1
|
Bae SDW, Nguyen R, Yuen L, Lam V, George J, Qiao L. Constitutive androstane receptor (CAR) functions as a tumor suppressor via regulating stemness in liver cancer. Sci Rep 2024; 14:30926. [PMID: 39730609 DOI: 10.1038/s41598-024-81571-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 11/27/2024] [Indexed: 12/29/2024] Open
Abstract
Constitutive androstane receptor (CAR) is a xenosensor that is almost exclusively expressed in the liver. Studies in rodents suggest an oncogenic role for CAR in liver cancer, but its role in human liver cancer is unclear. We aimed to investigate the functional roles of CAR in human liver cancer with a focus on the liver cancer stem cells. We used bioinformatics to increase our understanding of CAR in human liver cancer and associated stem cell markers. We studied the functional roles of CAR in human liver cancer with a focus on the liver cancer stem cell using siRNA, modulation of CAR activity, and tumorsphere formation assays. We have revealed significant associations between CAR and a wide variety of signalling pathways including stemness signalling. Further in vitro studies have shown that activation of CAR significantly reduces cancer cell stemness and represses proliferation, migration, invasion, and the tumorsphere-forming abilities of liver cancer cells (p < 0.05). Our data demonstrates the unequivocal tumor-suppressive role of CAR in liver cancer. While more detailed mechanistic studies are warranted, the efficacy of CAR xeno-activators in the treatment of advanced hepatocellular carcinoma (HCC) may potentially open a new avenue for liver cancer therapy.
Collapse
Affiliation(s)
- Sarah Da Won Bae
- Storr Liver Centre, Westmead Institute for Medical Research, Department of Medicine, the University of Sydney at Westmead Hospital, Westmead, NSW, 2145, Australia
| | - Romario Nguyen
- Storr Liver Centre, Westmead Institute for Medical Research, Department of Medicine, the University of Sydney at Westmead Hospital, Westmead, NSW, 2145, Australia
| | - Lawrence Yuen
- Department of Surgery, Westmead Hospital, Westmead, NSW, 2145, Australia
| | - Vincent Lam
- Department of Surgery, Westmead Hospital, Westmead, NSW, 2145, Australia
| | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, Department of Medicine, the University of Sydney at Westmead Hospital, Westmead, NSW, 2145, Australia.
- School of Medicine, University of Sydney, Sydney, Australia.
| | - Liang Qiao
- Storr Liver Centre, Westmead Institute for Medical Research, Department of Medicine, the University of Sydney at Westmead Hospital, Westmead, NSW, 2145, Australia.
| |
Collapse
|
2
|
Li Z, Sun M, Yang R, Wang Z, Zhu Q, Zhang Y, Yang H, Meng Z, Hu L, Sui L. Mediator complex subunit 1 promotes oral squamous cell carcinoma progression by activating MMP9 transcription and suppressing CD8 + T cell antitumor immunity. J Exp Clin Cancer Res 2024; 43:270. [PMID: 39343952 PMCID: PMC11440895 DOI: 10.1186/s13046-024-03191-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 09/13/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND The role of Mediator complex subunit 1 (MED1), a pivotal transcriptional coactivator implicated in diverse biological pathways, remains unexplored in the context of oral squamous cell carcinoma (OSCC). This study aims to elucidate the contributory mechanisms and potential impact of MED1 on the progression of OSCC. METHODS The expression and clinical significance of MED1 in OSCC tissues were evaluated through the bioinformatics analyses. The effects of MED1 on the biological behavior of OSCC cancer cells were assessed both in vitro and in vivo. Dual-luciferase reporter assay, chromatin immunoprecipitation (ChIP) assay, bioinformatic analysis, CD8+ T cell isolation experiment, coculture experiment, enzyme-linked immunosorbent assay (ELISA), and flow cytometric analysis were employed to elucidate the underlying mechanism through which MED1 operates in the progression of OSCC. RESULTS MED1 exhibited upregulation in both OSCC tissues and multiple OSCC cell lines, which correlated with decreased overall survival in patients. In vitro experiments demonstrated that knockdown of MED1 in metastatic OSCC cell lines SCC-9 and UPCI-SCC-154 hindered cell migration and invasion, while overexpression of MED1 promoted these processes. Whereas, MED1 knockdown had no impact on proliferation of cell lines mentioned above. In vivo studies further revealed that downregulation of MED1 effectively suppressed distant metastasis in OSCC. Mechanistically, MED1 enhanced the binding of transcription factors c-Jun and c-Fos to the matrix metalloprotein 9 (MMP9) promoters, resulting in a significant upregulation of MMP9 transcription. This process contributes to the migration and invasion of SCC-9 and UPCI-SCC-154 cells. Furthermore, MED1 modulated the expression of programmed death-ligand 1 (PD-L1) through the Notch signaling pathway, consequently impacting the tumor-killing capacity of CD8+ T cells in the tumor microenvironment. CONCLUSIONS Our findings indicate that MED1 plays a pivotal role in OSCC progression through the activation of MMP9 transcription and suppression of CD8+ T cell antitumor immunity, suggesting that MED1 may serve as a novel prognostic marker and therapeutic target in OSCC.
Collapse
Affiliation(s)
- Zhe Li
- Department of Prosthodontics, Tianjin Medical University School and Hospital of Stomatology, Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin Medical University Institute of Stomatology, 12 Qixiangtai Road, Tianjin, 300070, China
| | - Mengke Sun
- Immunology Department, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, 22 Qixiangtai Road, Tianjin, 300070, China
| | - Ruimeng Yang
- Department of Prosthodontics, Tianjin Medical University School and Hospital of Stomatology, Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin Medical University Institute of Stomatology, 12 Qixiangtai Road, Tianjin, 300070, China
| | - Zheng Wang
- Department of Prosthodontics, Tianjin Medical University School and Hospital of Stomatology, Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin Medical University Institute of Stomatology, 12 Qixiangtai Road, Tianjin, 300070, China
| | - Qianyu Zhu
- Immunology Department, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, 22 Qixiangtai Road, Tianjin, 300070, China
| | - Yue Zhang
- Department of Prosthodontics, Tianjin Medical University School and Hospital of Stomatology, Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin Medical University Institute of Stomatology, 12 Qixiangtai Road, Tianjin, 300070, China
| | - Haosun Yang
- Department of Prosthodontics, Tianjin Medical University School and Hospital of Stomatology, Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin Medical University Institute of Stomatology, 12 Qixiangtai Road, Tianjin, 300070, China
| | - Zhaosong Meng
- Department of Oral and Maxillofacial Surgery, Tianjin Medical University School and Hospital of Stomatology, Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin Medical University Institue of Stomatology, 12 Qixiangtai Road, Tianjin, 300070, China.
| | - Lizhi Hu
- Immunology Department, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), Tianjin Medical University, 22 Qixiangtai Road, Tianjin, 300070, China.
| | - Lei Sui
- Department of Prosthodontics, Tianjin Medical University School and Hospital of Stomatology, Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin Medical University Institute of Stomatology, 12 Qixiangtai Road, Tianjin, 300070, China.
| |
Collapse
|
3
|
Kim H. Regulation of Med1 protein by overexpression of BAP1 in breast cancer cells. Mol Cell Oncol 2024; 11:2347827. [PMID: 38708315 PMCID: PMC11067983 DOI: 10.1080/23723556.2024.2347827] [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: 01/18/2024] [Accepted: 04/23/2024] [Indexed: 05/07/2024]
Abstract
Med1 binds to a nuclear receptor and regulates transcription. Elevated Med1 protein expression promotes cancer growth in hormone-dependent breast and prostate cancers. Med1 protein expression was investigated by deubiquitinating enzymes (DUBs) overexpression in breast cancer cell lines. Various DNA constructs of SRT-DUBs were overexpressed in the MCF7 cell line, and Med1 protein expression was investigated by western blotting. The cell growth and in vitro invasion assay were performed in BRCA1-associated protein 1 (BAP1) wild type and mutant (C91A) overexpressed cells. Ubiquitination of the Med1 protein was observed, and Med1 protein expression and transcriptional activity were verified by various DUBs overexpressed. Although Med1 protein expression increased upon the overexpression of BAP1, it was not affected by the overexpression of BAP1 mutant (C91A). BAP1 was increased by the E2 treatment, which has an important effect on the breast cancer growth, and cell growth was decreased by BAP1 C91A overexpression. However, metastatic capacities were decreased by BAP1. In addition, the binding between the Med1 and the BAP1 protein was observed. These data suggested that BAP1 regulated Med1 protein expression in breast cancer cells and involved in cancer cell growth and metastasis by binding to Med1 protein.
