1
|
Ji X, Sun Z, Wu H, Zhang J, Liu S, Cao X, Wang B, Wang F, Zhang Y, Li B, Feng J, Zhao H. More powerful dysregulation of Helicobacter pylori East Asian-type CagA on intracellular signalings. BMC Microbiol 2024; 24:467. [PMID: 39528935 PMCID: PMC11552142 DOI: 10.1186/s12866-024-03619-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Chronic infection by Helicobacter pylori strains expressing cytotoxin-associated gene A (CagA) are the strongest risk factor for gastric cancer. CagA can be classified into East Asian-type and Western-type (CagAE and CagAW), with CagAE being more closely associated with gastric cancer. This study aimed to investigate the impact of CagAE on intracellular signaling pathways to explain its high oncogenicity. RESULTS Mutant H. pylori strains expressing either CagAE or CagAW were generated by transforming CagAE/W-expression plasmid into CagA-deleted G27 strain (G27ΔCagA). In human gastric epithelial cells (GES-1) infection, CagAE induced more severe cytopathic changes, including higher interleukin-8 (IL-8) secretion, reduced cell viability, more pronounced "hummingbird phenotype" alterations, and increased cell migration and invasion compared to CagAW. Transcriptome analysis revealed that CagAE had a stronger effect on the up-regulation of key intracellular processes, including tumor necrosis factor-ɑ (TNF-ɑ) signal pathway via nuclear factor kappa-B (NF-κB), inflammatory response, interferon-γ (IFN-γ) response, hypoxia, ultraviolet (UV) response, and Kirsten Rat Sarcoma Viral Oncogene Homolog (KRAS) signaling. A significant upregulation of hypoxia-related genes was a notable feature of CagAE. GES-1 cells infected with CagAE exhibited more severe intracellular hypoxia and higher levels of reactive oxygen species (ROS) than those infected with CagAW. Inhibition of hypoxia-inducible factor-1α (HIF-1α), which blocks hypoxia signaling, mitigated CagAE-induced cell migration, emphasizing the role of hypoxia in mediating CagAE effects. CONCLUSIONS The study provides transcriptome evidence of CagA-associated intracellular regulation during H. pylori infection, demonstrating that CagAE exerts stronger effects on intracellular signaling than CagAW. These findings offer insights into the heightened carcinogenic potential of CagAE in H. pylori-induced gastric cancer.
Collapse
Affiliation(s)
- Xiaofei Ji
- Binzhou Medical University, Yantai, China
| | - Zekun Sun
- Binzhou Medical University, Yantai, China
| | - Hao Wu
- Binzhou Medical University, Yantai, China
- Department of Blood Transfusion, Jining First People's Hospital, Jining, China
| | | | | | | | - Bin Wang
- Binzhou Medical University, Yantai, China
| | | | - Ying Zhang
- Binzhou Medical University, Yantai, China
| | - Boqing Li
- Binzhou Medical University, Yantai, China
| | | | | |
Collapse
|
2
|
Wu M, Tian C, Zou Z, Jin M, Liu H. Gastrointestinal Microbiota in Gastric Cancer: Potential Mechanisms and Clinical Applications-A Literature Review. Cancers (Basel) 2024; 16:3547. [PMID: 39456641 PMCID: PMC11506470 DOI: 10.3390/cancers16203547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 10/03/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024] Open
Abstract
Emerging evidence highlights the crucial role of gastrointestinal microbiota in the pathogenesis of gastric cancer. Helicobacter pylori (H. pylori) infection stands out as a primary pathogenic factor. However, interventions such as anti-H. pylori therapy, gastric surgeries, immunotherapy, and chronic inflammation significantly remodel the gastric microbiome, implicating a broader spectrum of microorganisms in cancer development. These microbial populations can modulate gastric carcinogenesis through various mechanisms, including sustained chronic inflammation, bacterial genotoxins, alterations in short-chain fatty acids, elevated gastrointestinal bile acids, impaired mucus barrier function, and increased concentrations of N-nitrosamines and lactic acid. The dynamic changes in gut microbiota also critically influence the outcomes of anti-cancer therapies by modifying drug bioavailability and metabolism, thus affecting therapeutic efficacy and side effect profiles. Additionally, the effectiveness of radiotherapy can be significantly impacted by gut microbiota alterations. Novel therapeutic strategies targeting the microbiome, such as dietary interventions, probiotic and synbiotic supplementation, and fecal microbiota transplantation, are showing promise in cancer treatment. Understanding the intricate relationship between the gut microbiota and gastric cancer is essential for developing new, evidence-based approaches to the prevention and treatment of this malignancy.
Collapse
Affiliation(s)
- Mengjiao Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (M.W.); (Z.Z.)
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chenjun Tian
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, China;
| | - Zhenwei Zou
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (M.W.); (Z.Z.)
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- The Eighth Hospital of Wuhan, Wuhan 430012, China
| | - Min Jin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (M.W.); (Z.Z.)
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Hongli Liu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (M.W.); (Z.Z.)
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan 430022, China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
3
|
Shirani M, Shariati S, Bazdar M, Sojoudi Ghamnak F, Moradi M, Shams Khozani R, Taki E, Arabsorkhi Z, Heidary M, Eskandari DB. The immunopathogenesis of Helicobacter pylori-induced gastric cancer: a narrative review. Front Microbiol 2024; 15:1395403. [PMID: 39035439 PMCID: PMC11258019 DOI: 10.3389/fmicb.2024.1395403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 05/06/2024] [Indexed: 07/23/2024] Open
Abstract
Helicobacter pylori infection is a well-established risk factor for the development of gastric cancer (GC). Understanding the immunopathogenesis underlying this association is crucial for developing effective preventive and therapeutic strategies. This narrative review comprehensively explores the immunopathogenesis of H. pylori-induced GC by delving into several key aspects, emphasizing the pivotal roles played by H. pylori virulence factors, including cytotoxin-associated gene A (cagA) and vacuolating cytotoxin A (vacA), blood group antigen-binding adhesin (babA), and sialic acid binding adhesin (sabA). Moreover, the review focuses on the role of toll-like receptors (TLRs) and cytokines in the complex interplay between chronic infection and gastric carcinogenesis. Finally, the study examines the association between H. pylori evasion of the innate and adaptive immune response and development of GC. A comprehensive understanding of the immunopathogenesis of H. pylori-induced GC is essential for designing targeted interventions to prevent and manage this disease. Further research is warranted to elucidate the intricate immune responses involved and identify potential therapeutic targets to improve patient outcomes.
Collapse
Affiliation(s)
- Maryam Shirani
- Toxicology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Saeedeh Shariati
- Toxicology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Monireh Bazdar
- School of Medicine, Razi Hospital, Ilam University of Medical Sciences, Ilam, Iran
| | | | - Melika Moradi
- Department of Microbiology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Elahe Taki
- Department of Microbiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zahra Arabsorkhi
- Department of Medical Genetics, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Mohsen Heidary
- Department of Laboratory Sciences, School of Paramedical Sciences, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | | |
Collapse
|
4
|
Zheng H, Xia P, Fu W, Ding S. Helicobacter pylori infection and inflammasomes. Helicobacter 2024; 29. [DOI: 10.1111/hel.13043] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 11/29/2023] [Indexed: 05/15/2025]
Abstract
AbstractHelicobacter pylori (H. pylori) causes the most prevalent bacterial infection worldwide, and more than half of the world's population is infected with H. pylori. Classified as a group 1 carcinogen of gastric cancer, H. pylori infection causes the most common chronic gastritis, which is able to progress to chronic atrophic gastritis, dysplasia, and even gastric cancer. The inflammasomes are important cytosolic multiprotein complexes to coordinate the host defense against foreign microorganisms and control the inflammatory response. It is also well‐known that inflammasome plays an important role in the occurrence of H. pylori‐induced gastric inflammation. During infection and inflammation, the activation process of inflammasome is tightly regulated by host immune system. However, excessive activation of inflammasome is closely related to the production of excessive cytokines that cause the body injury and resulting in various inflammatory diseases. In this review, we elaborate the activation and assembly mechanisms of inflammasome, the structure of different inflammasome complexes, host factors in vivo and drugs in vitro that regulate inflammasome signaling during H. pylori infection, aiming to provide novel insights and strategies for identifying new therapeutic targets for the treatment of H. pylori‐associated gastric mucosal diseases.
Collapse
Affiliation(s)
- Huiling Zheng
- Department of Gastroenterology Peking University Third Hospital Beijing China
- Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases (BZ0371) Beijing China
| | - Pengyan Xia
- Department of Immunology, School of Basic Medical Sciences Peking University Beijing China
| | - Weiwei Fu
- Department of Gastroenterology Peking University Third Hospital Beijing China
- Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases (BZ0371) Beijing China
| | - Shigang Ding
- Department of Gastroenterology Peking University Third Hospital Beijing China
- Beijing Key Laboratory for Helicobacter Pylori Infection and Upper Gastrointestinal Diseases (BZ0371) Beijing China
| |
Collapse
|
5
|
Jaroń K, Pietrzak A, Daniluk J, Adrych K, Gąsiorowska A, Skrzydło-Radomańska B, Małecka-Wojciesko E, Zwolińska-Wcisło M, Waluga M, Reguła J, Rydzewska G. Diagnostic and therapeutic recommendations on Helicobacter pylori infection. Recommendations of the Working Group of the Polish Society of Gastroenterology. PRZEGLAD GASTROENTEROLOGICZNY 2023; 18:225-248. [PMID: 37937106 PMCID: PMC10626381 DOI: 10.5114/pg.2023.131998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 09/27/2023] [Indexed: 11/09/2023]
Abstract
Helicobacter pylori remains a major health problem worldwide, causing considerable morbidity and mortality due to peptic ulcer disease and gastric cancer. These guidelines constitute an update of the previous "Recommendations on the diagnosis and management of Helicobacter pylori infection" issued in 2014. They have been developed by a Task Force organized by the Governing Board of the Polish Society of Gastroenterology. They discuss, with particular emphasis on new scientific data covering papers published since 2014: the epidemiology, clinical presentation, diagnostic principles and criteria for the diagnosis, and recommendations for the treatment of H. pylori infection. The guidelines in particular determine which patients need to be tested and treated for infection. The Task Force also discussed recommended treatment algorithms. Accordingly, a combination of available evidence and consensus-based expert opinion were used to develop these best practice advice statements. It is worth noting that guidelines are not mandatory to implement but they offer advice for pragmatic, relevant and achievable diagnostic and treatment pathways based on established key treatment principles and using local knowledge and available resources to guide regional practice.
