Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Gut Microbes ; 14(1): 2105102, 2022.
Article in English | MEDLINE | ID: mdl-35905376

ABSTRACT

Chronic mucosal pathogens have evolved multiple strategies to manipulate the host immune response; consequently, microbes contribute to the development of >2 million cases of cancer/year. Gastric adenocarcinoma is the fourth leading cause of cancer-related death and Helicobacter pylori confers the highest risk for this disease. Gastric innate immune effectors can either eliminate bacteria or mobilize adaptive immune responses including Toll-like receptors (TLRs), and cytosolic DNA sensor/adaptor proteins (e.g., stimulator of interferon genes, STING). The H. pylori strain-specific cag type IV secretion system (T4SS) augments gastric cancer risk and translocates DNA into epithelial cells where it activates the microbial DNA sensor TLR9 and suppresses injury in vivo; however, the ability of H. pylori to suppress additional nucleic acid PRRs within the context of chronic gastric inflammation and injury remains undefined. In this study, in vitro and ex vivo experiments identified a novel mechanism through which H. pylori actively suppresses STING and RIG-I signaling via downregulation of IRF3 activation. In vivo, the use of genetically deficient mice revealed that Th17 inflammatory responses are heightened following H. pylori infection within the context of Sting deficiency in conjunction with increased expression of a known host immune regulator, Trim30a. This novel mechanism of immune suppression by H. pylori is likely a critical component of a finely tuned rheostat that not only regulates the initial innate immune response, but also drives chronic gastric inflammation and injury.


Subject(s)
Gastrointestinal Microbiome , Helicobacter Infections , Helicobacter pylori , Nucleic Acids , Stomach Neoplasms , Animals , Gastric Mucosa/metabolism , Helicobacter Infections/microbiology , Helicobacter pylori/genetics , Immunity, Innate , Inflammation/metabolism , Mice , Nucleic Acids/metabolism , Stomach Neoplasms/microbiology
2.
Cell Commun Signal ; 17(1): 162, 2019 12 10.
Article in English | MEDLINE | ID: mdl-31823782

ABSTRACT

BACKGROUND: Interleukin-22 (IL-22) belongs to the IL-10 cytokine family and is mainly produced by activated Th1 cells. Although IL-22 expression is reported to be elevated in many cancers, and increased IL-22 expression correlates with tumor progression and poor prognosis, little is known about the role of IL-22 in papillary thyroid cancer (PTC). We previously demonstrated that IL-22 promotes PTC cell migration and invasion through the microRNA-595/Sox17 axis. METHODS: We used qRT-PCR and western blot to determine TRIM30, Sox17 and ß-catenin expression in PTC cells. Knockdown and overexpression were performed to detect the role of TRIM30/Sox17/ß-catenin axis on the migration and invasion PTC cells. Co-IP were used to determine the interaction between TRIM30 and Sox17. FINDINGS: In this study, we demonstrated that IL-22 triggered tripartite-motif protein 30 (TRIM30) association with Sox17, thereby mediating K48-linked polyubiquitination of Sox17. We then demonstrated that TRIM30 was a positive regulator of IL-22-regulated migration and invasion of PTC cells. We also found that IL-22 induced the transcriptional activity of ß-catenin and translocation of ß-catenin from cytosol to the nucleus. Upon investigating the mechanisms behind this event, we found that IL-22 disrupted Sox17/ß-catenin interactions by inducing TRIM30/Sox17 interactions, leading to promotion of ß-catenin-dependent signaling. The analysis of hundreds of clinical specimens revealed that IL-22, TRIM30 and ß-catenin levels were upregulated in PTC tissues compared with normal thyroid, and that their expression levels were closely correlated. Taken together, under the influence of IL-22, by sequestration of Sox17, TRIM30 promotes ß-catenin-dependent signaling that promotes PTC cell proliferation.


Subject(s)
Interleukins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , SOXF Transcription Factors/metabolism , Thyroid Cancer, Papillary/metabolism , Thyroid Neoplasms/metabolism , Ubiquitin/metabolism , Animals , Cell Line , Cell Movement , Cell Proliferation , Humans , Intracellular Signaling Peptides and Proteins/deficiency , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Signal Transduction , Thyroid Cancer, Papillary/pathology , Thyroid Neoplasms/pathology , Ubiquitination , Interleukin-22
SELECTION OF CITATIONS
SEARCH DETAIL