Collapse
Affiliation(s)
- Hyunju Kim
- Department of Health Sciences, The Graduate School of Dong-A University, Busan, South Korea
| |
Collapse
|
4
|
Lei ZY, Li ZH, Lin DN, Cao J, Chen JF, Meng SB, Wang JL, Liu J, Zhang J, Lin BL. Med1 inhibits ferroptosis and alleviates liver injury in acute liver failure via Nrf2 activation. Cell Biosci 2024; 14:54. [PMID: 38678227 PMCID: PMC11056072 DOI: 10.1186/s13578-024-01234-4] [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: 12/10/2023] [Accepted: 04/12/2024] [Indexed: 04/29/2024] Open
Abstract
BACKGROUND Extensive hepatocyte mortality and the absence of specific medical therapy significantly contribute to the unfavorable prognosis of acute liver failure (ALF). Ferroptosis is a crucial form of cell death involved in ALF. In this study, we aimed to determine the impact of Mediator complex subunit 1 (Med1) on ferroptosis and its potential hepatoprotective effects in ALF. RESULTS Med1 expression is diminished in the liver of lipopolysaccharide (LPS)/D-galactosamine (D-GalN)-induced ALF mice, as well as in hepatocytes damaged by H2O2 or TNF-α/D-GalN in vitro. Med1 overexpression mitigates liver injury and decreases the mortality rate of ALF mice by ferroptosis inhibition. The mechanism by which Med1 inhibits erastin-induced ferroptosis in hepatocytes involves the upregulation of nuclear factor erythroid 2-related factor 2 (Nrf2) and its downstream antioxidant genes heme oxygenase-1 (HO-1), glutamate cysteine ligase catalytic (GCLC), and NAD(P)H quinone oxidoreductase 1 (NQO1). Furthermore, Med1 overexpression suppresses the transcription of proinflammatory cytokines tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6) in the liver of mice with LPS/D-GalN-induced ALF. CONCLUSION Overall, our research findings indicate that Med1 suppresses ferroptosis and alleviates liver injury in LPS/D-GalN-induced ALF through the activation of Nrf2. These findings substantiate the therapeutic viability of targeting the Med1-Nrf2 axis as a means of treating individuals afflicted with ALF.
Collapse
Affiliation(s)
- Zi-Ying Lei
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, Guangdong, China
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Zhi-Hui Li
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, Guangdong, China
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Deng-Na Lin
- Department of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, Guangdong, China
| | - Jing Cao
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, Guangdong, China
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Jun-Feng Chen
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, Guangdong, China
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Shi-Bo Meng
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, Guangdong, China
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Jia-Lei Wang
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, Guangdong, China
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Jing Liu
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, Guangdong, China.
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China.
| | - Jing Zhang
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, Guangdong, China.
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China.
| | - Bing-Liang Lin
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, Guangdong, China.
- Guangdong Key Laboratory of Liver Disease Research, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China.
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
5
|
Kolonay DW, Sattler KM, Strawser C, Rafael-Fortney J, Mihaylova MM, Miller KE, Lepper C, Baskin KK. Temporal regulation of the Mediator complex during muscle proliferation, differentiation, regeneration, aging, and disease. Front Cell Dev Biol 2024; 12:1331563. [PMID: 38690566 PMCID: PMC11058648 DOI: 10.3389/fcell.2024.1331563] [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: 11/01/2023] [Accepted: 03/26/2024] [Indexed: 05/02/2024] Open
Abstract
Genesis of skeletal muscle relies on the differentiation and fusion of mono-nucleated muscle progenitor cells into the multi-nucleated muscle fiber syncytium. The temporally-controlled cellular and morphogenetic changes underlying this process are initiated by a series of highly coordinated transcription programs. At the core, the myogenic differentiation cascade is driven by muscle-specific transcription factors, i.e., the Myogenic Regulatory Factors (MRFs). Despite extensive knowledge on the function of individual MRFs, very little is known about how they are coordinated. Ultimately, highly specific coordination of these transcription programs is critical for their masterfully timed transitions, which in turn facilitates the intricate generation of skeletal muscle fibers from a naïve pool of progenitor cells. The Mediator complex links basal transcriptional machinery and transcription factors to regulate transcription and could be the integral component that coordinates transcription factor function during muscle differentiation, growth, and maturation. In this study, we systematically deciphered the changes in Mediator complex subunit expression in skeletal muscle development, regeneration, aging, and disease. We incorporated our in vitro and in vivo experimental results with analysis of publicly available RNA-seq and single nuclei RNA-seq datasets and uncovered the regulation of Mediator subunits in different physiological and temporal contexts. Our experimental results revealed that Mediator subunit expression during myogenesis is highly dynamic. We also discovered unique temporal patterns of Mediator expression in muscle stem cells after injury and during the early regeneration period, suggesting that Mediator subunits may have unique contributions to directing muscle stem cell fate. Although we observed few changes in Mediator subunit expression in aging muscles compared to younger muscles, we uncovered extensive heterogeneity of Mediator subunit expression in dystrophic muscle nuclei, characteristic of chronic muscle degeneration and regeneration cycles. Taken together, our study provides a glimpse of the complex regulation of Mediator subunit expression in the skeletal muscle cell lineage and serves as a springboard for mechanistic studies into the function of individual Mediator subunits in skeletal muscle.
Collapse
Affiliation(s)
- Dominic W. Kolonay
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Kristina M. Sattler
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Corinne Strawser
- Department of Pediatrics, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Jill Rafael-Fortney
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Maria M. Mihaylova
- Department of Biological Chemistry and Pharmacology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- The Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Katherine E. Miller
- Department of Pediatrics, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Institute for Genomic Medicine, Nationwide Children’s Hospital, Columbus, OH, United States
| | - Christoph Lepper
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Kedryn K. Baskin
- Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
6
|
Chaudhuri SM, Weinberg SE, Wang D, Yalom LK, Montauti E, Iyer R, Tang AY, Torres Acosta MA, Shen J, Mani NL, Wang S, Liu K, Lu W, Bui TM, Manzanares LD, Dehghani Z, Wai CM, Gao B, Wei J, Yue F, Cui W, Singer BD, Sumagin R, Zhang Y, Fang D. Mediator complex subunit 1 architects a tumorigenic Treg cell program independent of inflammation. Cell Rep Med 2024; 5:101441. [PMID: 38428427 PMCID: PMC10983042 DOI: 10.1016/j.xcrm.2024.101441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 12/18/2023] [Accepted: 02/01/2024] [Indexed: 03/03/2024]
Abstract
While immunotherapy has revolutionized cancer treatment, its safety has been hampered by immunotherapy-related adverse events. Unexpectedly, we show that Mediator complex subunit 1 (MED1) is required for T regulatory (Treg) cell function specifically in the tumor microenvironment. Treg cell-specific MED1 deletion does not predispose mice to autoimmunity or excessive inflammation. In contrast, MED1 is required for Treg cell promotion of tumor growth because MED1 is required for the terminal differentiation of effector Treg cells in the tumor. Suppression of these terminally differentiated Treg cells is sufficient for eliciting antitumor immunity. Both human and murine Treg cells experience divergent paths of differentiation in tumors and matched tissues with non-malignant inflammation. Collectively, we identify a pathway promoting the differentiation of a Treg cell effector subset specific to tumors and demonstrate that suppression of a subset of Treg cells is sufficient for promoting antitumor immunity in the absence of autoimmune consequences.
