1
|
Qian J, Liu KJ, Zhong CH, Xian LN, Hu ZH. Sivelestat sodium alleviated sepsis-induced acute lung injury by inhibiting TGF-β/Smad signaling pathways through upregulating microRNA-744-5p. J Thorac Dis 2024; 16:6616-6633. [PMID: 39552870 PMCID: PMC11565364 DOI: 10.21037/jtd-24-65] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 08/30/2024] [Indexed: 11/19/2024]
Abstract
Background Acute lung injury (ALI) is one of the most common critical illnesses in clinical practice, with sepsis being the most common cause of ALI. Sivelestat sodium (SV) hydrate is a highly effective inhibitor of neutrophil elastase, specifically targeting ALI related to systemic inflammatory response syndrome. The aim of this study is to examine the mechanisms by which SV can reduce the severity of ALI resulting from sepsis. Methods Cecum ligation and puncture (CLP) was employed for creating an animal model of ALI caused by sepsis. Primary human pulmonary microvascular endothelial cells (HPMECs) were treated with lipopolysaccharide (LPS) to develop an in vitro model of infection-induced ALI. Lung tissue damage was assessed by employing hematoxylin-eosin (H&E) and Masson staining. Lung edema was determined by calculating the lung wet-to-dry weight ratio. Lung tissue and cell samples were analyzed using Enzyme-linked immunosorbent assay (ELISA) to detect levels of tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, and IL-6. The 5-ethynyl-2'-deoxyuridine (EdU) and wound-healing assay were used to determine the cell proliferation and migration, while flow cytometry was used for detecting cell apoptosis. The association between microRNA (miR)-744 and transforming growth factor (TGF)-β1 was discovered and confirmed through the utilization of bioinformatics analyses and dual-luciferase gene reporter assay. The analysis of TGF-β1, p-Smad3, and Smad3 was carried out through western blotting and immunohistochemistry in both in vitro and in vivo scenarios. Results In both in vivo and in vitro settings of ALI models of sepsis, there was a significant decrease in the level of miR-744-5p, a significant elevation in the expression of inflammatory factors, and a significant intensification of lung tissue damage. Administration of SV resulted in a significant increase in the level of miR-744-5p, suppressed the inflammatory response, and ultimately improved lung injury. Cell proliferation was significantly enhanced by SV and cell apoptosis was inhibited. The protection of SV was significantly reversed by inhibiting the effect of miR-744-5p. The double-luciferase reporter gene assay revealed substantial interactions occurring between miR-744-5p and TGF-β1. The TGF-β/Smad signaling pathway was significantly inhibited by SV, however, the inhibitory effect can be counteracted by utilizing the miR-744-5p inhibitor. Conclusions The upregulation of miR-744-5p by SV inhibits the TGF-β/Smad signaling pathway, thereby reducing sepsis-induced ALI.
Collapse
Affiliation(s)
- Jin Qian
- Key Laboratory of Emergency and Trauma of Ministry of Education, Department of Intensive Care Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Ke-Jun Liu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Basic Medicine and Life Sciences, Hainan Medical University, Haikou, China
| | - Chang-Hui Zhong
- Key Laboratory of Emergency and Trauma of Ministry of Education, Department of Intensive Care Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Li-Na Xian
- Key Laboratory of Emergency and Trauma of Ministry of Education, Department of Intensive Care Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| | - Zhi-Hua Hu
- Key Laboratory of Emergency and Trauma of Ministry of Education, Department of Intensive Care Medicine, The First Affiliated Hospital of Hainan Medical University, Haikou, China
| |
Collapse
|
2
|
Yang Z, Wei J, Wang Y, Du Y, Song S, Li J, Su Z, Shi Y, Wu H. Irisin Ameliorates Renal Tubulointerstitial Fibrosis by Regulating the Smad4/β-Catenin Pathway in Diabetic Mice. Diabetes Metab Syndr Obes 2023; 16:1577-1593. [PMID: 37292142 PMCID: PMC10244207 DOI: 10.2147/dmso.s407734] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 05/06/2023] [Indexed: 06/10/2023] Open
Abstract
