colon cancer is lacking., Design: We infected two Apc -mutant mouse models and C57BL/6 mice with H. pylori and conducted a comprehensive analysis of H. pylori -induced changes in intestinal immune responses and epithelial signatures via flow cytometry, chip cytometry, immunohistochemistry and single cell RNA sequencing. Microbial signatures were characterised and evaluated in germ-free mice and via stool transfer experiments., Results: H. pylori infection accelerated tumour development in Apc -mutant mice. We identified a unique H. pylori -driven immune alteration signature characterised by a reduction in regulatory T cells and pro-inflammatory T cells. Furthermore, in the intestinal and colonic epithelium, H. pylori induced pro-carcinogenic STAT3 signalling and a loss of goblet cells, changes that have been shown to contribute-in combination with pro-inflammatory and mucus degrading microbial signatures-to tumour development. Similar immune and epithelial alterations were found in human colon biopsies from H. pylori -infected patients. Housing of Apc -mutant mice under germ-free conditions ameliorated, and early antibiotic eradication of H. pylori infection normalised the tumour incidence to the level of uninfected controls., Conclusions: Our studies provide evidence that H. pylori infection is a strong causal promoter of colorectal carcinogenesis. Therefore, implementation of H. pylori status into preventive measures of CRC should be considered., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2023. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2023
- Full Text
- View/download PDF
12. CagA-specific Gastric CD8 + Tissue-Resident T Cells Control Helicobacter pylori During the Early Infection Phase.
- Author
-
Koch MRA, Gong R, Friedrich V, Engelsberger V, Kretschmer L, Wanisch A, Jarosch S, Ralser A, Lugen B, Quante M, Vieth M, Vasapolli R, Schulz C, Buchholz VR, Busch DH, Mejías-Luque R, and Gerhard M
- Subjects
- Humans, Animals, Mice, CD8-Positive T-Lymphocytes, CD4-Positive T-Lymphocytes, Stomach, Gastric Mucosa microbiology, Antigens, Bacterial, Bacterial Proteins, Helicobacter pylori, Helicobacter Infections microbiology
- Abstract
Background & Aims: Infection with Helicobacter pylori strongly affects global health by causing chronic gastritis, ulcer disease, and gastric cancer. Although extensive research into the strong immune response against this persistently colonizing bacterium exists, the specific role of CD8
+ T cells remains elusive., Methods: We comprehensively characterize gastric H pylori-specific CD8+ T-cell responses in mice and humans by flow cytometry, RNA-sequencing, immunohistochemistry, and ChipCytometry, applying functional analyses including T-cell depletion, H pylori eradication, and ex vivo restimulation., Results: We define CD8+ T-cell populations bearing a tissue-resident memory (TRM ) phenotype, which infiltrate the gastric mucosa shortly after infection and mediate pathogen control by executing antigen-specific effector properties. These induced CD8+ tissue-resident memory T cells (TRM cells) show a skewed T-cell receptor beta chain usage and are mostly specific for cytotoxin-associated gene A, the distinctive oncoprotein injected by H pylori into host cells. As the infection progresses, we observe a loss of the TRM phenotype and replacement of CD8+ by CD4+ T cells, indicating a shift in the immune response during the chronic infection phase., Conclusions: Our results point toward a hitherto unknown role of CD8+ T-cell response in this bacterial infection, which may have important clinical implications for treatment and vaccination strategies against H pylori., (Copyright © 2023 AGA Institute. Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
13. Effects of Helicobacter pylori adhesin HopQ binding to CEACAM receptors in the human stomach.
- Author
-
Nguyen QA, Schmitt L, Mejías-Luque R, and Gerhard M
- Subjects
- Humans, Antigens, Bacterial genetics, Protein Transport, Adhesins, Bacterial metabolism, Stomach, Virulence Factors metabolism, Bacterial Proteins genetics, Helicobacter pylori
- Abstract
Helicobacter pylori has developed several strategies using its diverse virulence factors to trigger and, at the same time, limit the host's inflammatory responses in order to establish a chronic infection in the human stomach. One of the virulence factors that has recently received more attention is a member of the Helicobacter outer membrane protein family, the adhesin HopQ, which binds to the human Carcinoembryonic Antigen-related Cell Adhesion Molecules (CEACAMs) on the host cell surface. The HopQ-CEACAM interaction facilitates the translocation of the cytotoxin-associated gene A (CagA), an important effector protein of H. pylori , into host cells via the Type IV secretion system (T4SS). Both the T4SS itself and CagA are important virulence factors that are linked to many aberrant host signaling cascades. In the last few years, many studies have emphasized the prerequisite role of the HopQ-CEACAM interaction not only for the adhesion of this pathogen to host cells but also for the regulation of cellular processes. This review summarizes recent findings about the structural characteristics of the HopQ-CEACAM complex and the consequences of this interaction in gastric epithelial cells as well as immune cells. Given that the upregulation of CEACAMs is associated with many H. pylori -induced gastric diseases including gastritis and gastric cancer, these data may enable us to better understand the mechanisms of H. pylori 's pathogenicity., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2023 Nguyen, Schmitt, Mejías-Luque and Gerhard.)
- Published
- 2023
- Full Text
- View/download PDF
14. Immune Biology and Persistence of Helicobacter pylori in Gastric Diseases.
- Author
-
Fuchs S, Gong R, Gerhard M, and Mejías-Luque R
- Subjects
- Humans, Persistent Infection, Biology, Helicobacter pylori, Stomach Neoplasms
- Abstract
Helicobacter pylori is a prevalent pathogen, which affects more than 40% of the global population. It colonizes the human stomach and persists in its host for several decades or even a lifetime, if left untreated. The persistent infection has been linked to various gastric diseases, including gastritis, peptic ulcers, and an increased risk for gastric cancer. H. pylori infection triggers a strong immune response directed against the bacterium associated with the infiltration of innate phagocytotic immune cells and the induction of a Th1/Th17 response. Even though certain immune cells seem to be capable of controlling the infection, the host is unable to eliminate the bacteria as H. pylori has developed remarkable immune evasion strategies. The bacterium avoids its killing through innate recognition mechanisms and manipulates gastric epithelial cells and immune cells to support its persistence. This chapter focuses on the innate and adaptive immune response induced by H. pylori infection, and immune evasion strategies employed by the bacterium to enable persistent infection., (© 2023. The Author(s), under exclusive license to Springer Nature Switzerland AG.)
- Published
- 2023
- Full Text
- View/download PDF
15. JAK-STAT1 Signaling Pathway Is an Early Response to Helicobacter pylori Infection and Contributes to Immune Escape and Gastric Carcinogenesis.
- Author
-
Li X, Pan K, Vieth M, Gerhard M, Li W, and Mejías-Luque R
- Subjects
- Animals, B7-H1 Antigen genetics, B7-H1 Antigen metabolism, Carcinogenesis metabolism, Gastric Mucosa metabolism, Humans, Janus Kinases metabolism, Mice, STAT1 Transcription Factor genetics, STAT1 Transcription Factor metabolism, Signal Transduction genetics, Gastritis pathology, Helicobacter Infections complications, Helicobacter Infections genetics, Helicobacter pylori metabolism, Stomach Neoplasms pathology
- Abstract
Helicobacter pylori infection induces a number of pro-inflammatory signaling pathways contributing to gastric inflammation and carcinogenesis and has been identified as a major risk factor for the development of gastric cancer (GC). Janus kinase-signal transducer and activator of transcription (JAK-STAT) signaling mediates immune regulatory processes, including tumor-driven immune escape. Programmed death ligand 1 (PD-L1) expressed on gastric epithelium can suppress the immune system by shutting down T cell effector function. In a human cohort of subjects with gastric lesions and GC analyzed by proteomics, STAT1 increased along the cascade of progression of precancerous gastric lesions to GC and was further associated with a poor prognosis of GC (Hazard Ratio (95% confidence interval): 2.34 (1.04-5.30)). We observed that STAT1 was activated in human H. pylori -positive gastritis, while in GC, STAT1, and its target gene, PD-L1, were significantly elevated. To confirm the dependency of H. pylori , we infected gastric epithelial cells in vitro and observed strong activation of STAT1 and upregulation of PD-L1, which depended on cytokines produced by immune cells. To investigate the correlation of immune infiltration with STAT1 activation and PD-L1 expression, we employed a mouse model of H. pylori -induced gastric lesions in an Rnf43-deficient background. Here, phosphorylated STAT1 and PD-L1 were correlated with immune infiltration and proliferation. STAT1 and PD-L1 were upregulated in gastric tumor tissues compared with normal tissues and were associated with immune infiltration and poor prognosis based on the TCGA-STAD database. H. pylori -induced activation of STAT1 and PD-L1 expression may prevent immune surveillance in the gastric mucosa, allowing premalignant lesions to progress to gastric cancer.
- Published
- 2022
- Full Text
- View/download PDF
16. Engagement of CEACAM1 by Helicobacter pylori HopQ Is Important for the Activation of Non-Canonical NF-κB in Gastric Epithelial Cells.