Collapse
Affiliation(s)
- Krzysztof Jaroń
- Department of Internal Medicine and Gastroenterology and Subdivision for Treatment of Inflammatory Bowel Diseases, the National Medical Institute of the Ministry of Internal Affairs and Administration, Warsaw, Poland
| | - Anna Pietrzak
- Second Gastroenterology Clinic, Medical Centre for Postgraduate Education, Warsaw, Poland
- Gastroenterology Department, Bielański Hospital, Warsaw, Poland
| | - Jarosław Daniluk
- Department of Gastroenterology and Internal Diseases, Medical University of Bialystok, Bialystok, Poland
| | - Krystian Adrych
- Department of Gastroenterology and Hepatology, Medical University of Gdansk, Gdansk, Poland
| | - Anita Gąsiorowska
- Department of Gastroenterology, Central Clinical Hospital of the Medical University, Lodz, Poland
| | | | | | | | - Marek Waluga
- Department of Gastroenterology and Hepatology, Prof. Kornel Gibiński University Clinical Centre, Medical University of Silesia, Katowice, Poland
| | - Jarosław Reguła
- Department of Oncological Gastroenterology, Maria Skłodowska-Curie National Cancer Institute – National Research Institute, Warsaw, Poland
- Department of Gastroenterology, Hepatology and Clinical Oncology, Medical Centre for Postgraduate Education, Warsaw, Poland
| | - Grażyna Rydzewska
- Department of Internal Medicine and Gastroenterology and Subdivision for Treatment of Inflammatory Bowel Diseases, the National Medical Institute of the Ministry of Internal Affairs and Administration, Warsaw, Poland
- Department of Gastrointestinal Disease Prevention, Faculty of Medicine and Health Sciences, Jan Kochanowski University, Kielce, Poland
| |
Collapse
|
6
|
Jamal Eddin TM, Nasr SM, Gupta I, Zayed H, Al Moustafa AE. Helicobacter pylori and epithelial mesenchymal transition in human gastric cancers: An update of the literature. Heliyon 2023; 9:e18945. [PMID: 37609398 PMCID: PMC10440535 DOI: 10.1016/j.heliyon.2023.e18945] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/25/2023] [Accepted: 08/03/2023] [Indexed: 08/24/2023] Open
Abstract
Gastric cancer, a multifactorial disease, is considered one of the most common malignancies worldwide. In addition to genetic and environmental risk factors, infectious agents, such as Epstein-Barr virus (EBV) and Helicobacter pylori (H.pylori) contribute to the onset and development of gastric cancer. H. pylori is a type I carcinogen that colonizes the gastric epithelium of approximately 50% of the world's population, thus increasing the risk of gastric cancer development. On the other hand, epithelial mesenchymal transition (EMT) is a fundamental process crucial to embryogenic growth, wound healing, organ fibrosis and cancer progression. Several studies associate gastric pathogen infection of the epithelium with EMT initiation, provoking cancer metastasis in the gastric mucosa through various molecular signaling pathways. Additionally, EMT is implicated in the progression and development of H. pylori-associated gastric cancer. In this review, we recapitulate recent findings elucidating the association between H. pylori infection in EMT promotion leading to gastric cancer progression and metastasis.
Collapse
Affiliation(s)
- Tala M. Jamal Eddin
- College of Health Sciences, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Shahd M.O. Nasr
- College of Health Sciences, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Ishita Gupta
- College of Medicine, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Hatem Zayed
- College of Health Sciences, QU Health, Qatar University, PO Box 2713, Doha, Qatar
| | - Ala-Eddin Al Moustafa
- College of Medicine, QU Health, Qatar University, PO Box 2713, Doha, Qatar
- Biomedical Research Center, Qatar University, PO Box 2713, Doha, Qatar
- Oncology Department, Faculty of Medicine, McGill University, Montreal, QC, H3G 2M1, Canada
| |
Collapse
|
7
|
Kashyap D, Rele S, Bagde PH, Saini V, Chatterjee D, Jain AK, Pandey RK, Jha HC. Comprehensive insight into altered host cell-signaling cascades upon Helicobacter pylori and Epstein-Barr virus infections in cancer. Arch Microbiol 2023; 205:262. [PMID: 37310490 DOI: 10.1007/s00203-023-03598-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 06/14/2023]
Abstract
Cancer is characterized by mutagenic events that lead to disrupted cell signaling and cellular functions. It is one of the leading causes of death worldwide. Literature suggests that pathogens, mainly Helicobacter pylori and Epstein-Barr virus (EBV), have been associated with the etiology of human cancer. Notably, their co-infection may lead to gastric cancer. Pathogen-mediated DNA damage could be the first and crucial step in the carcinogenesis process that modulates numerous cellular signaling pathways. Altogether, it dysregulates the metabolic pathways linked with cell growth, apoptosis, and DNA repair. Modulation in these pathways leads to abnormal growth and proliferation. Several signaling pathways such RTK, RAS/MAPK, PI3K/Akt, NFκB, JAK/STAT, HIF1α, and Wnt/β-catenin are known to be altered in cancer. Therefore, this review focuses on the oncogenic roles of H. pylori, EBV, and its associated signaling cascades in various cancers. Scrutinizing these signaling pathways is crucial and may provide new insights and targets for preventing and treating H. pylori and EBV-associated cancers.
Collapse
Affiliation(s)
- Dharmendra Kashyap
- Lab No. POD 1B 602, Infection Bio-Engineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, Madhya Pradesh, 453552, India
| | - Samiksha Rele
- Lab No. POD 1B 602, Infection Bio-Engineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, Madhya Pradesh, 453552, India
| | - Pranit Hemant Bagde
- Lab No. POD 1B 602, Infection Bio-Engineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, Madhya Pradesh, 453552, India
| | - Vaishali Saini
- Lab No. POD 1B 602, Infection Bio-Engineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, Madhya Pradesh, 453552, India
| | | | | | - Rajan Kumar Pandey
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, 17177, Solna, Sweden
| | - Hem Chandra Jha
- Lab No. POD 1B 602, Infection Bio-Engineering Group, Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, Madhya Pradesh, 453552, India.
- Centre for Rural Development and Technology, Indian Institute of Technology Indore, Madhya Pradesh, 453552, Indore, India.
| |
Collapse
|
8
|
Li H, Lin J, Cheng S, Chi J, Luo J, Tang Y, Zhao W, Shu Y, Liu X, Xu C. Comprehensive analysis of differences in N6-methyladenosine RNA methylomes in Helicobacter pylori infection. Front Cell Dev Biol 2023; 11:1136096. [PMID: 37363723 PMCID: PMC10289286 DOI: 10.3389/fcell.2023.1136096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 04/13/2023] [Indexed: 06/28/2023] Open
Abstract
Background: Helicobacter pylori (H.pylori) infection is an important factor in the occurrence of human gastric diseases, but its pathogenic mechanism is not clear. N6-methyladenosine (m6A) is the most prevalent reversible methylation modification in mammalian RNA and it plays a crucial role in controlling many biological processes. However, there are no studies reported that whether H. pylori infection impacts the m6A methylation of stomach. In this study, we measured the overall level changes of m6A methylation of RNA under H. pylori infection through in vitro and in vivo experiment. Methods: The total quantity of m6A was quantified in gastric tissues of clinical patients and C57 mice with H. pylori infection, as well as acute infection model [H. pylori and GES-1 cells were cocultured for 48 h at a multiplicity of infection (MOI) from of 10:1 to 50:1]. Furthermore, we performed m6A methylation sequencing and RNA-sequencing on the cell model and RNA-sequencing on animal model. Results: Quantitative detection of RNA methylation showed that H. pylori infection group had higher m6A modification level. M6A methylation sequencing identified 2,107 significantly changed m6A methylation peaks, including 1,565 upregulated peaks and 542 downregulated peaks. A total of 2,487 mRNA was upregulated and 1,029 mRNA was downregulated. According to the comprehensive analysis of MeRIP-seq and RNA-seq, we identified 200 hypermethylation and upregulation, 129 hypermethylation but downregulation, 19 hypomethylation and downregulation and 106 hypomethylation but upregulation genes. The GO and KEGG pathway analysis of these differential methylation and regulatory genes revealed a wide range of biological functions. Moreover, combining with mice RNA-seq results, qRT- PCR showed that m6A regulators, METTL3, WTAP, FTO and ALKBH5, has significant difference; Two key genes, PTPN14 and ADAMTS1, had significant difference by qRT- PCR. Conclusion: These findings provide a basis for further investigation of the role of m6A methylation modification in H. pylori-associated gastritis.
Collapse
Affiliation(s)
- Huan Li
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jiahui Lin
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Sha Cheng
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Jingshu Chi
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Ju Luo
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yu Tang
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Wenfang Zhao
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Yufeng Shu
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xiaoming Liu
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Central South University, Changsha, Hunan, China
| | - Canxia Xu
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Non-Resolving Inflammation and Cancer, Central South University, Changsha, Hunan, China
| |
Collapse
|
9
|
Zhu S, Al-Mathkour M, Cao L, Khalafi S, Chen Z, Poveda J, Peng D, Lu H, Soutto M, Hu T, McDonald OG, Zaika A, El-Rifai W. CDK1 bridges NF-κB and β-catenin signaling in response to H. pylori infection in gastric tumorigenesis. Cell Rep 2023; 42:112005. [PMID: 36681899 PMCID: PMC9973518 DOI: 10.1016/j.celrep.2023.112005] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 10/31/2022] [Accepted: 01/03/2023] [Indexed: 01/22/2023] Open
Abstract
Infection with Helicobacter pylori (H. pylori) is the main risk factor for gastric cancer, a leading cause of cancer-related death worldwide. The oncogenic functions of cyclin-dependent kinase 1 (CDK1) are not fully understood in gastric tumorigenesis. Using public datasets, quantitative real-time PCR, western blot, and immunohistochemical (IHC) analyses, we detect high levels of CDK1 in human and mouse gastric tumors. H. pylori infection induces activation of nuclear factor κB (NF-κB) with a significant increase in CDK1 in in vitro and in vivo models (p < 0.01). We confirm active NF-κB binding sites on the CDK1 promoter sequence. CDK1 phosphorylates and inhibits GSK-3β activity through direct binding with subsequent accumulation and activation of β-catenin. CDK1 silencing or pharmacologic inhibition reverses these effects and impairs tumor organoids and spheroid formation. IHC analysis demonstrates a positive correlation between CDK1 and β-catenin. The results demonstrate a mechanistic link between infection, inflammation, and gastric tumorigenesis where CDK1 plays a critical role.