Background The primary pathophysiology of diabetic kidney disease (DKD) is tubulointerstitial fibrosis (TIF), and an essential contributing element is excessive extracellular matrix deposition. Irisin is a polypeptide formed by splitting fibronectin type III domain containing 5 (FNDC5), which participates in a number of physiological and pathological processes. Methods The purpose of this article is to examine irisin's function in DKD and analyze both its in vitro and in vivo effects. The Gene Expression Omnibus (GEO) database was used to download GSE30122, GSE104954, and GSE99325. Analysis of renal tubule samples from nondiabetic and diabetic mice identified 94 differentially expressed genes (DEGs). The transforming growth factor beta receptor 2 (TGFBR2), irisin, and TGF-β1 were utilized as DEGs to examine the impact of irisin on TIF in diabetic kidney tissue, according to the datasets retrieved from the GEO database and Nephroseq database. Additionally, the therapeutic impact of irisin was also examined using Western blot, RT-qPCR, immunofluorescence, immunohistochemistry, and kits for detecting mouse biochemical indices. Results In vitro, the findings demonstrated that irisin not only down-regulated the expression of Smad4 and β-catenin but also reduced the expression of proteins linked to fibrosis, the epithelial-mesenchymal transition (EMT), and mitochondrial dysfunction in HK-2 cells maintained in high glucose (HG) environment. In vivo, overexpressed FNDC5 plasmid was injected into diabetic mice to enhance its expression. Our studies found that overexpressed FNDC5 plasmid not only reversed the biochemical parameters and renal morphological characteristics of diabetic mice but also alleviated EMT and TIF by inhibiting Smad4/β-catenin signaling pathway. Conclusion The above experimental results revealed that irisin could reduce TIF in diabetic mice via regulating the Smad4/β-catenin pathway.
Collapse
Affiliation(s)
- Zhaohua Yang
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, People’s Republic of China
| | - Jinying Wei
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, People’s Republic of China
| | - Yashu Wang
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, People’s Republic of China
| | - Yunxia Du
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, People’s Republic of China
| | - Shan Song
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, People’s Republic of China
| | - Jiawei Li
- Basic Medical College, Hebei Medical University, Shijiazhuang, 050017, People’s Republic of China
| | - Ziyuan Su
- Basic Medical College, Hebei Medical University, Shijiazhuang, 050017, People’s Republic of China
| | - Yonghong Shi
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, People’s Republic of China
| | - Haijiang Wu
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, People’s Republic of China
| |
Collapse
|
3
|
Gaignage M, Zhang X, Stockis J, Dedobbeleer O, Michiels C, Cochez P, Dumoutier L, Coulie PG, Lucas S. Blocking GARP-mediated activation of TGF-β1 did not alter innate or adaptive immune responses to bacterial infection or protein immunization in mice. Cancer Immunol Immunother 2022; 71:1851-1862. [PMID: 34973084 PMCID: PMC9294018 DOI: 10.1007/s00262-021-03119-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 11/22/2021] [Indexed: 12/22/2022]
Abstract
Abstract Transmembrane protein GARP binds latent TGF-β1 to form GARP:(latent)TGF-β1 complexes on the surface of several cell types including Tregs, B-cells, and platelets. Upon stimulation, these cells release active TGF-β1. Blocking TGF-β1 activation by Tregs with anti-GARP:TGF-β1 mAbs overcomes resistance to PD1/PD-L1 blockade and induces immune-mediated regressions of murine tumors, indicating that Treg-derived TGF-β1 inhibits anti-tumor immunity. TGF-β1 exerts a vast array of effects on immune responses. For example, it favors differentiation of TH17 cells and B-cell switch to IgA production, two important processes for mucosal immunity. Here, we sought to determine whether treatment with anti-GARP:TGF-β1 mAbs would perturb immune responses to intestinal bacterial infection. We observed no aggravation of intestinal disease, no systemic dissemination, and no alteration of innate or adaptative immune responses upon oral gavage of C. rodentium in highly susceptible Il22r−/− mice treated with anti-GARP:TGF-β1 mAbs. To examine the effects of GARP:TGF-β1 blockade on Ig production, we compared B cell- and TH cell- responses to OVA or CTB protein immunization in mice carrying deletions of Garp in Tregs, B cells, or platelets. No alteration of adaptive immune responses to protein immunization was observed in the absence of GARP on any of these cells. Altogether, we show that antibody-mediated blockade of GARP:TGF-β1 or genetic deletion of Garp in Tregs, B cells or platelets, do not alter innate or adaptive immune responses to intestinal bacterial infection or protein immunization in mice. Anti-GARP:TGF-β1 mAbs, currently tested for cancer immunotherapy, may thus restore anti-tumor immunity without severely impairing other immune defenses. Précis Immunotherapy with GARP:TGF-β1 mAbs may restore anti-tumor immunity without impairing immune or inflammatory responses required to maintain homeostasis or host defense against infection, notably at mucosal barriers. Supplementary Information The online version contains supplementary material available at 10.1007/s00262-021-03119-8.