- Author
-
Taxauer K, Hamway Y, Ralser A, Dietl A, Mink K, Vieth M, Singer BB, Gerhard M, and Mejías-Luque R
- Abstract
The gastric pathogen Helicobacter pylori infects half of the world's population and is a major risk factor for gastric cancer development. In order to attach to human gastric epithelial cells and inject the oncoprotein CagA into host cells, H. pylori utilizes the outer membrane protein HopQ that binds to the cell surface protein CEACAM, which can be expressed on the gastric mucosa. Once bound, H. pylori activates a number of signaling pathways, including canonical and non-canonical NF-κB. We investigated whether HopQ-CEACAM interaction is involved in activating the non-canonical NF-κB signaling pathway. Different gastric cancer cells were infected with the H. pylori wild type, or HopQ mutant strains, and the activation of non-canonical NF-κB was related to CEACAM expression levels. The correlation between CEACAM levels and the activation of non-canonical NF-κB was confirmed in human gastric tissue samples. Taken together, our findings show that the HopQ-CEACAM interaction is important for activation of the non-canonical NF-κB pathway in gastric epithelial cells.
- Published
- 2021
- Full Text
- View/download PDF
17. Microbiota alteration at different stages in gastric lesion progression: a population-based study in Linqu, China.
- Author
-
Kadeerhan G, Gerhard M, Gao JJ, Mejías-Luque R, Zhang L, Vieth M, Ma JL, Bajbouj M, Suchanek S, Liu WD, Ulm K, Quante M, Li ZX, Zhou T, Schmid R, Classen M, Li WQ, Zhang Y, You WC, and Pan KF
- Abstract
In addition to Helicobacter pylori (H.pylori), gastric microbiota may be involved in carcinogenesis process. However, the longitudinal study to assess changes in the gastric microbiota associated with the development of gastric carcinogenesis is still limited. The aim of this study is to explore dynamic microbial alterations in gastric cancer (GC) development based on a 4-year endoscopic follow-up cohort in Linqu County, China. Microbial alterations were investigated by deep sequencing of the microbial 16S ribosomal RNA gene in 179 subjects with various gastric lesions, and validated in paired gastric biopsies prospectively collected before and after lesion progression and in non-progression controls. Significant differences were found in microbial diversity and community structure across various gastric lesions, with 62 candidate differential taxa between at least two lesion groups. Further validations identified Helicobacter, Bacillus, Capnocytophaga and Prevotella to be associated with lesion progression-to-dysplasia (DYS)/GC (all P < 0.05), especially for subjects progressing from intestinal metaplasia (IM) to DYS/GC. The combination of the four genera in a microbial dysbiosis index showed a significant difference after lesion progression-to-DYS/GC compared to controls (P = 0.027). The panel including the four genera identified subjects after progression-to-DYS/GC with an area under the receiver-operating curve (AUC) of 0.941. Predictive significance was found before lesion progression-to-DYS/GC with an AUC = 0.776 and an even better AUC (0.927) for subjects progressing from IM to DYS/GC. Microbiota may play different roles at different stages in gastric carcinogenesis. A panel of bacterial genera associated with gastric lesions may help to assess gastric microbial dysbiosis and show potential predictive values for lesion progression. Our findings provide new clues for the microbial mechanism of H.pylori-associated carcinogenesis., Competing Interests: None., (AJCR Copyright © 2021.)
- Published
- 2021
18. Senescence: A Novel Driver of Helicobacter pylori-Induced Gastric Atrophy.
- Author
-
Mejías-Luque R
- Subjects
- Atrophy, Humans, Gastritis, Atrophic, Helicobacter Infections, Helicobacter pylori
- Published
- 2021
- Full Text
- View/download PDF
19. Loss of RNF43 Function Contributes to Gastric Carcinogenesis by Impairing DNA Damage Response.
- Author
-
Neumeyer V, Brutau-Abia A, Allgäuer M, Pfarr N, Weichert W, Falkeis-Veits C, Kremmer E, Vieth M, Gerhard M, and Mejías-Luque R
- Subjects
- Adult, Aged, Aged, 80 and over, Animals, Apoptosis, Carcinogenesis metabolism, Case-Control Studies, Cell Proliferation, Female, Gastritis etiology, Gastritis metabolism, Helicobacter Infections microbiology, Helicobacter pylori isolation & purification, Humans, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Middle Aged, Stomach Neoplasms etiology, Stomach Neoplasms metabolism, Tumor Cells, Cultured, Ubiquitin-Protein Ligases genetics, Wnt Signaling Pathway, Xenograft Model Antitumor Assays, Carcinogenesis pathology, DNA Damage, Gastritis pathology, Helicobacter Infections complications, Stomach Neoplasms pathology, Ubiquitin-Protein Ligases metabolism, Ubiquitin-Protein Ligases physiology
- Abstract
Background & Aims: RING finger protein 43 (RNF43) is a tumor suppressor that frequently is mutated in gastric tumors. The link between RNF43 and modulation of Wingless-related integration site (WNT) signaling has not been shown clearly in the stomach. Because mutations in RNF43 are highly enriched in microsatellite-unstable gastric tumors, which show defects in DNA damage response (DDR), we investigated whether RNF43 is involved in DDR in the stomach., Methods: DDR activation and cell viability upon γ-radiation was analyzed in gastric cells where expression of RNF43 was depleted. Response to chemotherapeutic agents 5-fluorouracil and cisplatin was analyzed in gastric cancer cell lines and xenograft tumors. In addition, involvement of RNF43 in DDR activation was analyzed upon Helicobacter pylori infection in wild-type and Rnf43
ΔEx8 mice. Furthermore, a cohort of human gastric biopsy specimens was analyzed for RNF43 expression and mutation status as well as for activation of DDR., Results: RNF43 depletion conferred resistance to γ-radiation and chemotherapy by dampening the activation of DDR, thereby preventing apoptosis in gastric cells. Upon Helicobacter pylori infection, RNF43 loss of function reduced activation of DDR and apoptosis. Furthermore, RNF43 expression correlated with DDR activation in human gastric biopsy specimens, and RNF43 mutations found in gastric tumors conferred resistance to DNA damage. When exploring the molecular mechanisms behind these findings, a direct interaction between RNF43 and phosphorylated H2A histone family member X (γH2AX) was observed., Conclusions: We identified a novel function for RNF43 in the stomach as a regulator of DDR. Loss of RNF43 function in gastric cells confers resistance to DNA damage-inducing radiotherapy and chemotherapy, suggesting RNF43 as a possible biomarker for therapy selection., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2021
- Full Text
- View/download PDF
20. Cysteine Residues in Helicobacter pylori Adhesin HopQ are Required for CEACAM-HopQ Interaction and Subsequent CagA Translocation.
- Author
-
Hamway Y, Taxauer K, Moonens K, Neumeyer V, Fischer W, Schmitt V, Singer BB, Remaut H, Gerhard M, and Mejías-Luque R
- Abstract
Attachment to the host gastric mucosa is a key step in Helicobacter pylori infection. Recently, a novel adhesin, HopQ, was shown to bind distinct host CEACAM proteins-an interaction that was found to be essential for the translocation of CagA, a key virulence factor of H. pylori. The HopQ-CEACAM1 co-crystal structure revealed a binding mode dependent on loops in HopQ that are clasped by disulfide bonds. In this study, we investigated the importance of these cysteine residues for CEACAM1 engagement by H. pylori . We observed a loss of CEACAM1 binding and CagA translocation upon disruption of the disulfide bond in loop CL1 (connecting C103 to C132 in HopQ). Deletion of the Dsb-like oxidoreductase HP0231 did not affect cell surface expression of HopQ or alter the interaction of H. pylori with target cells. Although HP0231 deletion was previously described to impede CagA translocation, our results indicate that this occurs through a HopQ-independent mechanism. Together, our results open up new avenues to therapeutically target the HopQ-CEACAM1 interaction and reduce the burden of pathogenic H. pylori ., Competing Interests: The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.
- Published
- 2020
- Full Text
- View/download PDF
21. A mass spectrometry guided approach for the identification of novel vaccine candidates in gram-negative pathogens.
- Author
-
Hornburg D, Kruse T, Anderl F, Daschkin C, Semper RP, Klar K, Guenther A, Mejías-Luque R, Schneiderhan-Marra N, Mann M, Meissner F, and Gerhard M
- Subjects
- Antigens, Bacterial chemistry, Antigens, Bacterial immunology, Antigens, Surface chemistry, Antigens, Surface immunology, Bacterial Proteins chemistry, Bacterial Proteins immunology, Computational Biology methods, Proteomics methods, Reproducibility of Results, Bacterial Vaccines chemistry, Bacterial Vaccines immunology, Gram-Negative Bacteria immunology, Mass Spectrometry
- Abstract
Vaccination is the most effective method to prevent infectious diseases. However, approaches to identify novel vaccine candidates are commonly laborious and protracted. While surface proteins are suitable vaccine candidates and can elicit antibacterial antibody responses, systematic approaches to define surfomes from gram-negatives have rarely been successful. Here we developed a combined discovery-driven mass spectrometry and computational strategy to identify bacterial vaccine candidates and validate their immunogenicity using a highly prevalent gram-negative pathogen, Helicobacter pylori, as a model organism. We efficiently isolated surface antigens by enzymatic cleavage, with a design of experiment based strategy to experimentally dissect cell surface-exposed from cytosolic proteins. From a total of 1,153 quantified bacterial proteins, we thereby identified 72 surface exposed antigens and further prioritized candidates by computational homology inference within and across species. We next tested candidate-specific immune responses. All candidates were recognized in sera from infected patients, and readily induced antibody responses after vaccination of mice. The candidate jhp_0775 induced specific B and T cell responses and significantly reduced colonization levels in mouse therapeutic vaccination studies. In infected humans, we further show that jhp_0775 is immunogenic and activates IFNγ secretion from peripheral CD4
+ and CD8+ T cells. Our strategy provides a generic preclinical screening, selection and validation process for novel vaccine candidates against gram-negative bacteria, which could be employed to other gram-negative pathogens.- Published