Collapse
Affiliation(s)
- Shoumin Zhu
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Marwah Al-Mathkour
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Longlong Cao
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA; Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, China
| | - Shayan Khalafi
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Zheng Chen
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA; Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Julio Poveda
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Dunfa Peng
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Heng Lu
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Mohammed Soutto
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Tianling Hu
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Oliver G McDonald
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Alexander Zaika
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA; Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, USA; Department of Veterans Affairs, Miami Healthcare System, Miami, FL, USA
| | - Wael El-Rifai
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, USA; Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL, USA; Department of Veterans Affairs, Miami Healthcare System, Miami, FL, USA.
| |
Collapse
|
10
|
Song MY, Lee DY, Han YM, Kim EH. Anti-Inflammatory Effect of Korean Propolis on Helicobacter pylori-Infected Gastric Mucosal Injury Mice Model. Nutrients 2022; 14:nu14214644. [PMID: 36364906 PMCID: PMC9659254 DOI: 10.3390/nu14214644] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 10/28/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022] Open
Abstract
Propolis, a natural resinous substance obtained from a variety of buds and plants, has been reported to possess various biological functions. Several recent studies have demonstrated the inhibitory effects of propolis on the growth of Helicobacter pylori (H. pylori) in vitro; however, current research efforts on Korean propolis (KP) remain insufficient especially in vivo. Our study aims to investigate the anti-inflammatory effect and molecular mechanism of KP on mouse gastric mucosa during H. pylori infection. We examined an in vivo H. pylori-induced gastric mucosal injury mice model. We found that KP inhibited the growth of H. pylori and attenuated the expression of H. pylori virulence factors such as cytotoxin-associated gene A, encoding urease A subunit, surface antigen gene and neutrophil-activating protein A. Moreover, KP reduced both gross lesions and pathological scores in H. pylori-challenged mice. In addition, KP markedly restrained the production of pro-inflammatory cytokines and nitric oxide levels compared with an untreated H. pylori-infected group. In particular, we found that KP repressed the phosphorylation of IκBα and NF-κB p65 subunit, and subsequently suppressed their downstream target genes. Taken together, these findings demonstrate the beneficial effects of KP on inflammation through the inhibition of NF-κB signaling as well as inhibition of H. pylori growth in a mouse model infected with H. pylori. This suggests the potential application of KP as a natural supplement for patient’s suffering from gastric mucosal injury caused by H. pylori infection.
Collapse
|
11
|
Yang E, Chua W, Ng W, Roberts TL. Peripheral Cytokine Levels as a Prognostic Indicator in Gastric Cancer: A Review of Existing Literature. Biomedicines 2021; 9:1916. [PMID: 34944729 PMCID: PMC8698340 DOI: 10.3390/biomedicines9121916] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/07/2021] [Accepted: 12/07/2021] [Indexed: 12/16/2022] Open
Abstract
Although strong connections exist between the carcinogenesis of gastric cancer and chronic inflammation, gastric cancer is unique in that the chronic gastritis which frequently precedes carcinogenesis is strongly associated with H. pylori infection. The interplay between H. pylori virulence factors and host immune cells is complex but culminates in the activation of inflammatory pathways and transcription factors such as NF-κB, STAT3, and AP-1, all of which upregulate cytokine production. Due to the key role of cytokines in modulating the immune response against tumour cells as well as possibly stimulating tumour growth and proliferation, different patterns of cytokine secretion may be associated with varying patient outcomes. In relation to gastric cancer, interleukin-6, 8, 10, 17A, TNF, and IFN-γ may have pro-tumour properties, although interleukin-10, TNF, and IFN-γ may have anti-tumour effects. However, due to the lack of studies investigating patient outcomes, only a link between higher interleukin-6 levels and poorer prognosis has been demonstrated. Further investigations which link peripheral cytokine levels to patient prognosis may elucidate important pathological mechanisms in gastric cancer which adversely impact patient survival and allow treatments targeting these processes to be developed.
Collapse
Affiliation(s)
- Elton Yang
- School of Medicine, Western Sydney University, Campbelltown 2560, Australia; (E.Y.); (W.C.); (W.N.)
- Ingham Institute for Applied Medical Research, Liverpool 2170, Australia
| | - Wei Chua
- School of Medicine, Western Sydney University, Campbelltown 2560, Australia; (E.Y.); (W.C.); (W.N.)
- Ingham Institute for Applied Medical Research, Liverpool 2170, Australia
- Medical Oncology, Liverpool Hospital, Liverpool 2170, Australia
- Southwest Sydney Clinical School, University of New South Wales, Liverpool 2170, Australia
| | - Weng Ng
- School of Medicine, Western Sydney University, Campbelltown 2560, Australia; (E.Y.); (W.C.); (W.N.)
- Ingham Institute for Applied Medical Research, Liverpool 2170, Australia
- Medical Oncology, Liverpool Hospital, Liverpool 2170, Australia
- Southwest Sydney Clinical School, University of New South Wales, Liverpool 2170, Australia
| | - Tara Laurine Roberts
- School of Medicine, Western Sydney University, Campbelltown 2560, Australia; (E.Y.); (W.C.); (W.N.)
- Ingham Institute for Applied Medical Research, Liverpool 2170, Australia
- Southwest Sydney Clinical School, University of New South Wales, Liverpool 2170, Australia
| |
Collapse
|
12
|
Taxauer K, Hamway Y, Ralser A, Dietl A, Mink K, Vieth M, Singer BB, Gerhard M, Mejías-Luque R. Engagement of CEACAM1 by Helicobacterpylori HopQ Is Important for the Activation of Non-Canonical NF-κB in Gastric Epithelial Cells. Microorganisms 2021; 9:1748. [PMID: 34442827 PMCID: PMC8400456 DOI: 10.3390/microorganisms9081748] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/04/2021] [Accepted: 08/10/2021] [Indexed: 01/01/2023] Open
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.
Collapse
Affiliation(s)
- Karin Taxauer
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University Munich, 81675 Munich, Germany; (K.T.); (Y.H.); (A.R.); (A.D.); (K.M.); (M.G.)
| | - Youssef Hamway
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University Munich, 81675 Munich, Germany; (K.T.); (Y.H.); (A.R.); (A.D.); (K.M.); (M.G.)
| | - Anna Ralser
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University Munich, 81675 Munich, Germany; (K.T.); (Y.H.); (A.R.); (A.D.); (K.M.); (M.G.)
| | - Alisa Dietl
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University Munich, 81675 Munich, Germany; (K.T.); (Y.H.); (A.R.); (A.D.); (K.M.); (M.G.)
| | - Karin Mink
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University Munich, 81675 Munich, Germany; (K.T.); (Y.H.); (A.R.); (A.D.); (K.M.); (M.G.)
| | - Michael Vieth
- Institute of Pathology, Friedrich-Alexander University Erlangen-Nuremberg, Klinikum Bayreuth, 95445 Bayreuth, Germany;
| | - Bernhard B. Singer
- Institute of Anatomy, Medical Faculty, University of Duisburg-Essen, 45147 Essen, Germany;
| | - Markus Gerhard
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University Munich, 81675 Munich, Germany; (K.T.); (Y.H.); (A.R.); (A.D.); (K.M.); (M.G.)
| | - Raquel Mejías-Luque
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University Munich, 81675 Munich, Germany; (K.T.); (Y.H.); (A.R.); (A.D.); (K.M.); (M.G.)
| |
Collapse
|
13
|
Evaluation of miR-21 Expression Level in Helicobacter pylori-Infected Gastric Mucosa. Jundishapur J Microbiol 2020. [DOI: 10.5812/jjm.100724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background: Gastric cancer is one of the main causes of death worldwide. In this regard, Helicobacter pylori infection is considered as the main risk factor for gastric cancer. MicroRNA (mirNA) can interface with mRNA molecules as well as blocking their translation into proteins or inducing degradation. Objectives: The aim of this study was to compare the expression of mir-21 in biopsy samples of gastritis and healthy adjacent tissues. Methods: Between Feb-Dec 2017, 70 patients with dyspeptic symptoms from Taleghani Hospital were enrolled in this study. Accordingly, the expression level of mir-21 was evaluated using semi-quantitative RT-PCR in mucosal biopsy samples from those well-characterized patients. Moreover, the U6 gene was used as an internal control. Results: Our data indicated that mir-21 expression was significantly up-regulated in the infected samples with H. pylori compared to healthy samples. Conclusions: Our results confirm that H. pylori infection can alter the expression of mir-21 in gastric epithelial cells and gastric mucosal tissues. However, the exact role of the miRNA changes in H. pylori infection will require further experiments.
Collapse
|
14
|
Chen Y, Sheppard D, Dong X, Hu X, Chen M, Chen R, Chakrabarti J, Zavros Y, Peek RM, Chen LF. H. pylori infection confers resistance to apoptosis via Brd4-dependent BIRC3 eRNA synthesis. Cell Death Dis 2020; 11:667. [PMID: 32820150 PMCID: PMC7441315 DOI: 10.1038/s41419-020-02894-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 07/28/2020] [Accepted: 07/29/2020] [Indexed: 02/07/2023]
Abstract
H. pylori infection is one of the leading causes of gastric cancer and the pathogenicity of H. pylori infection is associated with its ability to induce chronic inflammation and apoptosis resistance. While H. pylori infection-induced expression of pro-inflammatory cytokines for chronic inflammation is well studied, the molecular mechanism underlying the apoptosis resistance in infected cells is not well understood. In this study, we demonstrated that H. pylori infection-induced apoptosis resistance in gastric epithelial cells triggered by Raptinal, a drug that directly activates caspase-3. This resistance resulted from the induction of cIAP2 (encoded by BIRC3) since depletion of BIRC3 by siRNA or inhibition of cIAP2 via BV6 reversed H. pylori-suppressed caspase-3 activation. The induction of cIAP2 was regulated by H. pylori-induced BIRC3 eRNA synthesis. Depletion of BIRC3 eRNA decreased H. pylori-induced cIAP2 and reversed H. pylori-suppressed caspase-3 activation. Mechanistically, H. pylori stimulated the recruitment of bromodomain-containing factor Brd4 to the enhancer of BIRC3 and promoted BIRC3 eRNA and mRNA synthesis. Inhibition of Brd4 diminished the expression of BIRC3 eRNA and the anti-apoptotic response to H. pylori infection. Importantly, H. pylori isogenic cagA-deficient mutant failed to activate the synthesis of BIRC3 eRNA and the associated apoptosis resistance. Finally, in primary human gastric epithelial cells, H. pylori also induced resistance to Raptinal-triggered caspase-3 activation by activating the Brd4-dependent BIRC3 eRNA synthesis in a CagA-dependent manner. These results identify a novel function of Brd4 in H. pylori-mediated apoptosis resistance via activating BIRC3 eRNA synthesis, suggesting that Brd4 could be a potential therapeutic target for H. pylori-induced gastric cancer.