Collapse
Affiliation(s)
- Mélanie Gaignage
- de Duve Institute, Université Catholique de Louvain, avenue Hippocrate 74, B1.74.04, 1200, Brussels, Belgium
| | - Xuhao Zhang
- de Duve Institute, Université Catholique de Louvain, avenue Hippocrate 74, B1.74.04, 1200, Brussels, Belgium
| | - Julie Stockis
- de Duve Institute, Université Catholique de Louvain, avenue Hippocrate 74, B1.74.04, 1200, Brussels, Belgium
| | - Olivier Dedobbeleer
- de Duve Institute, Université Catholique de Louvain, avenue Hippocrate 74, B1.74.04, 1200, Brussels, Belgium
| | - Camille Michiels
- de Duve Institute, Université Catholique de Louvain, avenue Hippocrate 74, B1.74.04, 1200, Brussels, Belgium
| | - Perrine Cochez
- de Duve Institute, Université Catholique de Louvain, avenue Hippocrate 74, B1.74.04, 1200, Brussels, Belgium
| | - Laure Dumoutier
- de Duve Institute, Université Catholique de Louvain, avenue Hippocrate 74, B1.74.04, 1200, Brussels, Belgium
| | - Pierre G Coulie
- de Duve Institute, Université Catholique de Louvain, avenue Hippocrate 74, B1.74.04, 1200, Brussels, Belgium
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wavre, Belgium
| | - Sophie Lucas
- de Duve Institute, Université Catholique de Louvain, avenue Hippocrate 74, B1.74.04, 1200, Brussels, Belgium.
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wavre, Belgium.
| |
Collapse
|
4
|
Wang J, Liu Z, Xu Y, Wang Y, Wang F, Zhang Q, Ni C, Zhen Y, Xu R, Liu Q, Fang W, Huang P, Liu X. Enterobacterial LPS-inducible LINC00152 is regulated by histone lactylation and promotes cancer cells invasion and migration. Front Cell Infect Microbiol 2022; 12:913815. [PMID: 35959377 PMCID: PMC9359126 DOI: 10.3389/fcimb.2022.913815] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 06/30/2022] [Indexed: 11/26/2022] Open
Abstract
Gut microbes participate in pathogenesis by interacting with the host genome through epigenetic mechanisms, such as long non-coding RNAs. However, the mechanisms by which the microbiota induce expression alteration of long non-coding RNAs remains unclear. Here, we quantified the transcriptome alteration of human colon cell lines after being infected by a common enteric pathogen Salmonella typhimurium SL1344. We observed a widespread lncRNAs expression alteration. Among them, the elevated expression of LINC00152 was verified and proved to be induced by enteric bacteria-derived lipopolysaccharide (LPS). The inducible LINC00152 were found to inhibit Salmonella invasion and inflammation response. LINC00152 was overexpressed in tumors of the clinical CRC samples compared with adjacent normal tissues. Accordingly, we also demonstrated that overexpression of LINC00152 promoted the migration and invasion of colorectal cancer cells. Consistently, we observed an increased abundance of gram-negative bacteria and LPS in tumors tissue. Taken together, the above data implicated that enriched gram-negative bacteria in tumor tissue might promote tumor growth through modulating the expression of LINC00152. Furthermore, we demonstrated that LPS upregulated the expression of LINC00152 by introducing histone lactylation on its promoter and decreasing the binding efficiency of the repressor, YY1, to it. Our results provide new insights into how enterobacteria affect host epigenetics in human disease.