- 2019
- Full Text
- View/download PDF
22. Helicobacter pylori Exploits the NLRC4 Inflammasome to Dampen Host Defenses.
- Author
-
Semper RP, Vieth M, Gerhard M, and Mejías-Luque R
- Subjects
- Animals, CARD Signaling Adaptor Proteins deficiency, Calcium-Binding Proteins deficiency, Epithelial Cells immunology, Epithelial Cells microbiology, Epithelial Cells pathology, Female, Humans, Inflammation immunology, Male, Mice, Mice, Knockout, Neutrophil Infiltration immunology, Tumor Cells, Cultured, CARD Signaling Adaptor Proteins immunology, Calcium-Binding Proteins immunology, Helicobacter pylori immunology, Inflammasomes immunology
- Abstract
Helicobacter pylori colonizes the stomach of around 50% of humans. This chronic infection can lead to gastric pathologic conditions such as gastric ulcers and gastric adenocarcinomas. The strong inflammatory response elicited by H. pylori is characterized by the induction of the expression of several cytokines. Among those, IL-18 is found highly upregulated in infected individuals, and its expression correlates with the severity of gastric inflammation. IL-18 is produced as inactive proform and has to be cleaved by the multiprotein complex inflammasome to be active. In immune cells, the NLRC4 inflammasome, which is activated by flagellin or bacterial secretion systems, was shown to be dispensable for H. pylori -induced inflammasome activation. However, apart from immune cells, gastric epithelial cells can also produce IL-18. In this study, we analyzed the role of the NLRC4 inflammasome during H. pylori infection. Our results indicate that NLRC4 and a functional type IV secretion system are crucial for the production of IL-18 from human and murine gastric epithelial cells. In vivo, Nlrc4
-/- mice failed to produce gastric IL-18 upon H. pylori infection. Compared with wild type mice, Nlrc4-/- mice controlled H. pylori better without showing strong inflammation. Moreover, H. pylori -induced IL-18 inhibits β-defensin 1 expression in a NF-κB-dependent manner, resulting in higher bacterial colonization. At the same time, inflammasome activation enhances neutrophil infiltration, resulting in inflammation. Thus, NLRC4 inflammasome activation and subsequent IL-18 production favors bacterial persistence by inhibiting antimicrobial peptide production and, at the same time, contributes to gastric inflammation., (Copyright © 2019 by The American Association of Immunologists, Inc.)- Published
- 2019
- Full Text
- View/download PDF
23. Concomitant Infection of S. mansoni and H. pylori Promotes Promiscuity of Antigen-Experienced Cells and Primes the Liver for a Lower Fibrotic Response.
- Author
-
Bhattacharjee S, Mejías-Luque R, Loffredo-Verde E, Toska A, Flossdorf M, Gerhard M, and Prazeres da Costa C
- Subjects
- Animals, Antigens immunology, Bone Marrow Cells metabolism, Coinfection microbiology, Coinfection parasitology, Cytokines metabolism, Dendritic Cells cytology, Dendritic Cells metabolism, Female, Gastritis immunology, Gastritis metabolism, Gastritis parasitology, Helicobacter Infections metabolism, Helicobacter Infections pathology, Humans, Interferon-gamma metabolism, Liver metabolism, Liver microbiology, Liver parasitology, Liver pathology, Liver Cirrhosis metabolism, Liver Cirrhosis pathology, Mice, Mice, Inbred C57BL, Schistosomiasis mansoni metabolism, Schistosomiasis mansoni pathology, Stomach immunology, Stomach microbiology, Stomach parasitology, Stomach pathology, Th1 Cells immunology, Th2 Cells immunology, Coinfection immunology, Gastritis microbiology, Helicobacter Infections immunology, Helicobacter pylori, Liver Cirrhosis microbiology, Liver Cirrhosis parasitology, Schistosomiasis mansoni immunology
- Abstract
Helicobacter pylori chronically colonizes the stomach and is strongly associated with gastric cancer. Its concomitant occurrence with helminths such as schistosomes has been linked to reduced cancer incidence, presumably due to suppression of H. pylori-associated pro-inflammatory responses. However, experimental evidence in support of such a causal link or the mutual interaction of both pathogens is lacking. We investigated the effects of co-infection during the different immune phases of S. mansoni infection. Surprisingly, co-infected mice had increased H. pylori gastric colonization during the interferon gamma (IFNγ) phase of schistosome infection but reduced infiltration of T cells in the stomach due to misdirection of antigen-experienced CXCR3
+ T cells to the liver. Unexpectedly, H. pylori co-infection resulted in partial protection from schistosome-induced liver damage. Here, we demonstrate that an increase in fibrosis-protective IL-13Ra2 is associated with H. pylori infection. Thus, our study strongly points to an immunological interaction of anatomically isolated pathogens, eventually resulting in altered disease pathology., (Copyright © 2019 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2019
- Full Text
- View/download PDF
24. Loss of endogenous RNF43 function enhances proliferation and tumour growth of intestinal and gastric cells.
- Author
-
Neumeyer V, Grandl M, Dietl A, Brutau-Abia A, Allgäuer M, Kalali B, Zhang Y, Pan KF, Steiger K, Vieth M, Anton M, Mejías-Luque R, and Gerhard M
- Subjects
- Animals, Caco-2 Cells, Carcinogenesis genetics, Carcinogenesis pathology, Cell Line, Cell Line, Tumor, Colorectal Neoplasms pathology, HT29 Cells, Humans, Mice, Mucous Membrane pathology, Mutation genetics, Stomach Neoplasms pathology, Ubiquitination genetics, Wnt Signaling Pathway genetics, Cell Proliferation genetics, Colorectal Neoplasms genetics, Intestines pathology, Stomach pathology, Stomach Neoplasms genetics, Ubiquitin-Protein Ligases genetics
- Abstract
Ring finger protein 43 (RNF43) is an E3 ubiquitin ligase that has been described to be frequently mutated in gastrointestinal cancers. RNF43 downregulation was associated with distant metastasis, TNM stage and poorer survival in patients with gastric and colorectal cancers. Functional analysis has shown that overexpressed RNF43 negatively regulates Wnt signalling by ubiquitinating Frizzled receptors and targeting them for degradation and by sequestering T-cell factor 4 (TCF4) to the nuclear membrane, thereby inhibiting Wnt-mediated transcription. In the stomach, RNF43 overexpression was shown to impair stem-like properties and to be negatively correlated with expression of Wnt-target genes. In this study, we show that RNF43 knockdown enhances the tumourigenic potential of gastric and colorectal cancer cell lines in vitro and in vivo. Thus, loss of RNF43 leads to increased proliferation and anchorage-independent growth as well as increased invasive capacity. In a xenograft model, RNF43 depletion enhanced tumour growth. Furthermore, we established two mouse models in which mutations in the RING domain of RNF43 were introduced. In the intestine and colon, loss of Rnf43 did not induce changes in epithelial architecture or proliferation. In contrast, in the stomach, thickening of the mucosa, hyperplasia and cellular atypia were observed in these mice. Notably, this was independent of elevated Wnt signalling. Together, our results show that RNF43 plays a tumour suppressive role in gastric and colorectal cancer cells and that the loss of its function alters gastric tissue homeostasis in vivo., (© The Author(s) 2018. Published by Oxford University Press. All rights reserved. For Permissions, please email: journals.permissions@oup.com.)
- Published
- 2019
- Full Text
- View/download PDF
25. Increased LIGHT expression and activation of non-canonical NF-κB are observed in gastric lesions of MyD88-deficient mice upon Helicobacter felis infection.