Collapse
Affiliation(s)
- Yanheng Chen
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, 61801, IL, USA
| | - Donald Sheppard
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, 61801, IL, USA
| | - Xingchen Dong
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, 61801, IL, USA
| | - Xiangming Hu
- Fujian Key Laboratory for Translational Research in Cancer and Neurodegenerative Diseases, Institute for Translational Medicine, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, China
| | - Meihua Chen
- The State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, 361101, China
| | - Ruichuan Chen
- The State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, 361101, China
| | - Jayati Chakrabarti
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, 45267, OH, USA
- Department of Cellular and Molevular Medicine, College of Medicine, University of Arizona-Tucson, Tucson, 85724, AZ, USA
| | - Yana Zavros
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, 45267, OH, USA
- Department of Cellular and Molevular Medicine, College of Medicine, University of Arizona-Tucson, Tucson, 85724, AZ, USA
| | - Richard M Peek
- Division of Gastroenterology, Department of Medicine and Cancer Biology, Vanderbilt University School of Medicine, Nashville, 37232, TN, USA
| | - Lin-Feng Chen
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, 61801, IL, USA.
- Carle R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, 61801, IL, USA.
| |
Collapse
|
15
|
Dang Y, Zhang Y, Xu L, Zhou X, Gu Y, Yu J, Jin S, Ji H, Shu Y, Zhang G, Cui S, Sun J. PUMA-mediated epithelial cell apoptosis promotes Helicobacter pylori infection-mediated gastritis. Cell Death Dis 2020; 11:139. [PMID: 32080167 PMCID: PMC7033162 DOI: 10.1038/s41419-020-2339-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 02/08/2020] [Accepted: 02/10/2020] [Indexed: 01/23/2023]
Abstract
The molecular mechanism responsible for Helicobacter pylori infection-mediated gastritis and carcinogenesis is not yet clear. Increased evidence suggests that chronic gastritis and elevated gastric epithelial cell (GEC) apoptosis are crucial events during stomach carcinoma transformation. PUMA is a potent proapoptotic Bcl-2 protein and mediates acute tissue injury. In this study, we aimed to investigate the role of PUMA in GEC apoptosis and inflammation induced by H. pylori infection. As a result, we found that PUMA expression was elevated in gastritis tissues compared with uninvolved tissues, and it was correlated with the severity of apoptosis and gastritis. In mice, PUMA mRNA and protein were markedly induced in GECs upon induction of gastritis by H. pylori. PUMA-deficient mice were highly resistant to apoptosis and gastritis induced by H. pylori. Furthermore, the transcription factor NF-κB p65 binds to PUMA promoter to activate PUMA transcription after H. pylori infection. In addition, NF-κB inhibitor could rescue H. pylori-induced apoptosis and gastritis. Finally, H. pylori-induced activation of p-p65 and PUMA was mediated via Toll-like receptor 2 (TLR2) and blocked in TLR2 knockout mice. Taken together, these results verified the pro-inflammatory effect of PUMA in H. pylori-infected gastric tissue. Moreover, TLR2/NF-κB-mediated transcriptional regulation of PUMA contributes to the pathogenesis of H. pylori-infected gastritis.
Collapse
Affiliation(s)
- Yini Dang
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yifeng Zhang
- Department of Gastroenterology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210001, China
| | - Lingyan Xu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xiaoying Zhou
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yanhong Gu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Jian Yu
- Department of Pathology and Radiation Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA
| | - Shidai Jin
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Haoming Ji
- Department of Oncology, Haian People's Hospital, Nantong, 226630, China
| | - Yongqian Shu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Guoxin Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Shiyun Cui
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Jing Sun
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
16
|
Sebrell TA, Hashimi M, Sidar B, Wilkinson RA, Kirpotina L, Quinn MT, Malkoç Z, Taylor PJ, Wilking JN, Bimczok D. A Novel Gastric Spheroid Co-culture Model Reveals Chemokine-Dependent Recruitment of Human Dendritic Cells to the Gastric Epithelium. Cell Mol Gastroenterol Hepatol 2019; 8:157-171.e3. [PMID: 30878664 PMCID: PMC6599165 DOI: 10.1016/j.jcmgh.2019.02.010] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 02/13/2019] [Accepted: 02/20/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND & AIMS Gastric dendritic cells (DCs) control the adaptive response to infection with Helicobacter pylori, a major risk factor for peptic ulcer disease and gastric cancer. We hypothesize that DC interactions with the gastric epithelium position gastric DCs for uptake of luminal H pylori and promote DC responses to epithelial-derived mediators. The aim of this study was to determine whether the gastric epithelium actively recruits DCs using a novel co-culture model of human gastric epithelial spheroids and monocyte-derived DCs. METHODS Spheroid cultures of primary gastric epithelial cells were infected with H pylori by microinjection. Co-cultures were established by adding human monocyte-derived DCs to the spheroid cultures and were analyzed for DC recruitment and antigen uptake by confocal microscopy. Protein array, gene expression polymerase chain reaction array, and chemotaxis assays were used to identify epithelial-derived chemotactic factors that attract DCs. Data from the co-culture model were confirmed using human gastric tissue samples. RESULTS Human monocyte-derived DCs co-cultured with gastric spheroids spontaneously migrated to the gastric epithelium, established tight interactions with the epithelial cells, and phagocytosed luminally applied H pylori. DC recruitment was increased upon H pylori infection of the spheroids and involved the activity of multiple chemokines including CXCL1, CXCL16, CXCL17, and CCL20. Enhanced chemokine expression and DC recruitment to the gastric epithelium also was observed in H pylori-infected human gastric tissue samples. CONCLUSIONS Our results indicate that the gastric epithelium actively recruits DCs for immunosurveillance and pathogen sampling through chemokine-dependent mechanisms, with increased recruitment upon active H pylori infection.
Collapse
Affiliation(s)
- Thomas A Sebrell
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana
| | - Marziah Hashimi
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana
| | - Barkan Sidar
- Department of Chemical and Biological Engineering and Center for Biofilm Engineering, Montana State University, Bozeman, Montana
| | - Royce A Wilkinson
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana
| | - Liliya Kirpotina
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana
| | - Mark T Quinn
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana
| | - Zeynep Malkoç
- Department of Chemical and Biological Engineering and Center for Biofilm Engineering, Montana State University, Bozeman, Montana
| | | | - James N Wilking
- Department of Chemical and Biological Engineering and Center for Biofilm Engineering, Montana State University, Bozeman, Montana
| | - Diane Bimczok
- Department of Microbiology and Immunology, Montana State University, Bozeman, Montana.
| |
Collapse
|
17
|
Hu Y, Liu JP, Li XY, Cai Y, He C, Li NS, Xie C, Xiong ZJ, Ge ZM, Lu NH, Zhu Y. Downregulation of tumor suppressor RACK1 by Helicobacter pylori infection promotes gastric carcinogenesis through the integrin β-1/NF-κB signaling pathway. Cancer Lett 2019; 450:144-154. [PMID: 30849478 DOI: 10.1016/j.canlet.2019.02.039] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/12/2019] [Accepted: 02/13/2019] [Indexed: 12/24/2022]
Abstract
Receptor of activated protein kinase C 1 (RACK1) is downregulated in gastric cancer and is involved in modulating NF-κB signaling pathway activity. However, the underlying molecular mechanisms regulating RACK1 expression are unclear. In this study, we demonstrated that downregulated expression of RACK1 was observed in gastric cancer tissue compared to adjacent normal tissue and was correlated with poor prognosis in patients. Helicobacter pylori (H. pylori) infection downregulated RACK1 expression in concert with canonical NF-κB signaling pathway activation in vivo and in vitro. RACK1 overexpression suppressed NF-κB signaling pathway activation as well as the release of downstream proinflammatory cytokines. In addition, RACK1 downregulation increased integrin β-1 expression, while integrin β-1 silencing decreased NF-κB signaling activation. Moreover, H. pylori infection downregulated RACK1 but upregulated integrin β-1 expression at the precancerous lesion stages in human subjects. Our data indicate that H. pylori infection promotes the upregulation of integrin β-1 expression via downregulation of RACK1 expression, which subsequently leads to the elevated activation of the NF-κB signaling pathway, an essential step in H. pylori-induced carcinogenesis.
Collapse
Affiliation(s)
- Yi Hu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China.
| | - Jian-Ping Liu
- Integrated Cardio Metabolic Centre, Karolinska Institute, Huddinge, Sweden.
| | - Xue-Yang Li
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China.
| | - Yan Cai
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China.
| | - Cong He
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China.
| | - Nian-Shuang Li
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China.
| | - Chuan Xie
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China.
| | - Zhi-Juan Xiong
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China.
| | - Zhong-Ming Ge
- Division of Comparative Medicine, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA, 02139, USA.
| | - Nong-Hua Lu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China.
| | - Yin Zhu
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China.
| |
Collapse
|
18
|
Kidane D. Molecular Mechanisms of H. pylori-Induced DNA Double-Strand Breaks. Int J Mol Sci 2018; 19:ijms19102891. [PMID: 30249046 PMCID: PMC6213211 DOI: 10.3390/ijms19102891] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 09/11/2018] [Accepted: 09/21/2018] [Indexed: 12/17/2022] Open
Abstract
Infections contribute to carcinogenesis through inflammation-related mechanisms. H. pylori infection is a significant risk factor for gastric carcinogenesis. However, the molecular mechanism by which H. pylori infection contributes to carcinogenesis has not been fully elucidated. H. pylori-associated chronic inflammation is linked to genomic instability via reactive oxygen and nitrogen species (RONS). In this article, we summarize the current knowledge of H. pylori-induced double strand breaks (DSBs). Furthermore, we provide mechanistic insight into how processing of oxidative DNA damage via base excision repair (BER) leads to DSBs. We review recent studies on how H. pylori infection triggers NF-κB/inducible NO synthase (iNOS) versus NF-κB/nucleotide excision repair (NER) axis-mediated DSBs to drive genomic instability. This review discusses current research findings that are related to mechanisms of DSBs and repair during H. pylori infection.
Collapse
Affiliation(s)
- Dawit Kidane
- Division of Pharmacology and Toxicology, College of Pharmacy, The University of Texas at Austin, Dell Pediatric Research Institute, 1400 Barbara Jordan Blvd. R1800, Austin, TX 78723, USA.
| |
Collapse
|
19
|
Molecular Mechanisms of Natural Honey Against H. pylori Infection Via Suppression of NF-κB and AP-1 Activation in Gastric Epithelial Cells. Arch Med Res 2017; 47:340-348. [PMID: 27751367 DOI: 10.1016/j.arcmed.2016.09.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2016] [Accepted: 09/02/2016] [Indexed: 12/31/2022]
Abstract
BACKGROUND AND AIMS Natural honey has been used as a medicine since ancient times. Honey is widely known for its antibacterial properties against H. pylori; however, the mechanisms of its antibacterial activity are not fully known. The present study was performed to examine the molecular mechanisms by which natural honey can inhibit H. pylori infection in gastric epithelial cells. METHODS Electrophoretic mobility shift assay was used to measure NF-κB- and AP-1-DNA binding activity. Western blotting was used to detect IκB-α and COX-2 expression. RESULTS H. pylori induced NF-κB and AP-1 DNA-binding activity in gastric epithelial cells. Manuka honey inhibited H. pylori-induced NF-κB and AP-1 in a time- and dose-dependent manner. Maximum inhibition of H. pylori-induced NF-κB and AP-1 by manuka honey was observed at concentrations of 20% at 1-2 h. Pre-treatment of AGS cells with other commercial natural honeys also inhibited H. pylori-induced NF-κB and AP-1 DNA-binding activity. Honey prevented H. pylori-induced degradation of IκB-α protein and downregulated COX-2 protein levels. CONCLUSIONS Our findings suggest that natural honey exerts its inhibitory effects against H. pylori by inhibiting NF-κB and AP-1 activation and downregulation of COX-2 expression. These results provide new mechanistic insights into honey effects in the suppression of H. pylori infection.