Collapse
Affiliation(s)
- Jianwei Wang
- Department of Pathogen Biology-Microbiology Division, Globe of health center, Nanjing Medical University, Nanjing, China
- Key Laboratory of Pathogen of Jiangsu Province and Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, China
- Institute of Pediatric Research, Children’s Hospital of Soochow University, Suzhou, China
| | - Zhi Liu
- Department of Pathogen Biology-Microbiology Division, Globe of health center, Nanjing Medical University, Nanjing, China
- Key Laboratory of Pathogen of Jiangsu Province and Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, China
| | - Yuyu Xu
- Department of Pathogen Biology-Microbiology Division, Globe of health center, Nanjing Medical University, Nanjing, China
- Key Laboratory of Pathogen of Jiangsu Province and Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, China
| | - Yipeng Wang
- Department of Pathogen Biology-Microbiology Division, Globe of health center, Nanjing Medical University, Nanjing, China
- Key Laboratory of Pathogen of Jiangsu Province and Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, China
| | - Fei Wang
- Department of Pathogen Biology-Microbiology Division, Globe of health center, Nanjing Medical University, Nanjing, China
| | - Qingqing Zhang
- Department of Pathogen Biology-Microbiology Division, Globe of health center, Nanjing Medical University, Nanjing, China
- Key Laboratory of Pathogen of Jiangsu Province and Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, China
| | - Chunhua Ni
- Department of Surgery , the Third Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Yi Zhen
- Cancer Biotherapy Center, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Rui Xu
- Department of Pathogen Biology-Microbiology Division, Globe of health center, Nanjing Medical University, Nanjing, China
- Key Laboratory of Pathogen of Jiangsu Province and Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, China
| | - Qisha Liu
- Department of Pathogen Biology-Microbiology Division, Globe of health center, Nanjing Medical University, Nanjing, China
- Key Laboratory of Pathogen of Jiangsu Province and Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, China
| | - Weijia Fang
- Cancer Biotherapy Center, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ping Huang
- Department of Surgery , the Third Affiliated Hospital, Nanjing Medical University, Nanjing, China
- *Correspondence: Xingyin Liu, ; Ping Huang,
| | - Xingyin Liu
- Department of Pathogen Biology-Microbiology Division, Globe of health center, Nanjing Medical University, Nanjing, China
- Key Laboratory of Pathogen of Jiangsu Province and Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, China
- Key Laboratory of Holistic Integrative Enterology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Xingyin Liu, ; Ping Huang,
| |
Collapse
|
5
|
Shealy NG, Yoo W, Byndloss MX. Colonization resistance: metabolic warfare as a strategy against pathogenic Enterobacteriaceae. Curr Opin Microbiol 2021; 64:82-90. [PMID: 34688039 DOI: 10.1016/j.mib.2021.09.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 09/22/2021] [Accepted: 09/28/2021] [Indexed: 12/19/2022]
Abstract
The intestine is home to a large and complex bacterial ecosystem (microbiota), which performs multiple beneficial functions for the host, including immune education, nutrition, and protection against invasion by enteric pathogens (colonization resistance). The host and microbiome symbiotic interactions occur in part through metabolic crosstalk. Thus, microbiota members have evolved highly diverse metabolic pathways to inhibit pathogen colonization via activation of protective immune responses and nutrient acquisition and utilization. Conversely, pathogenic Enterobacteriaceae actively induce an inflammation-dependent disruption of the gut microbial ecosystem (dysbiosis) to gain a competitive metabolic advantage against the resident microbiota. This review discusses the recent findings on the crucial role of microbiota metabolites in colonization resistance regulation. Additionally, we summarize metabolic mechanisms used by pathogenic Enterobacteriaceae to outcompete commensal microbes and cause disease.
Collapse
Affiliation(s)
- Nicolas G Shealy
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Woongjae Yoo
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Mariana X Byndloss
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
6
|
Zhang K, Wang N, Lu L, Ma X. Fermentation and Metabolism of Dietary Protein by Intestinal Microorganisms. Curr Protein Pept Sci 2021; 21:807-811. [PMID: 32048966 DOI: 10.2174/1389203721666200212095902] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 07/25/2019] [Accepted: 07/26/2019] [Indexed: 12/18/2022]
Abstract
Dietary protein is linked to the intestinal microorganisms. The decomposition of dietary protein can provide nutrients for microbial growth, which in turn can ferment protein to produce some metabolites. This review elaborates that the effects of different protein levels and types on intestinal microorganisms and their metabolites fermented by intestinal microorganisms, as well as the effects of these metabolites on organisms. It is well known that intestinal microbial imbalance can cause some diseases. Dietary protein supplementation can alter the composition of intestinal microorganisms and thus regulates the body health. However, protein can also produce some harmful metabolites. Therefore, how to rationally supplement protein is particularly important.