- Author
-
Mejías-Luque R, Lozano-Pope I, Wanisch A, Heikenwälder M, Gerhard M, and Obonyo M
- Subjects
- Animals, B-Lymphocytes metabolism, B-Lymphocytes pathology, Chemokine CXCL9 genetics, Chemokine CXCL9 metabolism, Helicobacter Infections metabolism, Helicobacter felis, Intercellular Adhesion Molecule-1 genetics, Intercellular Adhesion Molecule-1 metabolism, Male, Mice, Inbred C57BL, Mice, Mutant Strains, Myeloid Differentiation Factor 88 metabolism, STAT3 Transcription Factor metabolism, Signal Transduction, Stomach Diseases metabolism, Stomach Diseases microbiology, Stomach Diseases pathology, T-Lymphocytes metabolism, T-Lymphocytes pathology, Tumor Necrosis Factor Ligand Superfamily Member 14 genetics, Helicobacter Infections pathology, Myeloid Differentiation Factor 88 genetics, NF-kappa B metabolism, Tumor Necrosis Factor Ligand Superfamily Member 14 metabolism
- Abstract
Helicobacter pylori infection induces a number of pro-inflammatory signaling pathways contributing to gastric inflammation and carcinogenesis. Among those, NF-κB signaling plays a pivotal role during infection and malignant transformation of the gastric epithelium. However, deficiency of the adaptor molecule myeloid differentiation primary response 88 (MyD88), which signals through NF-κB, led to an accelerated development of gastric pathology upon H. felis infection, but the mechanisms leading to this phenotype remained elusive. Non-canonical NF-κB signaling was shown to aggravate H. pylori-induced gastric inflammation via activation of the lymphotoxin β receptor (LTβR). In the present study, we explored whether the exacerbated pathology observed in MyD88-deficient (Myd88
-/- ) mice was associated with aberrant activation of non-canonical NF-κB. Our results indicate that, in the absence of MyD88, H. felis infection enhances the activation of non-canonical NF-κB that is associated with increase in Cxcl9 and Icam1 gene expression and CD3+ lymphocyte recruitment. In addition, activation of signal transducer and activator of transcription 3 (STAT3) signaling was higher in Myd88-/- compared to wild type (WT) mice, indicating a link between MyD88 deficiency and STAT3 activation in response to H. felis infection. Thereby, MyD88 deficiency results in accelerated and aggravated gastric pathology induced by Helicobacter through activation of non-canonical NF-κB.- Published
- 2019
- Full Text
- View/download PDF
26. Mutated Rnf43 Aggravates Helicobacter Pylori -Induced Gastric Pathology.
- Author
-
Neumeyer V, Vieth M, Gerhard M, and Mejías-Luque R
- Abstract
The E3 ubiquitin ligase ring finger protein 43 (RNF43) is frequently mutated in gastric tumors and loss of RNF43 expression was suggested to be one of the key events during the transition from adenoma to gastric carcinoma. Functional studies on RNF43 have shown that it acts as a tumor suppressor by negatively regulating Wnt signaling. Interestingly, we observed that RNF43
H292R/H295R mice bearing two point mutations in the ring domain displayed thickening of the mucosa at early age but did not develop neoplasia. In this study, we infected these mice for 6 months with Helicobacter pylori , which has been described as one of the major risk factors for gastric cancer. Mice bearing mutant RNF43H292R/H295R showed higher gastritis scores upon H. pylori infection compared to wild-type mice, accompanied by increased lymphocyte infiltration and Ifng levels. Furthermore, infected Rnf43 mutant mice developed atrophy, hyperplasia and MUC2 expressing metaplasia and displayed higher levels of the gastric stem cell marker CD44 and canonical NF-κB signaling. In summary, our results show that transactivating mutations in the tumor suppressor Rnf43 can worsen H. pylori induced pathology.- Published
- 2019
- Full Text
- View/download PDF
27. Evidence suggests that germline RNF43 mutations are a rare cause of serrated polyposis.
- Author
-
Quintana I, Mejías-Luque R, Terradas M, Navarro M, Piñol V, Mur P, Belhadj S, Grau E, Darder E, Solanes A, Brunet J, Capellá G, Gerhard M, and Valle L
- Subjects
- DNA-Binding Proteins genetics, Humans, Mutation, Oncogene Proteins genetics, Ubiquitin-Protein Ligases, Colonic Polyps, Germ-Line Mutation
- Abstract
Competing Interests: Competing interests: None declared.
- Published
- 2018
- Full Text
- View/download PDF
28. Helicobacter pylori adhesin HopQ disrupts trans dimerization in human CEACAMs.
- Author
-
Moonens K, Hamway Y, Neddermann M, Reschke M, Tegtmeyer N, Kruse T, Kammerer R, Mejías-Luque R, Singer BB, Backert S, Gerhard M, and Remaut H
- Subjects
- Animals, Antigens, CD chemistry, Bacterial Proteins chemistry, CHO Cells, Cell Adhesion Molecules chemistry, Cell Line, Tumor, Cricetulus, Humans, Protein Multimerization, Antigens, CD physiology, Bacterial Proteins physiology, Cell Adhesion Molecules physiology, Helicobacter pylori physiology
- Abstract
The human gastric pathogen Helicobacter pylori is a major causative agent of gastritis, peptic ulcer disease, and gastric cancer. As part of its adhesive lifestyle, the bacterium targets members of the carcinoembryonic antigen-related cell adhesion molecule (CEACAM) family by the conserved outer membrane adhesin HopQ. The HopQ-CEACAM1 interaction is associated with inflammatory responses and enables the intracellular delivery and phosphorylation of the CagA oncoprotein via a yet unknown mechanism. Here, we generated crystal structures of HopQ isotypes I and II bound to the N-terminal domain of human CEACAM1 (C1ND) and elucidated the structural basis of H. pylori specificity toward human CEACAM receptors. Both HopQ alleles target the β-strands G, F, and C of C1ND, which form the trans dimerization interface in homo- and heterophilic CEACAM interactions. Using SAXS, we show that the HopQ ectodomain is sufficient to induce C1ND monomerization and thus providing H. pylori a route to influence CEACAM-mediated cell adherence and signaling events., (© 2018 The Authors.)
- Published
- 2018
- Full Text
- View/download PDF
29. Helicobacter pylori γ-glutamyl transferase contributes to colonization and differential recruitment of T cells during persistence.
- Author
-
Wüstner S, Anderl F, Wanisch A, Sachs C, Steiger K, Nerlich A, Vieth M, Mejías-Luque R, and Gerhard M
- Subjects
- Adaptive Immunity, Animals, CD8-Positive T-Lymphocytes enzymology, CD8-Positive T-Lymphocytes microbiology, Disease Models, Animal, Female, Helicobacter Infections enzymology, Helicobacter Infections microbiology, Helicobacter Infections pathology, Helicobacter pylori growth & development, Immunity, Innate, Interferon-gamma metabolism, Mice, Inbred C57BL, Mice, Knockout, Stomach enzymology, Stomach immunology, Stomach microbiology, Stomach pathology, Bacterial Proteins metabolism, CD8-Positive T-Lymphocytes immunology, Helicobacter Infections immunology, Helicobacter pylori enzymology, Virulence Factors metabolism, gamma-Glutamyltransferase metabolism
- Abstract
Helicobacter pylori γ-glutamyl transferase (gGT) is a key bacterial virulence factor that is not only important for bacterial gastric colonization but also related to the development of gastric pathology. Despite accumulating evidence for pathogenic and immunologic functions of H. pylori gGT, it is still unclear how it supports gastric colonization and how its specific effects on the host's innate and adaptive immune responses contribute to colonization and pathology. We have compared mice showing similar bacterial load after infection with gGT-proficient or gGT-deficient H. pylori to analyse the specific role of the enzyme during infection. Our data indicate that H. pylori gGT supports initial colonization. Nevertheless, bacteria lacking gGT can still colonize and persist. We observed that the presence of gGT during infection favoured a proinflammatory innate and adaptive immune response. Notably, H. pylori gGT activity was linked to increased levels of IFNγ, which were attributed to a differential recruitment of CD8
+ T cells to the stomach. Our data support an essential role for H. pylori gGT in gastric colonization and further suggest that gGT favours infiltration of CD8+ cells to the gastric mucosa, which might play an important and yet overlooked role in the pathogenesis of H. pylori.- Published
- 2017
- Full Text
- View/download PDF
30. Lymphotoxin β receptor signalling executes Helicobacter pylori -driven gastric inflammation in a T4SS-dependent manner.
- Author
-
Mejías-Luque R, Zöller J, Anderl F, Loew-Gil E, Vieth M, Adler T, Engler DB, Urban S, Browning JL, Müller A, Gerhard M, and Heikenwalder M
- Subjects
- Animals, Antigens, Bacterial metabolism, Bacterial Proteins metabolism, Cell Line, Tumor, Chemokine CCL2 metabolism, Chemokine CCL20 metabolism, Chemokine CXCL10 metabolism, Epithelial Cells metabolism, Female, Gastric Mucosa metabolism, Gastritis microbiology, Helicobacter Infections complications, Humans, Lymphotoxin beta Receptor antagonists & inhibitors, Lymphotoxin beta Receptor genetics, Mice, Mice, Inbred C57BL, RNA, Messenger, Signal Transduction, Transcription Factor RelB metabolism, Tumor Necrosis Factor Ligand Superfamily Member 14 metabolism, Tumor Necrosis Factor-alpha pharmacology, Chemokines metabolism, Gastritis metabolism, Helicobacter Infections metabolism, Helicobacter pylori, Lymphotoxin beta Receptor metabolism, NF-kappa B metabolism, Type IV Secretion Systems metabolism
- Abstract
Objective: Lymphotoxin β receptor (LTβR) signalling has been implicated in inflammation-associated tumour development in different tissues. We have analysed the role of LTβR and alternative NF-κB signalling in Helicobacter pylori- mediated gastric inflammation and pathology., Design: We analysed several ligands and receptors of the alternative NF-κB pathway, RelB, p52 nuclear translocation and target genes in tissue samples of H. pylori -infected patients with different degrees of gastritis or early gastric tumours by in situ hybridisation, immunohistochemistry, Western blot and real-time PCR analyses. Molecular mechanisms involved in LTβR activation by H. pylori were assessed in vitro using human gastric cancer cell lines and distinct H. pylori isolates. The effects of blocking or agonistically activating LTβR on gastric pathology during challenge with a human pathogenic H. pylori strain were studied in a mouse model., Results: Among the tested candidates, LT was significantly increased and activated alternative NF-κB signalling was observed in the gastric mucosa of H. pylori -infected patients. H. pylori induced LTβR-ligand expression in a type IV secretion system-dependent but CagA-independent manner, resulting in activation of the alternative NF-κB pathway, which was further enhanced by blocking canonical NF-κB during infection. Blocking LTβR signalling in vivo suppressed H. pylori- driven gastritis, whereas LTβR activation in gastric epithelial cells of infected mice induced a broadened pro-inflammatory chemokine milieu, resulting in exacerbated pathology., Conclusions: LTβR-triggered activation of alternative NF-κB signalling in gastric epithelial cells executes H. pylori -induced chronic gastritis, representing a novel target to restrict gastric inflammation and pathology elicited by H. pylori , while exclusively targeting canonical NF-κB may aggravate pathology by enhancing the alternative pathway., (Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://www.bmj.com/company/products-services/rights-and-licensing/.)