Collapse
|
20
|
Luo B, Wang M, Hou N, Hu X, Jia G, Qin X, Zuo X, Liu Y, Luo K, Song W, Wang K, Pang M. ATP-Dependent Lon Protease Contributes to Helicobacter pylori-Induced Gastric Carcinogenesis. Neoplasia 2017; 18:242-52. [PMID: 27108387 PMCID: PMC4840290 DOI: 10.1016/j.neo.2016.03.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 02/18/2016] [Accepted: 03/01/2016] [Indexed: 12/20/2022] Open
Abstract
Helicobacter pylori infection is the strongest risk factor for development of gastric cancer. Host cellular stress responses, including inflammatory and immune responses, have been reported highly linked to H. pylori-induced carcinogenesis. However, whether mitochondrial regulation and metabolic reprogramming, which are potently associated with various cancers, play a role in H. pylori-induced gastric carcinogenesis is largely unknown. Here we revealed that Lon protease (Lonp1), which is a key inductive of mitochondrial unfolded protein response (UPR(mt)) and is required to maintain the mitochondrial quality, was greatly induced in H. pylori infected gastric epithelial cells. Importantly, we uncovered that knockdown of Lonp1 expression significantly diminished the metabolic switch to glycolysis and gastric cell proliferation associated with low multiplicity of H. pylori infection. In addition, Lonp1 overexpression in gastric epithelial cells also promoted glycolytic switch and cell overgrowth, suggesting H. pylori effect is Lonp1 dependent. We further demonstrated that H. pylori induced Lonp1 expression and cell overgrowth, at least partially, via HIF-1α regulation. Collectively, our results concluded the relevance of Lonp1 for cell proliferation and identified Lonp1 as a key regulator of metabolic reprogramming in H. pylori-induced gastric carcinogenesis.
Collapse
Affiliation(s)
- Bin Luo
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu 610072, People's Republic of China
| | - Minggang Wang
- Department of Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 100043, People's Republic of China
| | - Nengyi Hou
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu 610072, People's Republic of China
| | - Xiao Hu
- Department of Gastroenterology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu 610072, People's Republic of China
| | - Guiqing Jia
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu 610072, People's Republic of China
| | - Xianpeng Qin
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu 610072, People's Republic of China
| | - Xiaofei Zuo
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu 610072, People's Republic of China
| | - Yang Liu
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu 610072, People's Republic of China
| | - Kun Luo
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu 610072, People's Republic of China
| | - Wei Song
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Kang Wang
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu 610072, People's Republic of China.
| | - Minghui Pang
- Department of Gastrointestinal Surgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu 610072, People's Republic of China.
| |
Collapse
|
21
|
Zhu S, Soutto M, Chen Z, Peng D, Romero-Gallo J, Krishna US, Belkhiri A, Washington MK, Peek R, El-Rifai W. Helicobacter pylori-induced cell death is counteracted by NF-κB-mediated transcription of DARPP-32. Gut 2017; 66:761-762. [PMID: 27590997 PMCID: PMC5334457 DOI: 10.1136/gutjnl-2016-312141] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 07/11/2016] [Accepted: 08/02/2016] [Indexed: 12/21/2022]
Abstract
OBJECTIVE DARPP-32 is a frequently amplified and overexpressed gene that promotes several oncogenic functions in gastric cancer. Herein, we investigated the relationship between Helicobacter pylori infection, proinflammatory NF-κB activation and regulation of DARPP-32. DESIGN The study used in vivo and in vitro experiments. Luciferase reporter, quantitative real-time PCR, immunoblot, chromatin immunoprecipitation (ChIP), cell viability, H. pylori infection, tissue microarrays and immunohistochemical assays were used. RESULTS Our results indicated that H. pylori infection increased the DARPP-32 mRNA and protein levels in gastric cancer cell lines and gastric mucosa of mice. H. pylori infection increased the activity of NF-κB reporter and p-NF-κB (S536) protein level in vitro and in vivo. To investigate the transcriptional regulation of DARPP-32, we cloned a 3019 bp of the DARPP-32 promoter into the luciferase reporter (pGL3-Luc). Both H. pylori infection and tumour necrosis factor-α treatment induced DARPP-32 reporter activity (p<0.01). Using deletion constructs of DARPP-32 promoter and ChIP assay, we demonstrated that the sequence -996 to -1008 bp containing putative NF-κB-binding sites is the most active region. The induction of DARPP-32 expression by H. pylori infection counteracted H. pylori-induced cell death through activation of serine/threonine-specific protein kinase (AKT), as determined by ATP-Glo and clonogenic survival assays. Immunohistochemistry analysis demonstrated a significant positive correlation between NF-κB and DARPP-32 expression levels in gastric cancer tissues (r2=0.43, p<0.01). CONCLUSIONS Given the high frequency of DARPP-32 overexpression and its prosurvival oncogenic functions, the induction of DARPP-32 expression following H. pylori infection and activation of NF-κB provides a link between infection, inflammation and gastric tumourigenesis.
Collapse
Affiliation(s)
- Shoumin Zhu
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Mohammed Soutto
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Zheng Chen
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - DunFa Peng
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Judith Romero-Gallo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Uma S Krishna
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Abbes Belkhiri
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - M Kay Washington
- Department of Pathology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Richard Peek
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Wael El-Rifai
- Department of Surgery, Vanderbilt University Medical Center, Nashville, Tennessee, USA,Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA,Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| |
Collapse
|
22
|
Chen J, Wang Z, Hu X, Chen R, Romero-Gallo J, Peek RM, Chen LF. BET Inhibition Attenuates Helicobacter pylori-Induced Inflammatory Response by Suppressing Inflammatory Gene Transcription and Enhancer Activation. THE JOURNAL OF IMMUNOLOGY 2016; 196:4132-42. [PMID: 27084101 DOI: 10.4049/jimmunol.1502261] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 03/16/2016] [Indexed: 12/25/2022]
Abstract
Helicobacter pylori infection causes chronic gastritis and peptic ulceration. H. pylori-initiated chronic gastritis is characterized by enhanced expression of many NF-κB-regulated inflammatory cytokines. Brd4 has emerged as an important NF-κB regulator and regulates the expression of many NF-κB-dependent inflammatory genes. In this study, we demonstrated that Brd4 was not only actively involved in H. pylori-induced inflammatory gene mRNA transcription but also H. pylori-induced inflammatory gene enhancer RNA (eRNA) synthesis. Suppression of H. pylori-induced eRNA synthesis impaired H. pylori-induced mRNA synthesis. Furthermore, H. pylori stimulated NF-κB-dependent recruitment of Brd4 to the promoters and enhancers of inflammatory genes to facilitate the RNA polymerase II-mediated eRNA and mRNA synthesis. Inhibition of Brd4 by JQ1 attenuated H. pylori-induced eRNA and mRNA synthesis for a subset of NF-κB-dependent inflammatory genes. JQ1 also inhibited H. pylori-induced interaction between Brd4 and RelA and the recruitment of Brd4 and RNA polymerase II to the promoters and enhancers of inflammatory genes. Finally, we demonstrated that JQ1 suppressed inflammatory gene expression, inflammation, and cell proliferation in H. pylori-infected mice. These studies highlight the importance of Brd4 in H. pylori-induced inflammatory gene expression and suggest that Brd4 could be a potential therapeutic target for the treatment of H. pylori-triggered inflammatory diseases and cancer.
Collapse
Affiliation(s)
- Jinjing Chen
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801; Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, China
| | - Zhen Wang
- Institute of Medicinal Biotechnology, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, China
| | - Xiangming Hu
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Ruichuan Chen
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen 361101, China
| | - Judith Romero-Gallo
- Division of Gastroenterology, Department of Medicine and Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN 37232; and
| | - Richard M Peek
- Division of Gastroenterology, Department of Medicine and Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN 37232; and
| | - Lin-Feng Chen
- Department of Biochemistry, University of Illinois at Urbana-Champaign, Urbana, IL 61801; Department of Medical Biochemistry, College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| |
Collapse
|
23
|
Libânio D, Dinis-Ribeiro M, Pimentel-Nunes P. Helicobacter pylori and microRNAs: Relation with innate immunity and progression of preneoplastic conditions. World J Clin Oncol 2015; 6:111-132. [PMID: 26468448 PMCID: PMC4600186 DOI: 10.5306/wjco.v6.i5.111] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Revised: 06/22/2015] [Accepted: 08/04/2015] [Indexed: 02/06/2023] Open
Abstract
The accepted paradigm for intestinal-type gastric cancer pathogenesis is a multistep progression from chronic gastritis induced by Helicobacter pylori (H. pylori) to gastric atrophy, intestinal metaplasia, dysplasia and ultimately gastric cancer. The genetic and molecular mechanisms underlying disease progression are still not completely understood as only a fraction of colonized individuals ever develop neoplasia suggesting that bacterial, host and environmental factors are involved. MicroRNAs are noncoding RNAs that may influence H. pylori-related pathology through the regulation of the transcription and expression of various genes, playing an important role in inflammation, cell proliferation, apoptosis and differentiation. Indeed, H. pylori have been shown to modify microRNA expression in the gastric mucosa and microRNAs are involved in the immune host response to the bacteria and in the regulation of the inflammatory response. MicroRNAs have a key role in the regulation of inflammatory pathways and H. pylori may influence inflammation-mediated gastric carcinogenesis possibly through DNA methylation and epigenetic silencing of tumor suppressor microRNAs. Furthermore, microRNAs influenced by H. pylori also have been found to be involved in cell cycle regulation, apoptosis and epithelial-mesenchymal transition. Altogether, microRNAs seem to have an important role in the progression from gastritis to preneoplastic conditions and neoplastic lesions and since each microRNA can control the expression of hundreds to thousands of genes, knowledge of microRNAs target genes and their functions are of paramount importance. In this article we present a comprehensive review about the role of microRNAs in H. pylori gastric carcinogenesis, identifying the microRNAs downregulated and upregulated in the infection and clarifying their biological role in the link between immune host response, inflammation, DNA methylation and gastric carcinogenesis.