Collapse
Affiliation(s)
- Shuvam M Chaudhuri
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Samuel E Weinberg
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Dongmei Wang
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Lenore K Yalom
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Elena Montauti
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Radhika Iyer
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Amy Y Tang
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Manuel A Torres Acosta
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Medical Scientist Training Program, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Jian Shen
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Nikita L Mani
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Shengnan Wang
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Kun Liu
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Weiyuan Lu
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Triet M Bui
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Laura D Manzanares
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Zeinab Dehghani
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Ching Man Wai
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Beixue Gao
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Juncheng Wei
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Feng Yue
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Weiguo Cui
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Benjamin D Singer
- Division of Pulmonary and Critical Care Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Ronen Sumagin
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Yana Zhang
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Deyu Fang
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA; Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
7
|
Gao J, Bao M, Xing Y, Ding Y, Han T, Wen E, Liu J, Yue S, Wang R, Wang L, Liu J, Zhao S, Huang J, Liu E, Bai L. Mediator subunit MED1 deficiency prevents carbon tetrachloride-induced hepatic fibrosis in mice. Am J Physiol Gastrointest Liver Physiol 2023; 325:G418-G428. [PMID: 37668531 DOI: 10.1152/ajpgi.00076.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 08/17/2023] [Accepted: 08/22/2023] [Indexed: 09/06/2023]
Abstract
Mediator subunit mediator 1 (MED1) mediates ligand-dependent binding of the mediator coactivator complex to various nuclear receptors and plays a critical role in embryonic development, lipid and glucose metabolism, liver regeneration, and tumorigenesis. However, the precise role of MED1 in the development of liver fibrosis has been unclear. Here, we showed that MED1 expression was increased in livers from nonalcoholic steatohepatitis (NASH) patients and mice and positively correlated with transforming growth factor β (TGF-β) signaling and profibrotic factors. Upon treatment with carbon tetrachloride (CCl4), hepatic fibrosis was much less in liver-specific MED1 deletion (MED1ΔLiv) mice than in MED1fl/fl littermates. TGF-β/Smad2/3 signaling pathway was inhibited, and gene expression of fibrotic markers, including α-smooth muscle actin (α-SMA), collagen type 1 α 1 (Col1a1), matrix metalloproteinase-2 (Mmp2), and metallopeptidase inhibitor 1 (Timp1) were decreased in livers of MED1ΔLiv mice with CCl4 injection. Transcriptomic analysis revealed that the differentially expressed genes in livers of CCl4-administered MED1ΔLiv mice were enriched in the pathway of oxidoreductase activity, followed by robustly reduced oxidoreductase activity-related genes, such as Gm4756, Txnrd3, and Etfbkmt. More importantly, we found that the reduction of reactive oxygen species (ROS) in MED1 knockdown hepatocytes blocked the activation of TGF-β/Smad2/3 pathway and the expression of fibrotic genes in LX2 cells. These results indicate that MED1 is a positive regulator for hepatic fibrogenesis, and MED1 may be considered as a potential therapeutic target for the regression of liver fibrosis.NEW & NOTEWORTHY In this study, we present the first evidence that liver mediator 1 (MED1) deficiency attenuated carbon tetrachloride-induced hepatic fibrosis in mouse. The underlying mechanism is that MED1 deficiency reduces reactive oxygen species (ROS) production in hepatocytes, thus restricts the activation of TGF-β/Smad2/3 signaling pathway and fibrogenic genes expression in hepatic stellate cells (HSCs). These data suggest that MED1 is an essential regulator for hepatic fibrogenesis, and MED1 may be considered as a potential therapeutic target for liver fibrosis.
Collapse
Affiliation(s)
- Jie Gao
- Department of Laboratory Animal Science, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, People's Republic of China
- Cardiometabolic Innovation Center, Ministry of Education, Xi'an, People's Republic of China
- School of Biological Science Technology and Engineering, Shaanxi University of Technology, Hanzhong, People's Republic of China
| | - Miaoye Bao
- Department of Laboratory Animal Science, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, People's Republic of China
- Cardiometabolic Innovation Center, Ministry of Education, Xi'an, People's Republic of China
| | - Yuanming Xing
- Cardiometabolic Innovation Center, Ministry of Education, Xi'an, People's Republic of China
| | - Yiming Ding
- Cardiometabolic Innovation Center, Ministry of Education, Xi'an, People's Republic of China
| | - Tuo Han
- Cardiometabolic Innovation Center, Ministry of Education, Xi'an, People's Republic of China
| | - Ergang Wen
- Department of Laboratory Animal Science, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, People's Republic of China
- Cardiometabolic Innovation Center, Ministry of Education, Xi'an, People's Republic of China
| | - Jun Liu
- Department of Laboratory Animal Science, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, People's Republic of China
- Cardiometabolic Innovation Center, Ministry of Education, Xi'an, People's Republic of China
| | - Shaoyun Yue
- Department of Laboratory Animal Science, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Rong Wang
- Department of Laboratory Animal Science, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, People's Republic of China
- Cardiometabolic Innovation Center, Ministry of Education, Xi'an, People's Republic of China
| | - Ling Wang
- School of Biological Science Technology and Engineering, Shaanxi University of Technology, Hanzhong, People's Republic of China
| | - Junhui Liu
- Department of Clinical Laboratory, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Sihai Zhao
- Department of Laboratory Animal Science, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, People's Republic of China
- Cardiometabolic Innovation Center, Ministry of Education, Xi'an, People's Republic of China
| | - Jiansheng Huang
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri, United States
| | - Enqi Liu
- Department of Laboratory Animal Science, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, People's Republic of China
- Cardiometabolic Innovation Center, Ministry of Education, Xi'an, People's Republic of China
| | - Liang Bai
- Department of Laboratory Animal Science, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, People's Republic of China
- Cardiometabolic Innovation Center, Ministry of Education, Xi'an, People's Republic of China
| |
Collapse
|
8
|
Mir R, Elfaki I, Elangeeb ME, Moawadh MS, Tayeb FJ, Barnawi J, Albalawi IA, Alharbi AA, Alhelali MH, Alsaedi BSO. Comprehensive Molecular Evaluation of HNF-1 Alpha, miR-27a, and miR-146 Gene Variants and Their Link with Predisposition and Progression in Type 2 Diabetes Patients. J Pers Med 2023; 13:1270. [PMID: 37623520 PMCID: PMC10455578 DOI: 10.3390/jpm13081270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/07/2023] [Accepted: 08/11/2023] [Indexed: 08/26/2023] Open
Abstract
BACKGROUND Type 2 diabetes (T2D) is a metabolic condition induced by insulin resistance and pancreatic beta cell dysfunction. MicroRNAs (miRNAs) have biological significance because they regulate processes such as the molecular signaling pathways involved in the pathophysiology of diabetes mellitus. The hepatocyte nuclear factor-1 alpha (HNF-1 alpha) is a transcription factor found in hepatocytes and the pancreas. Mutations in the HNF-1 alpha gene were reportedly associated with maturity-onset diabetes of the young (MODY). The objective of the present study was to examine the associations between MiR-27a, MiR-146, and HNF-1 alpha single-nucleotide variations (SNVs) with T2D risk in the Saudi population. METHODOLOGY We evaluated the association of SNVs of miR-27a rs895819 A>G, 146a-rs2910164 C>G, and HNF-1 alpha rs1169288 G>T (I27L) with the risk of T2D in Saudi patients with the Amplification Refractory Mutation System PCR (ARMS-PCR). For the miR-27a SNVs, we used 115 cases (82 males, 33 females) and 117 matched healthy controls (HCs); for the Mir-146 SNVs, we used 103 cases (70 males, 33 females) and 108 matched HCs; and for the HNF-1 alpha, we employed 110 patients (80 males, 30 females) and 110 HCs. The blood biochemistry of the participants was essayed using commercial kits, and the methods of statistical analysis used were the Chi-square test, the Fisher exact test, and a multivariate analysis based on logistic regression, like the odds ratio (OD) and risk ratio (RR), with 95% confidence intervals (CIs). RESULTS The MiR-27a rs895819 AG genotype was linked to increased T2D susceptibility, with OR = 2.01 and p-value = 0.011, and the miR-146 rs2910164 CG genotype and C allele were linked to an elevated risk of T2D, with OR = 2.75, p-value < 0.0016, OR = 1.77, and p-value = 0.004. The results also showed that the GT genotype and T allele of the HNF-1 alpha (rs1169288) G>T is linked to T2D, with OR = 2.18, p-value = 0.0061, and 1.77, p-value = 0.0059. CONCLUSIONS The SNVs in miR-27a, miR-146, and HNF-1 alpha can be potential loci for T2D risk. The limitations of this study include the relatively small sample size and the fact that it was a cross-sectional study. To our knowledge, this is the first study to highlight the association between miR-27a, miR-146, and HNF-1 alpha SNVs and the risk of T2D in the Saudi population. Future large-scale case-control studies, as well as studies on the functions of the proteins and protein interaction studies for HNF-1 alpha, are required to verify our findings. Furthermore, these findings can be used for the identification and stratification of at-risk populations via genetic testing for T2D-prevention strategies.