- Published
- 2017
- Full Text
- View/download PDF
31. Immune Evasion Strategies and Persistence of Helicobacter pylori.
- Author
-
Mejías-Luque R and Gerhard M
- Subjects
- Animals, Bacterial Proteins genetics, Bacterial Proteins immunology, Gastric Mucosa immunology, Gastric Mucosa microbiology, Helicobacter Infections microbiology, Helicobacter pylori genetics, Helicobacter pylori physiology, Humans, Virulence Factors genetics, Virulence Factors immunology, Helicobacter Infections immunology, Helicobacter pylori immunology, Immune Evasion
- Abstract
Helicobacter pylori infection is commonly acquired during childhood, can persist lifelong if not treated, and can cause different gastric pathologies, including chronic gastritis, peptic ulcer disease, and eventually gastric cancer. H. pylori has developed a number of strategies in order to cope with the hostile conditions found in the human stomach as well as successful mechanisms to evade the strong innate and adaptive immune responses elicited upon infection. Thus, by manipulating innate immune receptors and related signaling pathways, inducing tolerogenic dendritic cells and inhibiting effector T cell responses, H. pylori ensures low recognition by the host immune system as well as its persistence in the gastric epithelium. Bacterial virulence factors such as cytotoxin-associated gene A, vacuolating cytotoxin A, or gamma-glutamyltranspeptidase have been extensively studied in the context of bacterial immune escape and persistence. Further, the bacterium possesses other factors that contribute to immune evasion. In this chapter, we discuss in detail the main evasion and persistence strategies evolved by the bacterium as well as the specific bacterial virulence factors involved.
- Published
- 2017
- Full Text
- View/download PDF
32. Erratum: Helicobacter pylori adhesin HopQ engages in a virulence-enhancing interaction with human CEACAMs.
- Author
-
Javaheri A, Kruse T, Moonens K, Mejías-Luque R, Debraekeleer A, Asche CI, Tegtmeyer N, Kalali B, Bach NC, Sieber SA, Hill DJ, Königer V, Hauck CR, Moskalenko R, Haas R, Busch DH, Klaile E, Slevogt H, Schmidt A, Backert S, Remaut H, Singer BB, and Gerhard M
- Published
- 2016
- Full Text
- View/download PDF
33. Helicobacter pylori adhesin HopQ engages in a virulence-enhancing interaction with human CEACAMs.
- Author
-
Javaheri A, Kruse T, Moonens K, Mejías-Luque R, Debraekeleer A, Asche CI, Tegtmeyer N, Kalali B, Bach NC, Sieber SA, Hill DJ, Königer V, Hauck CR, Moskalenko R, Haas R, Busch DH, Klaile E, Slevogt H, Schmidt A, Backert S, Remaut H, Singer BB, and Gerhard M
- Subjects
- Adhesins, Bacterial chemistry, Antigens, Bacterial metabolism, Bacterial Proteins metabolism, Cell Line, Crystallography, X-Ray, Humans, Interleukin-8 metabolism, Protein Binding, Protein Conformation, Protein Transport, Virulence, Adhesins, Bacterial metabolism, Bacterial Adhesion, Cell Adhesion Molecules metabolism, Helicobacter pylori pathogenicity, Helicobacter pylori physiology, Host-Pathogen Interactions
- Abstract
Helicobacter pylori specifically colonizes the human gastric epithelium and is the major causative agent for ulcer disease and gastric cancer development. Here, we identify members of the carcinoembryonic antigen-related cell adhesion molecule (CEACAM) family as receptors of H. pylori and show that HopQ is the surface-exposed adhesin that specifically binds human CEACAM1, CEACAM3, CEACAM5 and CEACAM6. HopQ-CEACAM binding is glycan-independent and targeted to the N-domain. H. pylori binding induces CEACAM1-mediated signalling, and the HopQ-CEACAM1 interaction enables translocation of the virulence factor CagA into host cells and enhances the release of pro-inflammatory mediators such as interleukin-8. Based on the crystal structure of HopQ, we found that a β-hairpin insertion (HopQ-ID) in HopQ's extracellular 3+4 helix bundle domain is important for CEACAM binding. A peptide derived from this domain competitively inhibits HopQ-mediated activation of the Cag virulence pathway, as genetic or antibody-mediated abrogation of the HopQ function shows. Together, our data suggest the HopQ-CEACAM1 interaction to be a potentially promising novel therapeutic target to combat H. pylori-associated diseases.
- Published
- 2016
- Full Text
- View/download PDF
34. Helicobacter pylori γ-Glutamyltranspeptidase Induces Tolerogenic Human Dendritic Cells by Activation of Glutamate Receptors.
- Author
-
Käbisch R, Semper RP, Wüstner S, Gerhard M, and Mejías-Luque R
- Subjects
- Adaptive Immunity, Cells, Cultured, Cyclic AMP metabolism, Helicobacter Infections immunology, Humans, Interleukin-6 immunology, Signal Transduction, T-Lymphocytes, Regulatory immunology, Virulence Factors immunology, Bacterial Proteins immunology, Dendritic Cells immunology, Helicobacter pylori enzymology, Receptors, Glutamate metabolism, gamma-Glutamyltransferase immunology
- Abstract
Helicobacter pylori infection is characterized by chronic persistence of the bacterium. Different virulence factors, including H. pylori γ-glutamyltranspeptidase (gGT), have been reported to induce tolerogenicity by reprogramming dendritic cells (DCs). gGT is present in all bacterial isolates, indicating an important role for gGT in the course of infection. In the current study, we have analyzed the effect of H. pylori gGT on human DCs and the subsequent adaptive immune response. We show that glutamate produced due to H. pylori gGT enzymatic activity tolerizes DCs by inhibiting cAMP signaling and dampening IL-6 secretion in response to the infection. Together, our results provide a novel molecular mechanism by which H. pylori manipulates the host's immune response to persist within its host., (Copyright © 2016 by The American Association of Immunologists, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
35. Helicobacter pylori HP0231 Influences Bacterial Virulence and Is Essential for Gastric Colonization.
- Author
-
Zhong Y, Anderl F, Kruse T, Schindele F, Jagusztyn-Krynicka EK, Fischer W, Gerhard M, and Mejías-Luque R
- Subjects
- Antigens, Bacterial metabolism, Bacterial Proteins metabolism, Cell Line, Helicobacter pylori metabolism, Helicobacter pylori physiology, Protein Transport, Vacuoles microbiology, Virulence, Helicobacter pylori pathogenicity, Stomach microbiology
- Abstract
The Dsb protein family is responsible for introducing disulfide bonds into nascent proteins in prokaryotes, stabilizing the structure of many proteins. Helicobacter pylori HP0231 is a Dsb-like protein, shown to catalyze disulfide bond formation and to participate in redox homeostasis. Notably, many H. pylori virulence factors are stabilized by the formation of disulfide bonds. By employing H. pylori HP0231 deficient strains we analyzed the effect of lack of this bacterial protein on the functionality of virulence factors containing putative disulfide bonds. The lack of H. pylori HP0231 impaired CagA translocation into gastric epithelial cells and reduced VacA-induced cellular vacuolation. Moreover, H. pylori HP0231 deficient bacteria were not able to colonize the gastric mucosa of mice, probably due to compromised motility. Together, our data demonstrate an essential function for H. pylori HP0231 in gastric colonization and proper function of bacterial virulence factors related to gastric pathology.
- Published
- 2016
- Full Text
- View/download PDF
36. The E3 ligase RNF43 inhibits Wnt signaling downstream of mutated β-catenin by sequestering TCF4 to the nuclear membrane.