Collapse
|
24
|
Magalhães A, Marcos-Pinto R, Nairn AV, Dela Rosa M, Ferreira RM, Junqueira-Neto S, Freitas D, Gomes J, Oliveira P, Santos MR, Marcos NT, Xiaogang W, Figueiredo C, Oliveira C, Dinis-Ribeiro M, Carneiro F, Moremen KW, David L, Reis CA. Helicobacter pylori chronic infection and mucosal inflammation switches the human gastric glycosylation pathways. Biochim Biophys Acta Mol Basis Dis 2015; 1852:1928-39. [PMID: 26144047 DOI: 10.1016/j.bbadis.2015.07.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 05/29/2015] [Accepted: 07/01/2015] [Indexed: 02/06/2023]
Abstract
Helicobacter pylori exploits host glycoconjugates to colonize the gastric niche. Infection can persist for decades promoting chronic inflammation, and in a subset of individuals lesions can silently progress to cancer. This study shows that H. pylori chronic infection and gastric tissue inflammation result in a remodeling of the gastric glycophenotype with increased expression of sialyl-Lewis a/x antigens due to transcriptional up-regulation of the B3GNT5, B3GALT5, and FUT3 genes. We observed that H. pylori infected individuals present a marked gastric local pro-inflammatory signature with significantly higher TNF-α levels and demonstrated that TNF-induced activation of the NF-kappaB pathway results in B3GNT5 transcriptional up-regulation. Furthermore, we show that this gastric glycosylation shift, characterized by increased sialylation patterns, favors SabA-mediated H. pylori attachment to human inflamed gastric mucosa. This study provides novel clinically relevant insights into the regulatory mechanisms underlying H. pylori modulation of host glycosylation machinery, and phenotypic alterations crucial for life-long infection. Moreover, the biosynthetic pathways here identified as responsible for gastric mucosa increased sialylation, in response to H. pylori infection, can be exploited as drug targets for hindering bacteria adhesion and counteract the infection chronicity.
Collapse
Affiliation(s)
- Ana Magalhães
- Institute for Research and Innovation in Health (i3S), University of Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Portugal
| | - Ricardo Marcos-Pinto
- Centro Hospitalar do Porto (CHP), Gastroenterology Department, Portugal; Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Portugal; Medical Faculty, University of Porto, Portugal
| | - Alison V Nairn
- Complex Carbohydrate Research Center and Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, USA
| | - Mitche Dela Rosa
- Complex Carbohydrate Research Center and Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, USA
| | - Rui M Ferreira
- Institute for Research and Innovation in Health (i3S), University of Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Portugal
| | - Susana Junqueira-Neto
- Institute for Research and Innovation in Health (i3S), University of Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Portugal
| | - Daniela Freitas
- Institute for Research and Innovation in Health (i3S), University of Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Portugal
| | - Joana Gomes
- Institute for Research and Innovation in Health (i3S), University of Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Portugal
| | - Patrícia Oliveira
- Institute for Research and Innovation in Health (i3S), University of Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Portugal
| | - Marta R Santos
- Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Portugal
| | - Nuno T Marcos
- Institute for Research and Innovation in Health (i3S), University of Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Portugal; Section of Health Sciences, University of Aveiro, Portugal
| | - Wen Xiaogang
- Department of Pathology, Centro Hospitalar São João, Porto, Portugal; Centro Hospitalar Vila Nova de Gaia/Espinho, Portugal
| | - Céu Figueiredo
- Institute for Research and Innovation in Health (i3S), University of Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Portugal; Medical Faculty, University of Porto, Portugal
| | - Carla Oliveira
- Institute for Research and Innovation in Health (i3S), University of Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Portugal; Medical Faculty, University of Porto, Portugal
| | - Mário Dinis-Ribeiro
- Medical Faculty, University of Porto, Portugal; Gastroenterology Department, IPO Porto, Portugal; CIDES/CINTESIS, University of Porto, Portugal
| | - Fátima Carneiro
- Institute for Research and Innovation in Health (i3S), University of Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Portugal; Medical Faculty, University of Porto, Portugal; Department of Pathology, Centro Hospitalar São João, Porto, Portugal
| | - Kelley W Moremen
- Complex Carbohydrate Research Center and Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, USA
| | - Leonor David
- Institute for Research and Innovation in Health (i3S), University of Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Portugal; Medical Faculty, University of Porto, Portugal
| | - Celso A Reis
- Institute for Research and Innovation in Health (i3S), University of Porto, Portugal; Institute of Molecular Pathology and Immunology of the University of Porto (IPATIMUP), Portugal; Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Portugal; Medical Faculty, University of Porto, Portugal.
| |
Collapse
|
25
|
Subhash VV, Ho B. Inflammation and proliferation - a causal event of host response to Helicobacter pylori infection. MICROBIOLOGY (READING, ENGLAND) 2015; 161:1150-1160. [PMID: 25721850 DOI: 10.1099/mic.0.000066] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Helicobacter pylori is a major aetiological agent in the development of various gastroduodenal diseases. Its persistence in gastric mucosa is determined by the interaction between various host, microbial and environmental factors. The bacterium colonizes the gastric epithelium and induces activation of various chemokine mediators, including NFκB, the master regulator of inflammation. H. pylori infection is also associated with an increase in expression of cell cycle regulators, thereby leading to mucosal cell hyper-proliferation. Thus, H. pylori-associated infections manifest activation of key host response events, which inadvertently could lead to the establishment of chronic infection and neoplastic progression. This article reviews and elaborates the current knowledge in H. pylori-induced activation of various host signalling pathways that could promote cancer development. Special focus is placed on the inflammatory and proliferative responses that could serve as suitable biomarkers of infection, since a sustained cell proliferation in an environment rich in inflammatory cells is characteristic in H. pylori-associated gastric malignancies. Here, the role of ERK and WNT signalling in H. pylori-induced activation of inflammatory and proliferative responses respectively is discussed in detail. An in depth analysis of the underlying signalling pathways and interacting partners causing alterations in these crucial host responses could contribute to the development of successful therapeutic strategies for the prevention, management and treatment of H. pylori infection.
Collapse
Affiliation(s)
- Vinod Vijay Subhash
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, 117545, Singapore
| | - Bow Ho
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, 117545, Singapore
| |
Collapse
|
26
|
Helicobacter pylori-Induced Signaling Pathways Contribute to Intestinal Metaplasia and Gastric Carcinogenesis. BIOMED RESEARCH INTERNATIONAL 2015; 2015:737621. [PMID: 26064948 PMCID: PMC4441984 DOI: 10.1155/2015/737621] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 04/20/2015] [Indexed: 12/31/2022]
Abstract
Helicobacter pylori (H. pylori) induces chronic gastric inflammation, atrophic gastritis, intestinal metaplasia, and cancer. Although the risk of gastric cancer increases exponentially with the extent of atrophic gastritis, the precise mechanisms of gastric carcinogenesis have not been fully elucidated. H. pylori induces genetic and epigenetic changes in gastric epithelial cells through activating intracellular signaling pathways in a cagPAI-dependent manner. H. pylori eventually induces gastric cancer with chromosomal instability (CIN) or microsatellite instability (MSI), which are classified as two major subtypes of gastric cancer. Elucidation of the precise mechanisms of gastric carcinogenesis will also be important for cancer therapy.
Collapse
|
27
|
Tavares R, Pathak SK. Helicobacter pylori protein JHP0290 exhibits proliferative and anti-apoptotic effects in gastric epithelial cells. PLoS One 2015; 10:e0124407. [PMID: 25879227 PMCID: PMC4400171 DOI: 10.1371/journal.pone.0124407] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 03/13/2015] [Indexed: 01/06/2023] Open
Abstract
The influence of Helicobacter pylori infection on gastric epithelial cell proliferation, apoptosis and signaling pathways contributes to the development of infection-associated diseases. Here we report that JHP0290, which is a poorly functionally characterized protein from H. pylori, regulates multiple responses in human gastric epithelial cells. The differential expression and release of JHP0290 homologues was observed among H. pylori strains. JHP0290 existed in monomeric and dimeric forms in H. pylori cell extracts and culture broth. Recombinant purified JHP0290 (rJHP0290) also showed monomeric and dimeric forms, whereas the rJHP0290 C162A mutant exhibited only a monomeric form. The dimeric form of the protein was found to bind more efficiently to gastric epithelial cells than the monomeric form. The exposure of gastric epithelial cells to rJHP0290 induced proliferation in a dose-dependent manner. Faster progression into the cell cycle was observed in rJHP0290-challenged gastric epithelial cells. Furthermore, we detected an anti-apoptotic effect of rJHP0290 in gastric epithelial cells when the cells were treated with rJHP0290 in combination with Camptothecin (CPT), which is an inducer of apoptosis. CPT-induced caspase 3 activation was significantly reduced in the presence of rJHP0290. In addition, the activation of ERK MAPK and the transcription factor NFκB was observed in rJHP0290-challenged gastric epithelial cells lines. Our results suggest that JHP0290 may affect H. pylori-induced gastric diseases via the regulation of gastric epithelial cell proliferation and anti-apoptotic pathways.
Collapse
Affiliation(s)
- Raquel Tavares
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Sushil Kumar Pathak
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
- * E-mail: (SKP)
| |
Collapse
|
28
|
Bontems P, Aksoy E, Burette A, Segers V, Deprez C, Mascart F, Cadranel S. NF-κB activation and severity of gastritis in Helicobacter pylori-infected children and adults. Helicobacter 2014; 19:157-67. [PMID: 24661597 DOI: 10.1111/hel.12118] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND In contrast to adults, Helicobacter pylori gastritis in children is reported as milder and ulcer disease as uncommon, but unequivocal data are lacking. OBJECTIVES To compare the frequency of gastro-duodenal ulcers in children and adults as well as the proportion of Helicobacter pylori infection in these patients and to study the effect of chronological age on NF-κB activation and on severity of gastritis. DESIGN Patients referred in one pediatric and one adult facility for upper GI endoscopy were included. Gastric biopsies were obtained in consecutive Helicobacter pylori-infected patients and age-matched negative controls for immunohistochemistry and electrophoresis mobility shift assay. Three age groups were defined: younger than 8 years, 8-17 years, and adults. RESULTS Peptic ulcer disease was less frequent in children and less frequently associated with Helicobacter pylori infection. When comparing infected subjects to controls, densities of neutrophils and CD20 cells in the lamina propria increased in all age groups, CD3 cells increasing only in patients older than 8 years and CD8 cells only in adults. NF-κB-p65-positive cells were also increased only in infected adults as well as NF-κB-binding activity. A positive correlation was found between age and densities of neutrophils and CD3, but not of CD8 or CD20 cells. CONCLUSION Peptic ulcer disease was less frequent in children and less frequently caused by Helicobacter pylori infection. The different clinical outcome of the infection in children can be the consequence of the lower mucosal immune response.