Collapse
Affiliation(s)
- Rashid Mir
- Department of Medical Lab Technology, Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia; (M.S.M.); (F.J.T.); (J.B.)
| | - Imadeldin Elfaki
- Department of Biochemistry, Faculty of Science, University of Tabuk, Tabuk 47512, Saudi Arabia;
| | - M. E. Elangeeb
- Department of Basic Medical Sciences, Faculty of Applied Medical Sciences, University of Bisha, Bisha 67714, Saudia Arabia;
| | - Mamdoh S. Moawadh
- Department of Medical Lab Technology, Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia; (M.S.M.); (F.J.T.); (J.B.)
| | - Faris Jamal Tayeb
- Department of Medical Lab Technology, Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia; (M.S.M.); (F.J.T.); (J.B.)
| | - Jameel Barnawi
- Department of Medical Lab Technology, Prince Fahad Bin Sultan Chair for Biomedical Research, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk 71491, Saudi Arabia; (M.S.M.); (F.J.T.); (J.B.)
| | | | - Amnah A. Alharbi
- Department of Biochemistry, Faculty of Science, University of Tabuk, Tabuk 47512, Saudi Arabia;
| | - Marwan H. Alhelali
- Department of Statistics, University of Tabuk, Tabuk 47512, Saudi Arabia; (M.H.A.); (B.S.O.A.)
| | - Basim S. O. Alsaedi
- Department of Statistics, University of Tabuk, Tabuk 47512, Saudi Arabia; (M.H.A.); (B.S.O.A.)
| |
Collapse
|
9
|
Ilchuk LA, Kubekina MV, Okulova YD, Silaeva YY, Tatarskiy VV, Filatov MA, Bruter AV. Genetically Engineered Mice Unveil In Vivo Roles of the Mediator Complex. Int J Mol Sci 2023; 24:ijms24119330. [PMID: 37298278 DOI: 10.3390/ijms24119330] [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: 04/28/2023] [Revised: 05/16/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
The Mediator complex is a multi-subunit protein complex which plays a significant role in the regulation of eukaryotic gene transcription. It provides a platform for the interaction of transcriptional factors and RNA polymerase II, thus coupling external and internal stimuli with transcriptional programs. Molecular mechanisms underlying Mediator functioning are intensively studied, although most often using simple models such as tumor cell lines and yeast. Transgenic mouse models are required to study the role of Mediator components in physiological processes, disease, and development. As constitutive knockouts of most of the Mediator protein coding genes are embryonically lethal, conditional knockouts and corresponding activator strains are needed for these studies. Recently, they have become more easily available with the development of modern genetic engineering techniques. Here, we review existing mouse models for studying the Mediator, and data obtained in corresponding experiments.
Collapse
Affiliation(s)
- Leonid A Ilchuk
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Marina V Kubekina
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Yulia D Okulova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Yulia Yu Silaeva
- Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, 119334 Moscow, Russia
| | - Victor V Tatarskiy
- Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, 119334 Moscow, Russia
| | - Maxim A Filatov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Alexandra V Bruter
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
- Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology", Ministry of Health of the Russian Federation, Kashirskoe Sh. 24, 115478 Moscow, Russia
| |
Collapse
|
10
|
Yang Z, Liu Y, Cheng Q, Chen T. Targeting super enhancers for liver disease: a review. PeerJ 2023; 11:e14780. [PMID: 36726725 PMCID: PMC9885865 DOI: 10.7717/peerj.14780] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 01/03/2023] [Indexed: 01/28/2023] Open
Abstract
Background Super enhancers (SEs) refer to the ultralong regions of a gene accompanied by multiple transcription factors and cofactors and strongly drive the expression of cell-type-related genes. Recent studies have demonstrated that SEs play crucial roles in regulating gene expression related to cell cycle progression and transcription. Aberrant activation of SEs is closely related to the occurrence and development of liver disease. Liver disease, especially liver failure and hepatocellular carcinoma (HCC), constitutes a major class of diseases that seriously endanger human health. Currently, therapeutic strategies targeting SEs can dramatically prevent disease progression and improve the prognosis of animal models. The associated new approaches to the treatment of related liver disease are relatively new and need systematic elaboration. Objectives In this review, we elaborate on the features of SEs and discuss their function in liver disease. Additionally, we review their application prospects in clinical practice in the future. The article would be of interest to hepatologists, molecular biologists, clinicians, and all those concerned with targeted therapy and prognosis of liver disease. Methodology We searched three bibliographic databases (Web of Science Core Collection, Embase, PubMed) from 01/1981 to 06/2022 for peer-reviewed scientific publications focused on (1) gene treatment of liver disease; (2) current status of SE research; and (3) targeting SEs for liver disease. We included English language original studies only. Results The number of published studies considering the role of enhancers in liver disease is considerable. Since SEs were just defined in 2013, the corresponding data on SEs are scarce: approximately 50 papers found in bibliographic databases on the correlation between enhancers (or SEs) and liver disease. Remarkably, half of these papers were published in the past three years, indicating the growing interest of the scientific community in this issue. Studies have shown that treatments targeting components of SEs can improve outcomes in liver disease in animal and clinical trials. Conclusions The treatment of liver disease is facing a bottleneck, and new treatments are needed. Therapeutic regimens targeting SEs have an important role in the treatment of liver disease. However, given the off-target effect of gene therapy and the lack of clinical trials, the available experimental data are still fragmented and controversial.
Collapse
|
11
|
Meng Z, Li Z, Guo S, Wu D, Wei R, Liu J, Hu L, Sui L. MED1 Ablation Promotes Oral Mucosal Wound Healing via JNK Signaling Pathway. Int J Mol Sci 2022; 23:13414. [PMID: 36362197 PMCID: PMC9655393 DOI: 10.3390/ijms232113414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 10/31/2022] [Accepted: 10/31/2022] [Indexed: 08/12/2023] Open
Abstract
Mediator complex subunit 1 (MED1) is a coactivator of multiple transcription factors and plays a key role in regulating epidermal homeostasis as well as skin wound healing. It is unknown, however, whether it plays a role in healing oral mucosal wounds. In this study, we investigate MED1's functional effects on oral mucosal wound healing and its underlying mechanism. The epithelial-specific MED1 null (Med1epi-/-) mice were established using the Cre-loxP system with C57/BL6 background. A 3 mm diameter wound was made in the cheek mucosa of the 8-week-old mice. In vivo experiments were conducted using HE staining and immunostaining with Ki67 and uPAR antibodies. The in vitro study used lentiviral transduction, scratch assays, qRT-PCR, and Western blotting to reveal the underlying mechanisms. The results showed that ablation of MED1 accelerated oral mucosal wound healing in 8-week-old mice. As a result of ablation of MED1, Activin A/Follistatin expression was altered, resulting in an activation of the JNK/c-Jun pathway. Similarly, knockdown of MED1 enhanced the proliferation and migration of keratinocytes in vitro, promoting re-epithelialization, which accelerates the healing of oral mucosal wounds. Our study reveals a novel role for MED1 in oral keratinocytes, providing a new molecular therapeutic target for accelerated wound healing.
Collapse
Affiliation(s)
- Zhaosong Meng
- School of Stomatology, Tianjin Medical University, Tianjin 300014, China
| | - Zhe Li
- School of Stomatology, Tianjin Medical University, Tianjin 300014, China
| | - Shuling Guo
- School of Stomatology, Tianjin Medical University, Tianjin 300014, China
| | - Danfeng Wu
- School of Stomatology, Tianjin Medical University, Tianjin 300014, China
| | - Ran Wei
- School of Stomatology, Tianjin Medical University, Tianjin 300014, China
| | - Jiacheng Liu
- School of Stomatology, Tianjin Medical University, Tianjin 300014, China
| | - Lizhi Hu
- Immunology Department, Key Laboratory of Immune Microenvironment and Disease, Ministry of Education, Tianjin Medical University, Tianjin 300014, China
| | - Lei Sui
- School of Stomatology, Tianjin Medical University, Tianjin 300014, China
| |
Collapse
|
12
|
Xiao T, Guo Z, Fu M, Huang J, Wang X, Zhao Y, Tao L, Shen X. Amelioration of Alcoholic Liver Disease by Activating PXR-Cytochrome P450s Axis with Blackberry Extract. SEPARATIONS 2022; 9:321. [DOI: 10.3390/separations9100321] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2025] Open
Abstract
Blackberry is widely used in diets for its rich biological phytochemicals and health benefits. However, the relationship between the effect of blackberry extract (BBE) on ameliorating alcoholic liver disease (ALD) and the PXR-Cytochrome P450s axis in vivo and in vitro is unknown. In this study, 50% and 30% ethanol by gavage were used to establish acute and subacute ALD. Male mice were intragastrically administered BBE with 25, 50, and 100 mg/kg BW in the treatment groups. In the experiment, samples were collected, and related indices and histopathological observation were measured. In addition, the potential mechanism was predicted by network and docking studies, which were verified by qRT-PCR analysis, the detection of apoptosis, the measurement of mitochondrial membrane potential, the detection of ROS levels, and Western blotting in liver tissues and HepG2 cells. The acute and subacute ALD experiments indicated BBE ameliorated liver indices, AST, ALT, SOD, and MDA in serum, and the histopathology changed, as observed via H&E, Sirius red, and oil red O staining. The potential mechanism was predicted by network and docking studies, which were verified by experiments. Western blotting suggested BBE reduced the protein expression of NF-κB, TGF-β, IL-6, and α-SMA, and enhanced PXR and CAR in livers. In addition, qRT-PCR showed BBE significantly elevated the mRNA levels of PXR, CAR, CYP3A25, CYP3A11, and CYP2B10. In the experiment of the ethanol-induced apoptosis of HepG2 cells, BBE reduced the apoptosis of HepG2 cells by boosting mitochondrial membrane potential, reducing the apoptotic rate and ROS content, lessening the expression of Bax, and inducing the expression of PXR. For the first time, this study demonstrated BBE’s preventive effects on ALD, which are associated with the antioxidation and stimulation of the PXR-Cytochrome P450s axis. In addition, BBE is available as a nutritional agent.