- Author
-
Loregger A, Grandl M, Mejías-Luque R, Allgäuer M, Degenhart K, Haselmann V, Oikonomou C, Hatzis P, Janssen KP, Nitsche U, Gradl D, van den Broek O, Destree O, Ulm K, Neumaier M, Kalali B, Jung A, Varela I, Schmid RM, Rad R, Busch DH, and Gerhard M
- Subjects
- Animals, Basic Helix-Loop-Helix Leucine Zipper Transcription Factors genetics, Cell Line, Tumor, DNA-Binding Proteins genetics, Humans, Mutation, Nuclear Envelope genetics, Oncogene Proteins genetics, Transcription Factor 4, Transcription Factors genetics, Transcription, Genetic, Ubiquitin-Protein Ligases, Xenopus laevis, beta Catenin genetics, Basic Helix-Loop-Helix Leucine Zipper Transcription Factors metabolism, DNA-Binding Proteins metabolism, Nuclear Envelope metabolism, Oncogene Proteins metabolism, Transcription Factors metabolism, Wnt Signaling Pathway physiology, beta Catenin metabolism
- Abstract
Given its fundamental role in development and cancer, the Wnt-β-catenin signaling pathway is tightly controlled at multiple levels. RING finger protein 43 (RNF43) is an E3 ubiquitin ligase originally found in stem cells and proposed to inhibit Wnt signaling by interacting with the Wnt receptors of the Frizzled family. We detected endogenous RNF43 in the nucleus of human intestinal crypt and colon cancer cells. We found that RNF43 physically interacted with T cell factor 4 (TCF4) in cells and tethered TCF4 to the nuclear membrane, thus silencing TCF4 transcriptional activity even in the presence of constitutively active mutants of β-catenin. This inhibitory mechanism was disrupted by the expression of RNF43 bearing mutations found in human gastrointestinal tumors, and transactivation of the Wnt pathway was observed in various cells and in Xenopus embryos when the RING domain of RNF43 was mutated. Our findings indicate that RNF43 inhibits the Wnt pathway downstream of oncogenic mutations that activate the pathway. Mimicking or enhancing this inhibitory activity of RNF43 may be useful to treat cancers arising from aberrant activation of the Wnt pathway., (Copyright © 2015, American Association for the Advancement of Science.)
- Published
- 2015
- Full Text
- View/download PDF
37. Helicobacter pylori γ-glutamyltranspeptidase impairs T-lymphocyte function by compromising metabolic adaption through inhibition of cMyc and IRF4 expression.
- Author
-
Wüstner S, Mejías-Luque R, Koch MF, Rath E, Vieth M, Sieber SA, Haller D, and Gerhard M
- Subjects
- Cell Proliferation, Cytokines metabolism, Gastric Mucosa microbiology, Gastric Mucosa pathology, Glutamine metabolism, Host-Pathogen Interactions, Humans, Immune Evasion, Lymphocyte Activation, Helicobacter pylori enzymology, Interferon Regulatory Factors antagonists & inhibitors, Proto-Oncogene Proteins c-myc antagonists & inhibitors, T-Lymphocytes drug effects, T-Lymphocytes immunology, Virulence Factors metabolism, gamma-Glutamyltransferase metabolism
- Abstract
Helicobacter pylori (H. pylori) is a human-specific pathogen that has evolved to cope with the immune response elicited against the infection. We previously reported that H. pylori γ-glutamyltranspeptidase (gGT) impairs T-lymphocyte proliferation and thus might act as immune regulatory factor. In this study, we analysed the underlying mechanism and its implications for H. pylori persistence. We found that H. pylori gGT compromised T-cell proliferation, activation and effector cytokine expression by specifically depriving the extracellular space of glutamine. When assessing signalling cascades and transcription factors affected by H. pylori gGT, we found that expression of cMyc and IRF4, both required for metabolic adaptation of T-lymphocytes, was highly sensitive to extracellular glutamine levels and downregulated upon gGT treatment. Moreover, we could confirm decreased IRF4 expression in T-lymphocytes infiltrating the stomach of infected individuals. Thus, our results suggest that H. pylori gGT-mediated glutamine deprivation in the gastric mucosa may suppress T-cell function thereby contributing to bacterial persistence., (© 2014 John Wiley & Sons Ltd.)
- Published
- 2015
- Full Text
- View/download PDF
38. Helicobacter pylori-induced IL-1β secretion in innate immune cells is regulated by the NLRP3 inflammasome and requires the cag pathogenicity island.
- Author
-
Semper RP, Mejías-Luque R, Groß C, Anderl F, Müller A, Vieth M, Busch DH, Prazeres da Costa C, Ruland J, Groß O, and Gerhard M
- Subjects
- Animals, Bacterial Proteins genetics, Female, Genomic Islands genetics, Helicobacter Infections genetics, Helicobacter Infections pathology, Helicobacter pylori genetics, Humans, Male, Mice, NLR Family, Pyrin Domain-Containing 3 Protein, Bacterial Proteins immunology, Carrier Proteins immunology, Genomic Islands immunology, Helicobacter Infections immunology, Helicobacter pylori immunology, Immunity, Innate, Inflammasomes immunology, Interleukin-1beta immunology
- Abstract
Infection with the gram-negative bacterium Helicobacter pylori is the most prevalent chronic bacterial infection, affecting ∼50% of the world's population, and is the main risk factor of gastric cancer. The proinflammatory cytokine IL-1β plays a crucial role in the development of gastric tumors and polymorphisms in the IL-1 gene cluster leading to increased IL-1β production have been associated with increased risk for gastric cancer. To be active, pro-IL-1β must be cleaved by the inflammasome, an intracellular multiprotein complex implicated in physiological and pathological inflammation. Recently, H. pylori was postulated to activate the inflammasome in murine bone marrow-derived dendritic cells; however, the molecular mechanisms as well as the bacterial virulence factor acting as signal 2 activating the inflammasome remain elusive. In this study, we analyzed the inflammasome complex regulating IL-1β upon H. pylori infection as well as the molecular mechanisms involved. Our results indicate that H. pylori-induced IL-1β secretion is mediated by activation of the nucleotide-binding oligomerization domain-like receptor family, pyrin domain-containing 3 inflammasome. We also show that reactive oxygen species, potassium efflux, and lysosomal destabilization are the main cellular mechanisms responsible of nucleotide-binding oligomerization domain family, pyrin domain-containing 3 inflammasome activation upon H. pylori infection, and identify vacuolating cytotoxin A and cag pathogenicity island as the bacterial virulence determinants involved. Moreover, in vivo experiments indicate an important role for the inflammasome in the onset and establishment of H. pylori infection and in the subsequent inflammatory response of the host., (Copyright © 2014 by The American Association of Immunologists, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
39. Involvement of Toll-like receptors on Helicobacter pylori-induced immunity.
- Author
-
Käbisch R, Mejías-Luque R, Gerhard M, and Prinz C
- Subjects
- Antibodies, Neutralizing pharmacology, CD4-Positive T-Lymphocytes immunology, Cell Polarity, Dendritic Cells immunology, Humans, Interferon-gamma metabolism, Interleukin-10 metabolism, Interleukin-12 metabolism, Interleukin-17 metabolism, Interleukin-6 metabolism, Signal Transduction, T-Lymphocytes, Regulatory immunology, Dendritic Cells microbiology, Helicobacter Infections immunology, Helicobacter pylori immunology, Toll-Like Receptors physiology
- Abstract
Dendritic cells (DCs) play a major role in the innate immune response since they recognize a broad repertoire of PAMPs mainly via Toll-like receptors (TLRs). During Helicobacter pylori (H. pylori) infection, TLRs have been shown to be important to control cytokine response particularly in murine DCs. In the present study we analyzed the effect of blocking TLRs on human DCs. Co-incubation of human DCs with H. pylori resulted in the release of the pro-inflammatory cytokines IL-12p70, IL-6 and IL-10. Release of IL-12p70 and IL-10 was predominantly influenced when TLR4 signaling was blocked by adding specific antibodies, suggesting a strong influence on subsequent T cell responses through TLR4 activation on DCs. Co-incubation of H. pylori-primed DC with allogeneic CD4+ T cells resulted in the production of IFN-γ and IL-17A as well as the expression of Foxp3, validating a mixed Th1/Th17 and Treg response in vitro. Neutralization of TLR4 during H. pylori infection resulted in significantly decreased amounts of IL-17A and IFN-γ and reduced levels of Foxp3-expressing and IL-10-secreting T cells. Our findings suggest that DC cytokine secretion induced upon TLR4-mediated recognition of H. pylori influences inflammatory and regulatory T cell responses, which might facilitate the chronic bacterial persistence.