Collapse
Affiliation(s)
- Patrick Bontems
- Paediatric Gastroenterology-Hepatology, Queen Fabiola Children's University Hospital, Université Libre de Bruxelles, Av JJ Crocq 15, 1020, Brussels, Belgium; Laboratory of Vaccinology and Mucosal Immunity, Université Libre de Bruxelles, Route de Lennik 808, 1070, Brussels, Belgium
| | | | | | | | | | | | | |
Collapse
|
29
|
Wang F, Meng W, Wang B, Qiao L. Helicobacter pylori-induced gastric inflammation and gastric cancer. Cancer Lett 2014; 345:196-202. [PMID: 23981572 DOI: 10.1016/j.canlet.2013.08.016] [Citation(s) in RCA: 559] [Impact Index Per Article: 50.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2013] [Revised: 08/07/2013] [Accepted: 08/13/2013] [Indexed: 02/06/2023]
Abstract
Helicobacter pylori (H. pylori) infect over half of the world's population. The prevalence of H. pylori infection and the predominant genotype of H. pylori virulence factors vary considerably across different geographical regions. H. pylori could uniquely persist for decades in the harsh stomach environment, where it damages the gastric mucosa and changes the pattern of gastric hormone release, thereby affects gastric physiology. By utilizing various virulence factors, H. pylori targets different cellular proteins to modulate the host inflammatory response and initiate multiple "hits" on the gastric mucosa, resulting in chronic gastritis and peptic ulceration. Among the long-term consequences of H. pylori infection is gastric malignancies, particularly gastric cancer (GC) and gastric mucosa-associated lymphoid tissue (MALT) lymphoma. As such, H. pylori has been recognized as a class I carcinogen by the International Agency for Research on Cancer. Despite a close causal link between H. pylori infection and the development of gastric malignancies, the precise mechanisms involved in this process are still obscure. Studies over the past two decades have revealed that H. pylori exert oncogenic effects on gastric mucosa through a complex interaction between bacterial factors, host factors, and environmental factors. Numerous signaling pathways can be activated by H. pylori. In this review, we aim to elaborate on the recent developments in the pathophysiological mechanisms of H. pylori-induced gastric inflammation and gastric cancer.
Collapse
Affiliation(s)
- Fei Wang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi Province, China
| | - Wenbo Meng
- The Second Department of General Surgery, The First Hospital of Lanzhou University, Hepatopancreatobiliary Surgery Institute of Gansu Province, Clinical Medical College Cancer, Center of Lanzhou University, Lanzhou 730000, Gansu Province, China
| | - Bingyuan Wang
- Department of Gastroenterology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China.
| | - Liang Qiao
- Storr Liver Unit at Westmead Millennium Institute, The University of Sydney at Westmead Hospital, Westmead, NSW 2145, Australia.
| |
Collapse
|
30
|
Lamb A, Chen J, Blanke SR, Chen LF. Helicobacter pylori activates NF-κB by inducing Ubc13-mediated ubiquitination of lysine 158 of TAK1. J Cell Biochem 2014; 114:2284-92. [PMID: 23606331 DOI: 10.1002/jcb.24573] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 04/12/2013] [Indexed: 12/30/2022]
Abstract
The Helicobacter pylori virulence factor CagA targets a variety of host proteins to alter different cellular responses, including the induction of pro-inflammatory cytokines. We have previously shown that CagA-facilitated lysine 63-linked ubiquitination of TAK1 is essential for the H. pylori-induced NF-κB activation and the expression of proinflammatory cytokines. However, the molecular mechanism for TAK1 ubiquitination and activation in H. pylori-mediated NF-κB activation remains elusive. Here, we identify lysine 158 of TAK1 as the key residue undergoing lysine 63-linked ubiquitination in response to H. pylori infection. Mutation of lysine 158 to arginine prevents the ubiquitination of TAK1 and impairs H. pylori-induced TAK1 and NF-κB activation. Moreover, we demonstrate that E2 ubiquitin conjugating enzyme Ubc13 is involved in H. pylori-mediated TAK1 ubiquitination. Suppressing the activity of Ubc13 by a dominant-negative mutant or siRNA abolishes CagA-facilitated and H. pylori-induced TAK1 and NF-κB activation. These findings further underscore the importance of lysine 63-linked ubiquitination of TAK1 in H. pylori-induced NF-κB activation and NF-κB-mediated inflammatory response.
Collapse
Affiliation(s)
- Acacia Lamb
- Department of Biochemistry, College of Medicine, Urbana, Illinois 61801, USA
| | | | | | | |
Collapse
|
31
|
Lee KE, Khoi PN, Xia Y, Park JS, Joo YE, Kim KK, Choi SY, Jung YD. Helicobacter pylori and interleukin-8 in gastric cancer. World J Gastroenterol 2013; 19:8192-8202. [PMID: 24363509 PMCID: PMC3857441 DOI: 10.3748/wjg.v19.i45.8192] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Revised: 10/17/2013] [Accepted: 11/03/2013] [Indexed: 02/06/2023] Open
Abstract
Helicobacter pylori (H. pylori) is a major etiological factor in the development of gastric cancer. Large-scale epidemiological studies have confirmed the strong association between H. pylori infection and both cancer development and progression. Interleukin-8 (IL-8) is overexpressed in gastric mucosa exposed to H. pylori. The expression of IL-8 directly correlates with a poor prognosis in gastric cancer. IL-8 is multifunctional. In addition to its potent chemotactic activity, it can induce proliferation and migration of cancer cells. In this review, we focus on recent insights into the mechanisms of IL-8 signaling associated with gastric cancer. The relationship between IL-8 and H. pylori is discussed. We also summarize the current therapeutics against IL-8 in gastric cancer.
Collapse
|
32
|
Lamb A, Chen LF. Role of the Helicobacter pylori-induced inflammatory response in the development of gastric cancer. J Cell Biochem 2013; 114:491-7. [PMID: 22961880 DOI: 10.1002/jcb.24389] [Citation(s) in RCA: 155] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Accepted: 08/30/2012] [Indexed: 12/13/2022]
Abstract
Helicobacter pylori (H. pylori) infection causes chronic gastritis and peptic ulceration and is the strongest risk factor for the development of gastric cancer. The pathogenesis of H. pylori is believed to be associated with infection-initiated chronic gastritis, which is characterized by enhanced expression of many inflammatory genes. H. pylori utilizes various virulence factors, targeting different cellular proteins, to modulate the host inflammatory response. In this review, we explore the many different ways by which H. pylori initiates inflammation, leveling many "hits" on the gastric mucosa which can lead to the development of cancer. We also discuss some recent findings in understanding the pathogen-host interactions and the role of transcription factor NF-κB in H. pylori-induced inflammation.
Collapse
Affiliation(s)
- Acacia Lamb
- Department of Biochemistry, College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | | |
Collapse
|
33
|
Kim JM, Kim KM, Park EH, Seo JH, Song JY, Shin SC, Kang HL, Lee WK, Cho MJ, Rhee KH, Youn HS, Baik SC. Anthocyanins from black soybean inhibit Helicobacter pylori-induced inflammation in human gastric epithelial AGS cells. Microbiol Immunol 2013; 57:366-73. [PMID: 23668609 DOI: 10.1111/1348-0421.12049] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Revised: 03/18/2013] [Accepted: 03/21/2013] [Indexed: 12/11/2022]
Abstract
Infection with Helicobacter pylori leads to gastritis, peptic ulcers and gastric cancer. Moreover, when the gastric mucosa is exposed to H. pylori, gastric mucosal inflammatory cytokine interleukin-8 (Il-8) and reactive oxygen species increase. Anthocyanins have anti-oxidative, antibacterial and anti-inflammatory properties. However, the effect of anthocyanins in H. pylori-infected cells is not yet clear. In this study, therefore, the effect of anthocyanins on H. pylori-infected human gastric epithelial cells was examined. AGS cells were pretreated with anthocyanins for 24 hrs followed by H. pylori 26695 infection for up to 24 hrs. Cell viability and ROS production were examined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and 2',7'-dichlorofluorescein diacetate assay, respectively. Western blot analyses and RT-PCR were performed to assess gene and protein expression, respectively. IL-8 secretion in AGS cells was measured by ELISA. It was found that anthocyanins decrease H. pylori-induced ROS enhancement. Anthocyanins also inhibited phosphorylation of mitogen-activated protein kinases, translocation of nuclear factor-kappa B and Iκβα degradation. Furthermore anthocyanins inhibited H. pylori-induced inducible nitric oxide synthases and cyclooxygenase-2 mRNA expression and inhibited IL-8 production by 45.8%. Based on the above findings, anthocyanins might have an anti-inflammatory effect in H. pylori-infected gastric epithelial cells.
Collapse
Affiliation(s)
- Jung-Min Kim
- Department of Microbiology, Gyeongsang National University School of Medicine, Gyeongsang Institute of Health Science, Jinju 660-751, South Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Baud J, Varon C, Chabas S, Chambonnier L, Darfeuille F, Staedel C. Helicobacter pylori initiates a mesenchymal transition through ZEB1 in gastric epithelial cells. PLoS One 2013; 8:e60315. [PMID: 23565224 PMCID: PMC3614934 DOI: 10.1371/journal.pone.0060315] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Accepted: 02/26/2013] [Indexed: 02/07/2023] Open
Abstract
Chronic Helicobacter pylori infection provokes an inflammation of the gastric mucosa, at high risk for ulcer and cancer development. The most virulent strains harbor the cag pathogenicity island (cagPAI) encoding a type 4 secretion system, which allows delivery of bacterial effectors into gastric epithelial cells, inducing pro-inflammatory responses and phenotypic alterations reminiscent of an epithelial-to-mesenchymal transition (EMT). This study characterizes EMT features in H. pylori-infected gastric epithelial cells, and investigates their relationship with NF-κB activation. Cultured human gastric epithelial cell lines were challenged with a cagPAI+ H. pylori strain or cag isogenic mutants. Morphological changes, epithelial and mesenchymal gene expression and EMT-related microRNAs were studied. H. pylori up-regulates mesenchymal markers, including ZEB1. This transcription factor is prominently involved in the mesenchymal transition of infected cells and its up-regulation depends on cagPAI and NF-κB activation. ZEB1 expression and NF-κB activation were confirmed by immunohistochemistry in gastric mucosa from cagPAI+ H. pylori-infected patients. Gastric epithelial cell lines express high miR-200 levels, which are linked to ZEB1 in a reciprocal negative feedback loop and maintain their epithelial phenotype in non-infected conditions. However, miR-200b/c were increased upon infection, despite ZEB1 up-regulation and mesenchymal morphology. In the miR-200b-200a-429 cluster promoter, we identified a functional NF-κB binding site, recruiting NF-κB upon infection and trans-activating the microRNA cluster transcription. In conclusion, in gastric epithelial cells, cagPAI+ H. pylori activates NF-κB, which transactivates ZEB1, subsequently promoting mesenchymal transition. The unexpected N-FκB-dependent increase of miR-200 levels likely thwarts the irreversible loss of epithelial identity in that critical situation.