Collapse
Affiliation(s)
- Ting Xiao
- The State Key Laboratory of Functions and Applications of Medicinal Plants, The Department of Pharmaceutic Preparation of Chinse Medicine, The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China
| | - Zhenghong Guo
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, China
| | - Min Fu
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China
| | - Jiaoyan Huang
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China
| | - Xiaowei Wang
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China
| | - Yuqing Zhao
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Ling Tao
- The State Key Laboratory of Functions and Applications of Medicinal Plants, The Department of Pharmaceutic Preparation of Chinse Medicine, The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China
| | - Xiangchun Shen
- The State Key Laboratory of Functions and Applications of Medicinal Plants, The Department of Pharmaceutic Preparation of Chinse Medicine, The High Educational Key Laboratory of Guizhou Province for Natural Medicinal Pharmacology and Druggability, School of Pharmaceutical Sciences, Guizhou Medical University, Guiyang 550025, China
- The Key Laboratory of Optimal Utilization of Natural Medicine Resources, School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guiyang 550025, China
| |
Collapse
|
13
|
Jiao A, Liu H, Ding R, Zheng H, Zhang C, Feng Z, Lei L, Wang X, Su Y, Yang X, Sun C, Zhang L, Bai L, Sun L, Zhang B. Med1 Controls Effector CD8+ T Cell Differentiation and Survival through C/EBPβ-Mediated Transcriptional Control of T-bet. THE JOURNAL OF IMMUNOLOGY 2022; 209:855-863. [DOI: 10.4049/jimmunol.2200037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 06/25/2022] [Indexed: 01/04/2023]
Abstract
Abstract
Effector CD8+ T cells are crucial players in adaptive immunity for effective protection against invading pathogens. The regulatory mechanisms underlying CD8+ T cell effector differentiation are incompletely understood. In this study, we defined a critical role of mediator complex subunit 1 (Med1) in controlling effector CD8+ T cell differentiation and survival during acute bacterial infection. Mice with Med1-deficient CD8+ T cells exhibited significantly impaired expansion with evidently reduced killer cell lectin-like receptor G1+ terminally differentiated and Ly6c+ effector cell populations. Moreover, Med1 deficiency led to enhanced cell apoptosis and expression of multiple inhibitory receptors (programmed cell death 1, T cell Ig and mucin domain–containing-3, and T cell immunoreceptor with Ig and ITIM domains). RNA-sequencing analysis revealed that T-bet– and Zeb2-mediated transcriptional programs were impaired in Med1-deficient CD8+ T cells. Overexpression of T-bet could rescue the differentiation and survival of Med1-deficient CD8+ effector T cells. Mechanistically, the transcription factor C/EBPβ promoted T-bet expression through interacting with Med1 in effector T cells. Collectively, our findings revealed a novel role of Med1 in regulating effector CD8+ T cell differentiation and survival in response to bacterial infection.
Collapse
Affiliation(s)
- Anjun Jiao
- *Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, Shaanxi, China
- †Institute of Infection and Immunity, Translational Medicine Institute, Xi’an Jiaotong University, Xi’an, Shaanxi, China
- ‡Key Laboratory of Environment and Genes Related to Diseases, Xi’an Jiaotong University, Ministry of Education, Xi’an, Shaanxi, China
- §Xi’an Key Laboratory of Immune Related Diseases, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Haiyan Liu
- *Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, Shaanxi, China
- †Institute of Infection and Immunity, Translational Medicine Institute, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Renyi Ding
- *Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, Shaanxi, China
- †Institute of Infection and Immunity, Translational Medicine Institute, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Huiqiang Zheng
- *Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, Shaanxi, China
- †Institute of Infection and Immunity, Translational Medicine Institute, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Cangang Zhang
- *Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, Shaanxi, China
- †Institute of Infection and Immunity, Translational Medicine Institute, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Zhao Feng
- *Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, Shaanxi, China
- †Institute of Infection and Immunity, Translational Medicine Institute, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Lei Lei
- *Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, Shaanxi, China
- †Institute of Infection and Immunity, Translational Medicine Institute, Xi’an Jiaotong University, Xi’an, Shaanxi, China
- ‡Key Laboratory of Environment and Genes Related to Diseases, Xi’an Jiaotong University, Ministry of Education, Xi’an, Shaanxi, China
- §Xi’an Key Laboratory of Immune Related Diseases, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Xin Wang
- *Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, Shaanxi, China
- †Institute of Infection and Immunity, Translational Medicine Institute, Xi’an Jiaotong University, Xi’an, Shaanxi, China
- ‡Key Laboratory of Environment and Genes Related to Diseases, Xi’an Jiaotong University, Ministry of Education, Xi’an, Shaanxi, China
- §Xi’an Key Laboratory of Immune Related Diseases, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Yanhong Su
- *Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, Shaanxi, China
- †Institute of Infection and Immunity, Translational Medicine Institute, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Xiaofeng Yang
- *Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, Shaanxi, China
- †Institute of Infection and Immunity, Translational Medicine Institute, Xi’an Jiaotong University, Xi’an, Shaanxi, China
- ‡Key Laboratory of Environment and Genes Related to Diseases, Xi’an Jiaotong University, Ministry of Education, Xi’an, Shaanxi, China
- §Xi’an Key Laboratory of Immune Related Diseases, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Chenming Sun
- *Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, Shaanxi, China
- †Institute of Infection and Immunity, Translational Medicine Institute, Xi’an Jiaotong University, Xi’an, Shaanxi, China
- ‡Key Laboratory of Environment and Genes Related to Diseases, Xi’an Jiaotong University, Ministry of Education, Xi’an, Shaanxi, China
- §Xi’an Key Laboratory of Immune Related Diseases, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Lianjun Zhang
- ¶Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- ‖Suzhou Institute of Systems Medicine, Suzhou, Jiangsu, China; and
| | - Liang Bai
- #Institute of Cardiovascular Science, Xi’an Jiaotong University Health Science Center, Xi’an, Shaanxi, China
| | - Lina Sun
- *Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, Shaanxi, China
- †Institute of Infection and Immunity, Translational Medicine Institute, Xi’an Jiaotong University, Xi’an, Shaanxi, China
- ‡Key Laboratory of Environment and Genes Related to Diseases, Xi’an Jiaotong University, Ministry of Education, Xi’an, Shaanxi, China
- §Xi’an Key Laboratory of Immune Related Diseases, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| | - Baojun Zhang
- *Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi’an Jiaotong University, Xi’an, Shaanxi, China
- †Institute of Infection and Immunity, Translational Medicine Institute, Xi’an Jiaotong University, Xi’an, Shaanxi, China
- ‡Key Laboratory of Environment and Genes Related to Diseases, Xi’an Jiaotong University, Ministry of Education, Xi’an, Shaanxi, China
- §Xi’an Key Laboratory of Immune Related Diseases, Xi’an Jiaotong University, Xi’an, Shaanxi, China
| |
Collapse
|
14
|
Wu S, Li Q, Cao Y, Luo S, Wang Z, Zhang T. Mediator complex subunit 8 is a prognostic biomarker in hepatocellular carcinoma. Am J Transl Res 2022; 14:1765-1777. [PMID: 35422940 PMCID: PMC8991165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Accepted: 02/16/2022] [Indexed: 06/14/2023]
Abstract
BACKGROUND Mediator complex subunit 8 (MED8) is known for its role in encoding a subunit of the mediator complex (MED), that is critical for transcription. MED8 is significantly expressed in various tumors and has been correlated with an unfavorable prognosis. Nevertheless, no relationships have been found between MED8 and the clinical characteristics of hepatocellular carcinoma (HCC). METHODS To conduct an evaluation of correlations between clinicopathologic characteristics and MED8 expression, the logistic regression, Wilcoxon signed-rank test, and Kruskal-Wallis test were used. To perform analysis of factors contributing to prognosis, the Kaplan-Meier approach and the Cox regression analyses were used. A nomogram on the basis of a Cox multivariate analysis was employed to anticipate the influence of MED8 on patient prognosis. The receiver operating characteristic (ROC) curves were plotted and the areas under the curve (AUC) were calculated to assess the prognostic value of MED8. Both immune infiltration analysis and Gene Set Enrichment Analysis (GSEA) were applied to reveal significant enrichment differences among TCGA data. Quantitative RT-PCR (qRT-PCR) and western blotting were used to verify the difference in the expression of MED8 in normal and hepatocellular carcinoma cells. The immunohistochemical method was used to validate the MED8 expression in tumor and adjoining tissues of HCC patients. RESULTS A univariate analysis showed that high MED8 expression predicts poor disease-specific survival (DSS) (HR: 2.57; 95% confidence interval (CI) 1.62, 4.07; P<0.001). Multivariate regression analysis showed that high MED8 (adjusted HR: 3.032 (1.817, 5.060); P<0.001) expression and M stage (adjusted HR=4.075 (1.179-14.091) for M1 vs. M0, P=0.026) served as prognostic indicators of unfavorable overall survival in an independent manner in patients with HCC. The C-index for the nomogram was 0.732 (95% CI: 0.698, 0.766) and the AUC of MED8 was 0.817 (95% CI: 0.778, 0.857). Functional analysis showed that the cell cycle checkpoints, p53 dependent G1-DNA damage response, mitotic G1-G1-S phases, and mitotic G2-G2-M phases, were significantly enriched in DEGs associated with MED8 expression. Th2 cells were positively correlated with MED8 expression. CONCLUSIONS MED8 predicts poor prognosis in HCC, possibly through modulating the cell cycle and Th2 cells.