Collapse
Affiliation(s)
- Ke Zhang
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University,
Beijing 100193, China
| | - Nan Wang
- China Institute of Veterinary Drug Control, Beijing 100081,China
| | - Lin Lu
- Animal Science and Technology College, Beijing University of Agriculture, Beijing 102206, China
| | - Xi Ma
- State Key Laboratory of Animal Nutrition, College of Animal Science and Technology, China Agricultural University,
Beijing 100193, China
| |
Collapse
|
7
|
Guo P, Zhang K, Ma X, He P. Clostridium species as probiotics: potentials and challenges. J Anim Sci Biotechnol 2020; 11:24. [PMID: 32099648 PMCID: PMC7031906 DOI: 10.1186/s40104-019-0402-1] [Citation(s) in RCA: 287] [Impact Index Per Article: 57.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 11/11/2019] [Indexed: 02/06/2023] Open
Abstract
Clostridium species, as a predominant cluster of commensal bacteria in our gut, exert lots of salutary effects on our intestinal homeostasis. Up to now, Clostridium species have been reported to attenuate inflammation and allergic diseases effectively owing to their distinctive biological activities. Their cellular components and metabolites, like butyrate, secondary bile acids and indolepropionic acid, play a probiotic role primarily through energizing intestinal epithelial cells, strengthening intestinal barrier and interacting with immune system. In turn, our diets and physical state of body can shape unique pattern of Clostridium species in gut. In view of their salutary performances, Clostridium species have a huge potential as probiotics. However, there are still some nonnegligible risks and challenges in approaching application of them. Given this, this review summarized the researches involved in benefits and potential risks of Clostridium species to our health, in order to develop Clostridium species as novel probiotics for human health and animal production.
Collapse
Affiliation(s)
- Pingting Guo
- State Key Laboratory of Animal Nutrition, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193 China
| | - Ke Zhang
- State Key Laboratory of Animal Nutrition, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193 China
| | - Xi Ma
- State Key Laboratory of Animal Nutrition, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193 China
| | - Pingli He
- State Key Laboratory of Animal Nutrition, China Agricultural University, No. 2 Yuanmingyuan West Road, Beijing, 100193 China
| |
Collapse
|
8
|
Zhang S, Al-Maghout T, Cao H, Pelzl L, Salker MS, Veldhoen M, Cheng A, Lang F, Singh Y. Gut Bacterial Metabolite Urolithin A (UA) Mitigates Ca 2+ Entry in T Cells by Regulating miR-10a-5p. Front Immunol 2019; 10:1737. [PMID: 31417547 PMCID: PMC6685097 DOI: 10.3389/fimmu.2019.01737] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 07/09/2019] [Indexed: 12/16/2022] Open
Abstract
The gut microbiota influences several biological functions including immune responses. Inflammatory bowel disease is favorably influenced by consumption of several dietary natural plant products such as pomegranate, walnuts, and berries containing polyphenolic compounds such as ellagitannins and ellagic acid. The gut microbiota metabolizes ellagic acid resulting in the formation of bioactive urolithins A, B, C, and D. Urolithin A (UA) is the most active and effective gut metabolite and acts as a potent anti-inflammatory and anti-oxidant agent. However, whether gut metabolite UA affects the function of immune cells remains incompletely understood. T cell proliferation is stimulated by store operated Ca2+ entry (SOCE) resulting from stimulation of Orai1 by STIM1/STIM2. We show here that treatment of murine CD4+ T cells with UA (10 μM, 3 days) significantly blunted SOCE in CD4+ T cells, an effect paralleled by significant downregulation of Orai1 and STIM1/2 transcript levels and protein abundance. UA treatment further increased miR-10a-5p abundance in CD4+ T cells in a dose dependent fashion. Overexpression of miR-10a-5p significantly decreased STIM1/2 and Orai1 mRNA and protein levels as well as SOCE in CD4+ T cells. UA further decreased CD4+ T cell proliferation. Thus, the gut bacterial metabolite UA increases miR-10a-5p levels thereby downregulating Orai1/STIM1/STIM2 expression, store operated Ca2+ entry, and proliferation of murine CD4+ T cells.
Collapse
Affiliation(s)
- Shaqiu Zhang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Tamer Al-Maghout
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Hang Cao
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Lisann Pelzl
- Department of Physiology, University of Tübingen, Tübingen, Germany
- Centre for Clinical Transfusion Medicine, Tübingen University, Tübingen, Germany
| | - Madhuri S. Salker
- Research Institute of Women's Health, University of Tübingen, Tübingen, Germany
| | - Marc Veldhoen
- Instituto de Medicina Molecular, Joâo Lobo Antunes, Faculdade de Medicina da Universidade de Lisboa, Lisbon, Portugal
| | - Anchun Cheng
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Florian Lang
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | - Yogesh Singh
- Department of Physiology, University of Tübingen, Tübingen, Germany
- Institute of Medical Genetics and Applied Genomics, Tübingen University, Tübingen, Germany
| |
Collapse
|