- Published
- 2014
- Full Text
- View/download PDF
40. The stem cell factor SOX2 regulates the tumorigenic potential in human gastric cancer cells.
- Author
-
Hütz K, Mejías-Luque R, Farsakova K, Ogris M, Krebs S, Anton M, Vieth M, Schüller U, Schneider MR, Blum H, Wagner E, Jung A, and Gerhard M
- Subjects
- Animals, Base Sequence, Carcinogenesis, Cell Line, Tumor, DNA Primers, Female, Humans, Mice, Real-Time Polymerase Chain Reaction, SOXB1 Transcription Factors genetics, SOXB1 Transcription Factors physiology, Stomach Neoplasms pathology
- Abstract
Gastric cancer (GC) is still one of the most common causes of cancer-related death worldwide, which is mainly attributable to late diagnosis and poor treatment options. Infection with Helicobacter pylori, different environmental factors and genetic alterations are known to influence the risk of developing gastric tumors. However, the molecular mechanisms involved in gastric carcinogenesis are still not fully understood, making it difficult to design targeted therapeutic approaches. Aberrant expression of the specific gastric differentiation marker SOX2 has been observed in stomach cancer. However, the role of SOX2 in gastric tumors has not been well established to date. To elucidate the role of SOX2 in gastric tumorigenesis, SOX2 transcriptional activity was blocked in AZ-521 cells. Interestingly, inhibition of SOX2 reduced cell proliferation and migration, increased apoptosis and induced changes in cell cycle. Blocking of SOX2 also reduced the tumorigenic potential of AZ-521 cells in vivo. In addition, correlation of SOX2 expression and proliferation was observed in a subset of human gastric tumors. Finally, target genes of SOX2 were for the first time identified by RNA microarray in GC cells. Taken together, the results presented here indicate that SOX2 controls several aspects related to GC development and progression by regulating the expression of members of important signaling pathways. These findings could provide new therapeutic options for a subset of GCs exhibiting SOX2 deregulation.
- Published
- 2014
- Full Text
- View/download PDF
41. Helicobacter pylori cytotoxin-associated gene A impairs human dendritic cell maturation and function through IL-10-mediated activation of STAT3.
- Author
-
Kaebisch R, Mejías-Luque R, Prinz C, and Gerhard M
- Subjects
- Antigens, Bacterial metabolism, Bacterial Proteins metabolism, Cells, Cultured, Cytokines biosynthesis, Humans, Signal Transduction, T-Lymphocytes, Regulatory immunology, T-Lymphocytes, Regulatory metabolism, Antigens, Bacterial genetics, Bacterial Proteins genetics, Dendritic Cells immunology, Dendritic Cells metabolism, Helicobacter pylori genetics, Interleukin-10 metabolism, STAT3 Transcription Factor metabolism
- Abstract
Helicobacter pylori infection induces chronic gastric inflammation that can progress to cancer. In this process, the virulence factor cytotoxin-associated gene A (CagA) plays a central role by directly altering epithelial cell signaling and inducing a strong Th1 immune response, which contributes to carcinogenesis. It is still barely understood how the bacterium evades clearance despite this solid immune response and persists lifelong. Dendritic cells (DCs) play a major role in determining the adaptive immune response toward H. pylori, and high levels of regulatory T cells have been detected infiltrating the gastric mucosa of H. pylori-infected patients, which contribute to bacterial persistence. Although murine studies indicate that H. pylori induces tolerization of DCs and impairs DC maturation, the virulence determinants involved are still controversial. Moreover, the signaling cascades engaged in human DC tolerization upon H. pylori infection remain unknown. In the current study, we analyzed the effect of H. pylori infection on human DC maturation and function, focusing on the virulence factors implicated and signaling pathways involved. Our results reveal that CagA is crucial for DC tolerization by modulating IL-10 secretion and, in turn, STAT3 phosphorylation, favoring a regulatory T cell immune response. Our findings help to unravel the paradox why CagA-positive strains, although eliciting a stronger inflammatory response, have overcome evolutionary pressure and persisted in their human host.
- Published
- 2014
- Full Text
- View/download PDF
42. H. pylori virulence factors: influence on immune system and pathology.
- Author
-
Kalali B, Mejías-Luque R, Javaheri A, and Gerhard M
- Subjects
- Adhesins, Bacterial metabolism, Bacterial Adhesion, Bacterial Outer Membrane Proteins metabolism, Helicobacter Infections pathology, Humans, Immune System, Inflammation, Stomach microbiology, Symbiosis, Treatment Outcome, Helicobacter Infections immunology, Helicobacter pylori pathogenicity, Virulence Factors metabolism
- Abstract
Helicobacter pylori is the most widespread chronic bacterial agent in humans and is well recognized for its association with ulcer disease and gastric cancer, with both representing major global health and socioeconomic issues. Given the high level of adaptation and the coevolution of this bacterium with its human host, a thorough and multidirectional view of the specific microbiological characteristics of this infection as well as the host physiology is needed in order to develop novel means of prevention of therapy. This review aims to pinpoint some of these potentially important angles, which have to be considered mutually when studying H. pylori's pathogenicity. The host's biological changes due to the virulence factors are a valuable pillar of H. pylori research as are the mechanisms by which bacteria provoke these changes. In this context, necessary adhesion molecules and significant virulence factors of H. pylori are discussed. Moreover, metabolism of the bacteria, one of the most important aspects for a better understanding of bacterial physiology and consequently possible therapeutic and prophylactic strategies, is addressed. On the other hand, we discuss the recent experimental proofs of the "hygiene hypothesis" in correlation with Helicobacter's infection, which adds another aspect of complexity to this infection.
- Published
- 2014
- Full Text
- View/download PDF
43. Activation of the NF-kB pathway downregulates TFF-1 in gastric carcinogenesis.
- Author
-
Cobler L, Mejías-Luque R, Garrido M, Pera M, Badia-Garrido E, and de Bolós C
- Subjects
- Aged, Blotting, Western, Down-Regulation, Female, Fluorescent Antibody Technique, Humans, Immunohistochemistry, Male, Middle Aged, Reverse Transcriptase Polymerase Chain Reaction, Trefoil Factor-1, Cell Transformation, Neoplastic metabolism, NF-kappa B metabolism, Signal Transduction physiology, Stomach Neoplasms metabolism, Tumor Suppressor Proteins metabolism
- Abstract
Trefoil factor 1 (TFF1) is expressed in the normal superficial epithelium of the stomach and is implicated in the maintenance of gastric epithelial structure and function. During gastric carcinogenesis, in which pro-inflammatory cytokines play a crucial role, its expression level decreases suggesting a role as tumor suppressor factor. We have compared expression of TFF1 in gastric mucosa from cancer patients, in which several degrees of inflammatory infiltrate are present, with that in normal mucosa from non-cancer patients without infiltrating inflammatory cells. TFF1 is less expressed in the superficial gastric epithelium from cancer patients than in that from normal individuals in which the nuclear factor (NF)-κB pathway is not activated. We analyzed TFF1 expression in ex vivo samples of gastric mucosa from cancer patients, and in MKN45 gastric cancer cell line after exposure to proinflammatory cytokines interleukin (IL)-1β or tumor necrosis factor (TNF)-α, that activate the NF-κB pathway. We found that IL-1β and TNF-α activate the NF-κB pathway, as reflected in the nuclear expression of p65 and the activation of p-IκBα, and downregulate TFF1 expression after 1 or 2 h of exposure. Moreover, cells in the superficial gastric epithelium in ex vivo samples co-expressed TFF1/p65 at cellular level, whereas tumor cells did not. In summary, downregulation of TFF1 expression during gastric neoplastic transformation is associated with activation of the NF-κB pathway through IL-1β or TNF-α, but other regulatory mechanisms might also be involved.
- Published
- 2013
- Full Text
- View/download PDF
44. Helicobacter bilis gamma-glutamyltranspeptidase enhances inflammatory stress response via oxidative stress in colon epithelial cells.
- Author
-
Javed S, Mejías-Luque R, Kalali B, Bolz C, and Gerhard M
- Subjects
- Cell Line, Tumor, Colonic Neoplasms genetics, Colonic Neoplasms metabolism, Colonic Neoplasms microbiology, Cyclic AMP Response Element-Binding Protein metabolism, Glutamine metabolism, Helicobacter Infections genetics, Humans, Interleukin-8 biosynthesis, NF-kappa B metabolism, Reactive Oxygen Species metabolism, Signal Transduction, Transcription Factor AP-1 metabolism, Transcriptional Activation, Colon metabolism, Colon microbiology, Helicobacter enzymology, Helicobacter Infections metabolism, Intestinal Mucosa metabolism, Intestinal Mucosa microbiology, Oxidative Stress, gamma-Glutamyltransferase metabolism
- Abstract
Helicobacter bilis (H. bilis) infection is associated with cases of inflammatory bowel Disease, thyphlocolitis, hepatitis and cholecystitis. However, little is known about the bacterial virulence determinants or the molecular mechanisms involved. Recently, H. bilis γ-glutamyltranspeptidase (HBgGT) was shown to be a virulence factor decreasing host cell viability. Bacterial gGTs play a key role in synthesis and degradation of glutathione and enables the bacteria to utilize extracellular glutamine and glutathione as sources of glutamate. gGT-mediated loss of cell viability has so far been linked to DNA damage via oxidative stress, but the signaling cascades involved herein have not been described. In this study, we identified enhanced ROS production induced by HBgGT as a central factor involved in the activation of the oxidative stress response cascades, which finally activate CREB, AP-1 and NF-κB in H. bilis infected colon cancer cells. IL-8, an important pro-inflammatory chemokine that is a common downstream target of these transcription factors, was up-regulated upon H. bilis infection in an HBgGT dependent manner. Moreover, the induction of these signaling responses and inflammatory cytokine production in host cells could be linked to HBgGT-mediated glutamine deprivation. This study implicates for the first time HBgGT as an important regulator of signaling cascades regulating inflammation in H. bilis infected host epithelial cells that could be responsible for induction of inflammatory disorders by the bacterium.