Collapse
Affiliation(s)
- Shuang Wu
- Clinical Laboratory, The Affiliated Children Hospital of Xi’an Jiaotong UniversityXi’an, Shaanxi, China
| | - Qiao Li
- Clinical Laboratory, The Affiliated Children Hospital of Xi’an Jiaotong UniversityXi’an, Shaanxi, China
| | - Yuan Cao
- Department of Pulmonary and Critical Care Medicine, The Second Affiliated Hospital of Xi’an Jiaotong University (Xibei Hospital)Xi’an, Shaanxi, China
| | - Senyuan Luo
- Department of Pathology, Taihe Hospital, Hubei Medical UniversityShiyan, Hubei, China
| | - Zengguo Wang
- Clinical Laboratory, The Affiliated Children Hospital of Xi’an Jiaotong UniversityXi’an, Shaanxi, China
| | - Taoyuan Zhang
- Department of Anesthesiology, Rizhao International Heart HospitalRizhao, Shandong, China
| |
Collapse
|
15
|
Zhou F, Ding M, Gu Y, Fan G, Liu C, Li Y, Sun R, Wu J, Li J, Xue X, Li H, Li X. Aurantio-Obtusin Attenuates Non-Alcoholic Fatty Liver Disease Through AMPK-Mediated Autophagy and Fatty Acid Oxidation Pathways. Front Pharmacol 2022; 12:826628. [PMID: 35087411 PMCID: PMC8787202 DOI: 10.3389/fphar.2021.826628] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 12/21/2021] [Indexed: 12/18/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD), manifested as the aberrant accumulation of lipids in hepatocytes and inflammation, has become an important cause of advanced liver diseases and hepatic malignancies worldwide. However, no effective therapy has been approved yet. Aurantio-obtusin (AO) is a main bioactive compound isolated from Cassia semen that has been identified with multiple pharmacological activities, including improving adiposity and insulin resistance. However, the ameliorating effects of AO on diet-induced NAFLD and underlying mechanisms remained poorly elucidated. Our results demonstrated that AO significantly alleviated high-fat diet and glucose-fructose water (HFSW)-induced hepatic steatosis in mice and oleic acid and palmitic acid (OAPA)-induced lipid accumulation in hepatocytes. Remarkably, AO was found to distinctly promote autophagy flux and influence the degradation of lipid droplets by inducing AMPK phosphorylation. Additionally, the induction of AMPK triggered TFEB activation and promoted fatty acid oxidation (FAO) by activating PPARα and ACOX1 and decreasing the expression of genes involved in lipid biosynthesis. Meanwhile, the lipid-lowing effect of AO was significantly prevented by the pretreatment with inhibitors of autophagy, PPARα or ACOX1, respectively. Collectively, our study suggests that AO ameliorates hepatic steatosis via AMPK/autophagy- and AMPK/TFEB-mediated suppression of lipid accumulation, which opens new opportunities for pharmacological treatment of NAFLD and associated complications.
Collapse
Affiliation(s)
- Fei Zhou
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Mingning Ding
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Yiqing Gu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Guifang Fan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Chuanyang Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yijie Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Rong Sun
- The Second Hospital of University, Jinan, China.,Advanced Medical Research Institute, Shandong University, Jinan, China
| | - Jianzhi Wu
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Jianchao Li
- The Second Hospital of University, Jinan, China.,Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiaoyong Xue
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Hongjuan Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaojiaoyang Li
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
16
|
van Liempd S, Cabrera D, Pilzner C, Kollmus H, Schughart K, Falcón-Pérez JM. Impaired beta-oxidation increases vulnerability to influenza A infection. J Biol Chem 2021; 297:101298. [PMID: 34637789 PMCID: PMC8564733 DOI: 10.1016/j.jbc.2021.101298] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 12/20/2022] Open
Abstract
Influenza A virus (IAV) infection casts a significant burden on society. It has particularly high morbidity and mortality rates in patients suffering from metabolic disorders. The aim of this study was to relate metabolic changes with IAV susceptibility using well-characterized inbred mouse models. We compared the highly susceptible DBA/2J (D2) mouse strain for which IAV infection is lethal with the C57BL/6J (B6) strain, which exhibits a moderate course of disease and survives IAV infection. Previous studies showed that D2 has higher insulin and glucose levels and is predisposed to develop diet-induced type 2 diabetes. Using high-resolution liquid chromatography–coupled MS, the plasma metabolomes of individual animals were repeatedly measured up to 30 days postinfection. The biggest metabolic difference between these strains in healthy and infected states was in the levels of malonylcarnitine, which was consistently increased 5-fold in D2. Other interstrain and intrastrain differences in healthy and infected animals were observed for acylcarnitines, glucose, branched-chain amino acids, and oxidized fatty acids. By mapping metabolic changes to canonical pathways, we found that mitochondrial beta-oxidation is likely disturbed in D2 animals. In noninfected D2 mice, this leads to increased glycerolipid production and reduced acylcarnitine production, whereas in infected D2 animals, peroxisomal beta-oxidation becomes strongly increased. From these studies, we conclude that metabolic changes caused by a distortion of mitochondrial and peroxisomal metabolism might impact the innate immune response in D2, leading to high viral titers and mortality.
Collapse
Affiliation(s)
| | - Diana Cabrera
- Metabolomics Platform CIC bioGUNE-BRTA, Derio, Spain
| | - Carolin Pilzner
- Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Heike Kollmus
- Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Klaus Schughart
- Department of Infection Genetics, Helmholtz Centre for Infection Research, Braunschweig, Germany; University of Veterinary Medicine Hannover, Hannover, Germany; Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Juan M Falcón-Pérez
- Metabolomics Platform CIC bioGUNE-BRTA, Derio, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
17
|
Liu J, Zhu J, Zhang X, Jia Y, Lee X, Gao Z. Hsa-miR-637 inhibits human hepatocyte proliferation by targeting Med1-interacting proteins. LIVER RESEARCH 2021; 5:88-96. [PMID: 39959344 PMCID: PMC11791805 DOI: 10.1016/j.livres.2021.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/13/2020] [Accepted: 01/18/2021] [Indexed: 11/30/2022]
Abstract
Background Recent studies have shown that mediator complex subunit 1 (Med1) can significantly affect hepatocyte proliferation and differentiation. Acting as a tumor suppressor, microRNA-637 (hsa-miR-637) can inhibit the growth of hepatocarcinoma cells and further induce cell apoptosis. However, the function of hsa-miR-637 and its target genes during liver regeneration remains to be elucidated. Methods This study used co-immunoprecipitation (Co-IP) assay, transfection, luciferase reporter assay, functional assay by cell counting kit-8 (CCK-8), Annexin V-FITC/propidium iodide apoptosis assay, and quantitative polymerase chain reaction analysis of chromatin immunoprecipitation (ChIP) for analysis. Results Hsa-miR-637 has been suggested to suppress the expression of two Med1-interacting nuclear receptors, identified as the peroxisome proliferator-activated receptor alpha (PPARA) and thyroid hormone receptor alpha (THRA) at the transcriptional and translational levels in the human liver HL-7702 cell line. The interaction between Med1 and PPARA/THRA in HL-7702 cells was then confirmed. The transcriptional repression of hsa-miR-637 on PPARA and THRA was also demonstrated. Moreover, hsa-miR-637 has been determined to suppress the proliferation of HL-7702 cells. Furthermore, cell cycle arrest of HL-7702 cells was induced by transfection of hsa-miR-637 at the S phase, but its apoptosis failed. Finally, PPARA was indicated to directly bind to the promoter of some transcription factors, like β-catenin, mouse double minute 2 (MDM2), and p53. Conclusions This study has confirmed that hsa-miR-637 plays an antiproliferative role during liver regeneration, which may contribute in understanding the regenerative process of the liver.