Collapse
Affiliation(s)
- Jessica Baud
- University Bordeaux, ARNA Laboratory, Bordeaux, France
- INSERM, U869, ARNA Laboratory, Bordeaux, France
| | - Christine Varon
- University Bordeaux, Laboratoire de Bactériologie, Bordeaux, France
- INSERM, U853, Laboratoire de Bactériologie, Bordeaux, France
| | - Sandrine Chabas
- University Bordeaux, ARNA Laboratory, Bordeaux, France
- INSERM, U869, ARNA Laboratory, Bordeaux, France
| | - Lucie Chambonnier
- University Bordeaux, Laboratoire de Bactériologie, Bordeaux, France
- INSERM, U853, Laboratoire de Bactériologie, Bordeaux, France
| | - Fabien Darfeuille
- University Bordeaux, ARNA Laboratory, Bordeaux, France
- INSERM, U869, ARNA Laboratory, Bordeaux, France
- * E-mail: (CS); (FD)
| | - Cathy Staedel
- University Bordeaux, ARNA Laboratory, Bordeaux, France
- INSERM, U869, ARNA Laboratory, Bordeaux, France
- * E-mail: (CS); (FD)
| |
Collapse
|
35
|
Abstract
Helicobacter pylori infection leads to long-lasting chronic inflammation and represents the most common risk factor underlying gastric cancer. Recently, new insights into the mechanisms through which H. pylori and mucosal inflammation lead to cancer development have emerged. H. pylori virulence factors, in particular specific CagA genotypes, represent main factors in gastric cancer, inducing altered intracellular signaling in epithelial cells. The chronic nature of H. pylori infection appears to relate to the VacA virulence factor and Th17/Treg mechanisms. A role of H. pylori infection in epigenetic and microRNA deregulation has been shown. Mutation of the epithelial cell genome, a hallmark of cancer, was demonstrated to accumulate in H. pylori infected stomach partly due to inadequate DNA repair. Gastric stem cells were shown to be targets of oxidative injury in the Helicobacter-inflammatory milieu. Recent advances emphasizing the contribution of bacterial factors, inflammatory mediators, and the host epithelial response in gastric carcinogenesis are reviewed.
Collapse
|
36
|
Yoon J, Cho SJ, Ko YS, Park J, Shin DH, Hwang IC, Han SY, Nam SY, Kim MA, Chang MS, Lee HS, Kim WH, Lee BL. A synergistic interaction between transcription factors nuclear factor-κB and signal transducers and activators of transcription 3 promotes gastric cancer cell migration and invasion. BMC Gastroenterol 2013; 13:29. [PMID: 23402362 PMCID: PMC3583822 DOI: 10.1186/1471-230x-13-29] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Accepted: 01/30/2013] [Indexed: 11/13/2022] Open
Abstract
Background The transcription factor nuclear factor-κB (NF-κB) has been implicated in gastric cancer metastasis, but the underlying molecular mechanisms remain unclear. We investigated the role of the interaction between NF-κB and signal transducers and activators of transcription 3 (STAT3) in controlling metastatic potential of gastric cancer cells. Methods Immunohistochemistry for NF-κB p65 (RelA), phospho-Tyr705-STAT3 (pSTAT3), or matrix metalloproteinase 9 (MMP9) was performed on tissue array slides containing 255 gastric carcinoma specimens. NF-κB inhibition in SNU-638 and MKN1 gastric cancer cell lines were performed by transduction with a retroviral vector containing NF-κB repressor mutant of IκBα, and STAT3 was silenced by RNA interference. We also did luciferase reporter assay, double immunofluorescence staining and immunoblotting. Cell migration and invasion were determined by wound-healing assay and invasion assay, respectively. Results NF-κB and STAT3 were constitutively activated and were positively correlated (P = 0.038) in gastric cancer tissue specimens. In cell culture experiments, NF-κB inhibition reduced STAT3 expression and activation, whereas STAT3 silencing did not affect NF-κB activation. Moreover, both NF-κB inhibition and STAT3 silencing decreased gastric cancer cell migration and invasion in a synergistic manner. In addition, both NF-κB activation and STAT3 activation were positively correlated with MMP9 in gastric cancer tissues (P = 0.001 and P = 0.022, respectively), decreased E-cadherin expression and increased Snail and MMP9 expressions in cultured cells. Conclusion NF-κB and STAT3 are positively associated and synergistically contribute to the metastatic potential of gastric cancer cells. Thus, dual use of NF-κB and STAT3 inhibitors may enhance the efficacy of the anti-metastatic treatment of gastric cancer.
Collapse
Affiliation(s)
- Jiyeon Yoon
- Department of Anatomy, Seoul National University College of Medicine, Seoul, 110-799, South Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Papadakos KS, Sougleri IS, Mentis AF, Hatziloukas E, Sgouras DN. Presence of terminal EPIYA phosphorylation motifs in Helicobacter pylori CagA contributes to IL-8 secretion, irrespective of the number of repeats. PLoS One 2013; 8:e56291. [PMID: 23409168 PMCID: PMC3567036 DOI: 10.1371/journal.pone.0056291] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2012] [Accepted: 01/07/2013] [Indexed: 12/23/2022] Open
Abstract
CagA protein contributes to pro-inflammatory responses during H. pylori infection, following its intracellular delivery to gastric epithelial cells. Here, we report for the first time in an isogenic background, on the subtle role of CagA phosphorylation on terminal EPIYA-C motifs in the transcriptional activation and expression of IL-8. We utilized isogenic H. pylori mutants of P12 reference strain, expressing CagA with varying number of EPIYA-C motifs and the corresponding phosphorylation defective EPIFA-C motifs while preserving intact the CM multimerization motifs. These mutants had been previously closely scrutinized in terms of type IV secretion system functionality, CagA translocation and its subsequent phosphorylation. Following infection of gastric epithelial cell lines, transcriptional activation of IL-8 gene and secreted IL-8 levels were found to be strictly dependent upon the functionality of the EPIYA-C phosphorylation motifs, as EPIFA-C phosphorylation-deficient CagA expression failed to induce full IL-8 transcriptional activity. Interestingly, levels of IL-8 gene activation and of secreted IL-8 were the same, irrespective of the number of EPIYA-C terminal repeats. We monitored IkBα phosphorylation and confirmed CagA involvement in NF-kB activation. Furthermore, we observed that presence of EPIYA-C functional phosphorylation motifs contributed to NF-kB activation. NF-kB upstream signaling events, such as early ERK1/2 and AKT activation were confirmed to be independent of EPIYA-C phosphorylation. On the contrary, use of TAK1 specific inhibitor 5Z-7-Oxozeaenol resulted in complete arrest of IL-8 secretion, in a dose-dependent manner, irrespective of CagA status. H. pylori-infected TAK1-/- mouse embryonic fibroblasts (MEFs) failed to induce NF-kB activity, unlike the respective control MEFs. CagA and TAK1 were found to immunoprecipitate together, irrespective of CagA EPIYA-C status, thus confirming earlier reports of TAK1 and CagA protein interaction. Our data suggest that CagA may potentially interfere with TAK1 activity during NF-kB activation for IL-8 induction in early H. pylori infection.
Collapse
Affiliation(s)
| | - Ioanna S. Sougleri
- Laboratory of Medical Microbiology, Hellenic Pasteur Institute, Athens, Greece
| | - Andreas F. Mentis
- Laboratory of Medical Microbiology, Hellenic Pasteur Institute, Athens, Greece
| | - Efstathios Hatziloukas
- Laboratory of Molecular Biology, Department of Biological Applications and Technology, University of Ioannina, Ioannina, Greece
| | - Dionyssios N. Sgouras
- Laboratory of Medical Microbiology, Hellenic Pasteur Institute, Athens, Greece
- * E-mail:
| |
Collapse
|
38
|
Tsang YHN, Lamb A, Chen LF. New insights into the inactivation of gastric tumor suppressor RUNX3: the role of H. pylori infection. J Cell Biochem 2011; 112:381-6. [PMID: 21268057 DOI: 10.1002/jcb.22964] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Runt-related transcription factor 3, or RUNX3, is a tumor suppressor in gastric cancer. Inactivation of RUNX3 is causally associated with the genesis of gastric cancer, since RUNX3 is frequently inactivated in gastric cancers by hemizygous deletion, hypermethylation of its promoter, or protein mislocalization. Infection with Helicobacter pylori is the strongest risk factor for the development of gastric cancer. Recent studies have indicated that H. pylori infection plays an important role in the inactivation of RUNX3, and that this inactivation contributes to the pathogenesis of H. pylori. Here we summarize these recent advances and discuss their significances in understanding the initiation and development of gastric cancer.
Collapse
Affiliation(s)
- Ying-Hung Nicole Tsang
- Department of Biochemistry, College of Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, USA
| | | | | |
Collapse
|
39
|
Jones KR, Whitmire JM, Merrell DS. A Tale of Two Toxins: Helicobacter Pylori CagA and VacA Modulate Host Pathways that Impact Disease. Front Microbiol 2010; 1:115. [PMID: 21687723 PMCID: PMC3109773 DOI: 10.3389/fmicb.2010.00115] [Citation(s) in RCA: 116] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2010] [Accepted: 09/27/2010] [Indexed: 12/16/2022] Open
Abstract
Helicobacter pylori is a pathogenic bacterium that colonizes more than 50% of the world's population, which leads to a tremendous medical burden. H. pylori infection is associated with such varied diseases as gastritis, peptic ulcers, and two forms of gastric cancer: gastric adenocarcinoma and mucosa-associated lymphoid tissue lymphoma. This association represents a novel paradigm for cancer development; H. pylori is currently the only bacterium to be recognized as a carcinogen. Therefore, a significant amount of research has been conducted to identify the bacterial factors and the deregulated host cell pathways that are responsible for the progression to more severe disease states. Two of the virulence factors that have been implicated in this process are cytotoxin-associated gene A (CagA) and vacuolating cytotoxin A (VacA), which are cytotoxins that are injected and secreted by H. pylori, respectively. Both of these virulence factors are polymorphic and affect a multitude of host cellular pathways. These combined facts could easily contribute to differences in disease severity across the population as various CagA and VacA alleles differentially target some pathways. Herein we highlight the diverse types of cellular pathways and processes targeted by these important toxins.
Collapse
Affiliation(s)
- Kathleen R Jones
- Department of Microbiology and Immunology, Uniformed Services University of the Health Sciences Bethesda, MD, USA
| | | | | |
Collapse
|