- Published
- 2013
- Full Text
- View/download PDF
45. Regulation of glycosyltransferases and Lewis antigens expression by IL-1β and IL-6 in human gastric cancer cells.
- Author
-
Padró M, Mejías-Luque R, Cobler L, Garrido M, Pérez-Garay M, Puig S, Peracaula R, and de Bolós C
- Subjects
- Animals, Cytokine Receptor gp130 genetics, Cytokine Receptor gp130 metabolism, Gene Expression Regulation, Neoplastic, Glycosyltransferases genetics, Humans, Lewis Blood Group Antigens genetics, Mice, Oligosaccharides genetics, RNA, Messenger metabolism, STAT3 Transcription Factor genetics, STAT3 Transcription Factor metabolism, Sialyl Lewis X Antigen, Stomach Neoplasms genetics, Transplantation, Heterologous, Tumor Cells, Cultured, Glycosyltransferases metabolism, Interleukin-1beta pharmacology, Interleukin-6 pharmacology, Lewis Blood Group Antigens metabolism, Oligosaccharides metabolism, Stomach Neoplasms enzymology, Stomach Neoplasms immunology
- Abstract
Inflammation of stomach mucosa has been postulated as initiator of gastric carcinogenesis and the presence of pro-inflammatory cytokines can regulate specific genes involved in this process. The cellular expression pattern of glycosyltransferases and Lewis antigens detected in the normal mucosa changed during the neoplassic transformation. The aim of this work was to determine the regulation of specific fucosyltransferases and sialyltransferases by IL-1β and IL-6 pro-inflammatory cytokines in MKN45 gastric cancer cells. IL-1β induced significant increases in the mRNA levels of FUT1, FUT2 and FUT4, and decreases of FUT3 and FUT5. In IL-6 treatments, enhanced FUT1 and lower FUT3 and FUT5 mRNA expression were detected. No substantial changes were observed in the levels of ST3GalIII and ST3GalIV. The activation of FUT1, FUT2 and FUT4 by IL-1β is through the NF-κB pathway and the down-regulation of FUT3 and FUT5 by IL-6 is through the gp130/STAT-3 pathway, since they are inhibited specifically by panepoxydone and AG490, respectively. The levels of Lewis antigens after IL-1β or IL-6 stimulation decreased for sialyl-Lewis x, and no significant differences were found in the rest of the Lewis antigens analyzed, as it was also observed in subcutaneous mice tumors from MKN45 cells treated with IL-1β or IL-6. In addition, in 61 human intestinal-type gastric tumors, sialyl-Lewis x was highly detected in samples from patients that developed metastasis. These results indicate that the expression of the fucosyltransferases involved in the synthesis of Lewis antigens in gastric cancer cells can be specifically modulated by IL-1β and IL-6 inflammatory cytokines.
- Published
- 2011
- Full Text
- View/download PDF
46. IL-6 induces MUC4 expression through gp130/STAT3 pathway in gastric cancer cell lines.
- Author
-
Mejías-Luque R, Peiró S, Vincent A, Van Seuningen I, and de Bolós C
- Subjects
- Cell Line, Tumor, Gene Expression Regulation, Neoplastic drug effects, Humans, Mucin-2, Mucin-4, Mucins genetics, Promoter Regions, Genetic genetics, Protein Binding, STAT3 Transcription Factor genetics, Cytokine Receptor gp130 metabolism, Interleukin-6 pharmacology, Mucins metabolism, STAT3 Transcription Factor metabolism, Signal Transduction drug effects, Stomach Neoplasms metabolism
- Abstract
The gastric mucosal levels of the pro-inflammatory cytokine Interleukin 6 (IL-6) have been reported to be increased in Helicobacter pylori-infected subjects and, in gastric adenocarcinomas, the up-regulation of intestinal mucin genes (MUC2 and MUC4) has been detected. To analyse the regulatory effects of IL-6 on the activation of intestinal mucins, six gastric cancer cell lines were treated for different times with several concentrations of IL-6, and the expression of MUC2 and MUC4 was evaluated. IL-6 induced MUC4 expression, detected by quantitative RT-PCR, Western blot and immunofluorescence, and MUC2 expression was not affected. MUC4 mRNA levels decreased after blocking the gp130/STAT3 pathway at the level of the receptor, and at the level of STAT3 activation using the AG490 specific inhibitor. MUC4 presents two putative binding sites for STAT factors that may regulate MUC4 transcription after a pro-inflammatory stimulus as IL-6. By EMSA, ChIP and site-directed mutagenesis we show that STAT3 binds to a cis-element at -123/-115, that conveys IL-6 mediated up-regulation of MUC4 transcriptional activity. We also demonstrated that p-STAT3 binds to MUC4 promoter and a three-fold increase in p-STAT3 binding was observed after treating GP220 cells with IL-6. In conclusion, IL-6 treatment induced MUC4 expression through the gp130/STAT3 pathway, indicating the direct role of IL-6 on the activation of the intestinal mucin gene MUC4 in gastric cancer cells.
- Published
- 2008
- Full Text
- View/download PDF
47. Changes in the invasive and metastatic capacities of HT-29/M3 cells induced by the expression of fucosyltransferase 1.
- Author
-
Mejías-Luque R, López-Ferrer A, Garrido M, Fabra A, and de Bolós C
- Subjects
- Adenocarcinoma enzymology, Colonic Neoplasms enzymology, E-Selectin metabolism, Fucosyltransferases metabolism, HT29 Cells, Humans, Neoplasm Invasiveness, Neoplasm Metastasis, Oligosaccharides analysis, Protein Binding, Sialyl Lewis X Antigen, Transfection, Galactoside 2-alpha-L-fucosyltransferase, Adenocarcinoma pathology, Colonic Neoplasms pathology, Fucosyltransferases genetics
- Abstract
Lewis antigens are terminal fucosylated oligosaccharides synthesized by the sequential action of several glycosyltransferases. The fucosyltransferases are the enzymes responsible for the addition of terminal fucose to precursor oligosaccharides attached to proteins or lipids. These oligosaccharides, defined as cell surface markers, have been implicated in different types of intercellular interactions and in adhesion and invasion processes. Transfection of HT-29/M3 colon cancer cells with the full length of human fucosyltransferase (FUT1), induces the synthesis of H type 2 and Lewis y antigens, associated with a decrease of sialyl-Lewis x. The capacity to develop primary tumors when cells were injected intrasplenically was similar in parental and FUT1-transfected cells, but the capacity to colonize the liver after spleen removal was significantly reduced in M3/FUT1 transfected cells. These results indicate that the expression of FUT1 induces changes in the metastatic capacity of HT-29/M3 colon cancer cells, as a consequence of the altered expression pattern of type 2 Lewis antigens. Also, an association between MUC5AC expression and the degree of gland differentiation in both primary splenic tumors and hepatic metastases was detected.
- Published
- 2007
- Full Text
- View/download PDF
48. Mucin genes (MUC2, MUC4, MUC5AC, and MUC6) detection in normal and pathological endometrial tissues.
- Author
-
Alameda F, Mejías-Luque R, Garrido M, and de Bolós C
- Subjects
- Female, Gene Expression Regulation, Humans, Hyperplasia genetics, Hyperplasia pathology, Neoplasms genetics, Neoplasms pathology, Endometrium cytology, Endometrium metabolism, Mucins genetics, Uterine Diseases genetics, Uterine Diseases pathology
- Abstract
Changes in the composition and physical properties of the mucous gel covering the endometrial surface are detected during the menstrual cycle and in pathological conditions. The aim of this study is to analyze the expression patterns of the 11p15 secreted mucins, MUC2, MUC5AC, and MUC6, and the membrane-bound mucin MUC4 in proliferative and secretory normal endometrium, simple and complex hyperplasia, and endometrial adenocarcinoma. A total of 98 samples, 19 of normal endometrium (11 proliferative and 8 secretor), 44 of endometrial hyperplasia (23 simple, 21 complex), and 35 of endometrial endometrioid adenocarcinomas were analyzed by immunohistochemical techniques using specific antimucin antibodies. In the endometrial proliferative glandular epithelium, only MUC4 is detected (36.3% cases). During the secretory phase, increased levels of MUC2 are found (37.5%), whereas MUC4 is less detected (12.5%). In simple hyperplasia, higher levels of mucins are expressed in the endometrial glands: MUC2 is detected in 8.7%, MUC4 in 43.4%, and MUC5AC and MUC6 in 13% of the samples, whereas in complex hyperplasia, decreased levels of mucin expression are found: MUC2 and MUC5AC are not detected, and MUC4 (28.5%) and MUC6 (20.4%) are positive. In endometrial adenocarcinoma, MUC4 is highly detected (77.1%) and increased levels of MUC5AC and MUC6 are found (61.7% and 48.5%), whereas MUC2 is poorly detected (8.5%). These findings suggest that during endometrial neoplasic transformation, increased levels of MUC4, MUC5AC, and MUC6 are detected, whereas MUC2 is only significantly detected in the secretory endometrium.
- Published
- 2007
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.