Collapse
Affiliation(s)
- Jing Liu
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jianyun Zhu
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaohong Zhang
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yuzhi Jia
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Xuejun Lee
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhiliang Gao
- Department of Infectious Diseases, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
18
|
Lei L, Yang X, Su Y, Zheng H, Liu J, Liu H, Zou Y, Jiao A, Wang X, Zhang C, Zhang X, Zhang J, Zhang D, Zhou X, Shi L, Liu E, Bai L, Sun C, Zhang B. Med1 controls CD8 T cell maintenance through IL-7R-mediated cell survival signalling. J Cell Mol Med 2021; 25:4870-4876. [PMID: 33733611 PMCID: PMC8107092 DOI: 10.1111/jcmm.16465] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/01/2021] [Accepted: 03/03/2021] [Indexed: 12/15/2022] Open
Abstract
Under steady‐state conditions, the pool size of peripheral CD8+ T cells is maintained through turnover and survival. Beyond TCR and IL‐7R signals, the underlying mechanisms are less well understood. In the present study, we found a significant reduction of CD8+ T cell proportion in spleens but not in thymi of mice with T cell‐specific deletion of Mediator Subunit 1 (Med1). A competitive transfer of wild‐type (WT) and Med1‐deficient CD8+ T cells reproduced the phenotype in the same recipients and confirmed intrinsic role of Med1. Furthermore, we observed a comparable degree of migration and proliferation but a significant increase of cell death in Med1‐deficient CD8+ T cells compared with WT counterparts. Finally, Med1‐deficient CD8+ T cells exhibited a decreased expression of interleukin‐7 receptor α (IL‐7Rα), down‐regulation of phosphorylated‐STAT5 (pSTAT5) and Bim up‐regulation. Collectively, our study reveals a novel role of Med1 in the maintenance of CD8+ T cells through IL‐7Rα/STAT5 pathway‐mediated cell survival.
Collapse
Affiliation(s)
- Lei Lei
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China.,Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, China.,Xi'an Key Laboratory of Immune Related Diseases, Xi'an, China
| | - Xiaofeng Yang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China.,Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, China.,Xi'an Key Laboratory of Immune Related Diseases, Xi'an, China
| | - Yanhong Su
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China.,Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Huiqiang Zheng
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China.,Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Jun Liu
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China.,Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Haiyan Liu
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China.,Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Yujing Zou
- Duke University Medical Center, Durham, NC, USA
| | - Anjun Jiao
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China.,Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Xin Wang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China.,Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Cangang Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China.,Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Xingzhe Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China.,Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Jiahui Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China.,Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Dan Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China.,Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Xiaobo Zhou
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China.,Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Lin Shi
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China.,Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Enqi Liu
- Institute of Cardiovascular Science, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Liang Bai
- Institute of Cardiovascular Science, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Chenming Sun
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China.,Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, China.,Xi'an Key Laboratory of Immune Related Diseases, Xi'an, China
| | - Baojun Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China.,Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, China.,Xi'an Key Laboratory of Immune Related Diseases, Xi'an, China
| |
Collapse
|
19
|
Li K, Zhao B, Wei D, Wang W, Cui Y, Qian L, Liu G. miR‑146a improves hepatic lipid and glucose metabolism by targeting MED1. Int J Mol Med 2019; 45:543-555. [PMID: 31894315 PMCID: PMC6984781 DOI: 10.3892/ijmm.2019.4443] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 11/19/2019] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is one of the most common chronic liver diseases worldwide. Increasing evidence has shown that microRNAs (miRNAs) play a vital role in the progression of NAFLD. The aim of the present study was to examine the expression level and roles of miR-146a in fatty liver of high-fat diet (HFD) and ob/ob mice and fatty acid-treated hepatic cells using RT-qPCR and western blot analysis. The results showed that the expression of miR-146a was significantly decreased in the livers of high-fat diet (HFD) and ob/ob mice and free fatty acid-stimulated cells by RT-qPCR. Overexpression of hepatic miR-146a improved glucose and insulin tolerance as well as lipid accumulation in the liver by promoting the oxidative metabolism of fatty acids. In addition, the overexpression of miR-146a increased the amount of mitochondria and promoted mitochondrial respiration in hepatocytes. Similarly, inhibition of miR-146a expression levels significantly reduced mitochondrial numbers in AML12 cells as well as the expression of mitochondrial respiration related genes. Additionally, MED1 was a direct target of miR-146a and restoring MED1 abolished the metabolic effects of miR-146a on lipid metabolism and mitochondrial function. Therefore, results of the present study identified a novel function of miR-146a in glucose and lipid metabolism in targeting MED1, suggesting that miR-146a serves as a potential therapeutic target for metabolic syndrome disease.
Collapse
Affiliation(s)
- Kun Li
- Department of Biomedical and Health Science, School of Life and Health Science, Anhui Science and Technology University, Fengyang, Anhui 233100, P.R. China
| | - Bao Zhao
- Department of Otorhinolaryngology Head and Neck Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Diandian Wei
- Department of Biomedical and Health Science, School of Life and Health Science, Anhui Science and Technology University, Fengyang, Anhui 233100, P.R. China
| | - Wenrui Wang
- Department of Biotechnology, School of Life Science and Technology, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Yixuan Cui
- Department of Otorhinolaryngology Head and Neck Surgery, First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233004, P.R. China
| | - Lisheng Qian
- Department of Biomedical and Health Science, School of Life and Health Science, Anhui Science and Technology University, Fengyang, Anhui 233100, P.R. China
| | - Guodong Liu
- Department of Biomedical and Health Science, School of Life and Health Science, Anhui Science and Technology University, Fengyang, Anhui 233100, P.R. China
| |
Collapse
|
20
|
Huck I, Gunewardena S, Espanol-Suner R, Willenbring H, Apte U. Hepatocyte Nuclear Factor 4 Alpha Activation Is Essential for Termination of Liver Regeneration in Mice. Hepatology 2019; 70:666-681. [PMID: 30520062 PMCID: PMC6551324 DOI: 10.1002/hep.30405] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 11/23/2018] [Indexed: 12/14/2022]
Abstract
Hepatocyte nuclear factor 4 alpha (HNF4α) is critical for hepatic differentiation. Recent studies have highlighted its role in inhibition of hepatocyte proliferation and tumor suppression. However, the role of HNF4α in liver regeneration (LR) is not known. We hypothesized that hepatocytes modulate HNF4α activity when navigating between differentiated and proliferative states during LR. Western blotting analysis revealed a rapid decline in nuclear and cytoplasmic HNF4α protein levels, accompanied with decreased target gene expression, within 1 hour after two-thirds partial hepatectomy (post-PH) in C57BL/6J mice. HNF4α protein expression did not recover to pre-PH levels until day 3. Hepatocyte-specific deletion of HNF4α (HNF4α-KO [knockout]) in mice resulted in 100% mortality post-PH, despite increased proliferative marker expression throughout regeneration. Sustained loss of HNF4α target gene expression throughout regeneration indicated that HNF4α-KO mice were unable to compensate for loss of HNF4α transcriptional activity. Deletion of HNF4α resulted in sustained proliferation accompanied by c-Myc and cyclin D1 overexpression and a complete deficiency of hepatocyte function after PH. Interestingly, overexpression of degradation-resistant HNF4α in hepatocytes delayed, but did not prevent, initiation of regeneration after PH. Finally, adeno-associated virus serotype 8 (AAV8)-mediated reexpression of HNF4α in hepatocytes of HNF4α-KO mice post-PH restored HNF4α protein levels, induced target gene expression, and improved survival of HNF4α-KO mice post-PH. Conclusion: In conclusion, these data indicate that HNF4α reexpression following initial decrease is critical for hepatocytes to exit from cell cycle and resume function during the termination phase of LR. These results indicate the role of HNF4α in LR and have implications for therapy of liver failure.
Collapse
Affiliation(s)
- Ian Huck
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS
| | - Sumedha Gunewardena
- Department of Biostatistics University of Kansas Medical Center, Kansas City, KS
| | | | - Holger Willenbring
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research,Liver Center, Division of Transplantation, University of California San Francisco, San Francisco, CA,Department of Surgery, Division of Transplantation, University of California San Francisco, San Francisco, CA
| | - Udayan Apte
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS
| |
Collapse
|
21
|
Shete V, Liu N, Jia Y, Viswakarma N, Reddy JK, Thimmapaya B. Mouse Cardiac Pde1C Is a Direct Transcriptional Target of Pparα. Int J Mol Sci 2018; 19:ijms19123704. [PMID: 30469494 PMCID: PMC6321386 DOI: 10.3390/ijms19123704] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 11/16/2018] [Accepted: 11/16/2018] [Indexed: 12/29/2022] Open
Abstract
Phosphodiesterase 1C (PDE1C) is expressed in mammalian heart and regulates cardiac functions by controlling levels of second messenger cyclic AMP and cyclic GMP (cAMP and cGMP, respectively). However, molecular mechanisms of cardiac Pde1c regulation are currently unknown. In this study, we demonstrate that treatment of wild type mice and H9c2 myoblasts with Wy-14,643, a potent ligand of nuclear receptor peroxisome-proliferator activated receptor alpha (PPARα), leads to elevated cardiac Pde1C mRNA and cardiac PDE1C protein, which correlate with reduced levels of cAMP. Furthermore, using mice lacking either Pparα or cardiomyocyte-specific Med1, the major subunit of Mediator complex, we show that Wy-14,643-mediated Pde1C induction fails to occur in the absence of Pparα and Med1 in the heart. Finally, using chromatin immunoprecipitation assays we demonstrate that PPARα binds to the upstream Pde1C promoter sequence on two sites, one of which is a palindrome sequence (agcTAGGttatcttaacctagc) that shows a robust binding. Based on these observations, we conclude that cardiac Pde1C is a direct transcriptional target of PPARα and that Med1 may be required for the PPARα mediated transcriptional activation of cardiac Pde1C.
Collapse
Affiliation(s)
- Varsha Shete
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Ning Liu
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Yuzhi Jia
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Navin Viswakarma
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612, USA.
| | - Janardan K Reddy
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Bayar Thimmapaya
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
22
|
Nagpal N, Sharma S, Maji S, Durante G, Ferracin M, Thakur JK, Kulshreshtha R. Essential role of MED1 in the transcriptional regulation of ER-dependent oncogenic miRNAs in breast cancer. Sci Rep 2018; 8:11805. [PMID: 30087366 PMCID: PMC6081450 DOI: 10.1038/s41598-018-29546-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 07/12/2018] [Indexed: 01/24/2023] Open
Abstract
Mediator complex has been extensively shown to regulate the levels of several protein-coding genes; however, its role in the regulation of miRNAs in humans remains unstudied so far. Here we show that MED1, a Mediator subunit in the Middle module of Mediator complex, is overexpressed in breast cancer and is a negative prognostic factor. The levels of several miRNAs (miR-100-5p, -191-5p, -193b-3p, -205-5p, -326, -422a and -425-5p) were found to be regulated by MED1. MED1 induces miR-191/425 cluster in an estrogen receptor-alpha (ER-α) dependent manner. Occupancy of MED1 on estrogen response elements (EREs) upstream of miR-191/425 cluster is estrogen and ER-α-dependent and ER-α-induced expression of these miRNAs is MED1-dependent. MED1 mediates induction of cell proliferation and migration and the genes associated with it (JUN, FOS, EGFR, VEGF, MMP1, and ERBB4) in breast cancer, which is abrogated when used together with miR-191-inhibition. Additionally, we show that MED1 also regulates the levels of direct miR-191 target genes such as SATB1, CDK6 and BDNF. Overall, the results show that MED1/ER-α/miR-191 axis promotes breast cancer cell proliferation and migration and may serve as a novel target for therapy.
Collapse
Affiliation(s)
- Neha Nagpal
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi, 110016, India.,Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Shivani Sharma
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi, 110016, India
| | - Sourobh Maji
- National Institute of Plant Genome Research, Aruna Asaf Ali Marg, New Delhi, 110067, India
| | - Giorgio Durante
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126, Bologna, Italy
| | - Manuela Ferracin
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, 40126, Bologna, Italy
| | - Jitendra K Thakur
- National Institute of Plant Genome Research, Aruna Asaf Ali Marg, New Delhi, 110067, India.
| | - Ritu Kulshreshtha
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, New Delhi, 110016, India.
| |
Collapse
|
23
|
Biological processes and signal transduction pathways regulated by the protein methyltransferase SETD7 and their significance in cancer. Signal Transduct Target Ther 2018; 3:19. [PMID: 30013796 PMCID: PMC6043541 DOI: 10.1038/s41392-018-0017-6] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 02/05/2018] [Accepted: 03/21/2018] [Indexed: 02/07/2023] Open
Abstract
Protein methyltransferases have been shown to methylate histone and non-histone proteins, leading to regulation of several biological processes that control cell homeostasis. Over the past few years, the histone-lysine N-methyltransferase SETD7 (SETD7; also known as SET7/9, KIAA1717, KMT7, SET7, SET9) has emerged as an important regulator of at least 30 non-histone proteins and a potential target for the treatment of several human diseases. This review discusses current knowledge of the structure and subcellular localization of SETD7, as well as its function as a histone and non-histone methyltransferase. This work also underlines the putative contribution of SETD7 to the regulation of gene expression, control of cell proliferation, differentiation and endoplasmic reticulum stress, which indicate that SETD7 is a candidate for novel targeted therapies with the aim of either stimulating or inhibiting its activity, depending on the cell signaling context.
Collapse
|
24
|
Jia Y, Liu N, Viswakarma N, Sun R, Schipma MJ, Shang M, Thorp EB, Kanwar YS, Thimmapaya B, Reddy JK. PIMT/NCOA6IP Deletion in the Mouse Heart Causes Delayed Cardiomyopathy Attributable to Perturbation in Energy Metabolism. Int J Mol Sci 2018; 19:ijms19051485. [PMID: 29772707 PMCID: PMC5983783 DOI: 10.3390/ijms19051485] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 05/03/2018] [Accepted: 05/09/2018] [Indexed: 01/09/2023] Open
Abstract
PIMT/NCOA6IP, a transcriptional coactivator PRIP/NCOA6 binding protein, enhances nuclear receptor transcriptional activity. Germline disruption of PIMT results in early embryonic lethality due to impairment of development around blastocyst and uterine implantation stages. We now generated mice with Cre-mediated cardiac-specific deletion of PIMT (csPIMT−/−) in adult mice. These mice manifest enlargement of heart, with nearly 100% mortality by 7.5 months of age due to dilated cardiomyopathy. Significant reductions in the expression of genes (i) pertaining to mitochondrial respiratory chain complexes I to IV; (ii) calcium cycling cardiac muscle contraction (Atp2a1, Atp2a2, Ryr2); and (iii) nuclear receptor PPAR- regulated genes involved in glucose and fatty acid energy metabolism were found in csPIMT−/− mouse heart. Elevated levels of Nppa and Nppb mRNAs were noted in csPIMT−/− heart indicative of myocardial damage. These hearts revealed increased reparative fibrosis associated with enhanced expression of Tgfβ2 and Ctgf. Furthermore, cardiac-specific deletion of PIMT in adult mice, using tamoxifen-inducible Cre-approach (TmcsPIMT−/−), results in the development of cardiomyopathy. Thus, cumulative evidence suggests that PIMT functions in cardiac energy metabolism by interacting with nuclear receptor coactivators and this property could be useful in the management of heart failure.
Collapse
Affiliation(s)
- Yuzhi Jia
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Ning Liu
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Navin Viswakarma
- Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, IL 60612, USA.
| | - Ruya Sun
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Mathew J Schipma
- Next Generation Sequencing Core Facility, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Meng Shang
- Feinberg Cardiovascular Research Institute and Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Edward B Thorp
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Yashpal S Kanwar
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Bayar Thimmapaya
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| | - Janardan K Reddy
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA.
| |
Collapse
|
25
|
Amoasii L, Olson EN, Bassel-Duby R. Control of Muscle Metabolism by the Mediator Complex. Cold Spring Harb Perspect Med 2018; 8:cshperspect.a029843. [PMID: 28432117 DOI: 10.1101/cshperspect.a029843] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Exercise represents an energetic challenge to whole-body homeostasis. In skeletal muscle, exercise activates a variety of signaling pathways that culminate in the nucleus to regulate genes involved in metabolism and contractility; however, much remains to be learned about the transcriptional effectors of exercise. Mediator is a multiprotein complex that links signal-dependent transcription factors and other transcriptional regulators with the basal transcriptional machinery, thereby serving as a transcriptional "hub." In this article, we discuss recent studies highlighting the role of Mediator subunits in metabolic regulation and glucose metabolism, as well as exercise responsiveness. Elucidation of the roles of Mediator subunits in metabolic control has revealed new mechanisms and molecular targets for the modulation of metabolism and metabolic disorders.
Collapse
Affiliation(s)
- Leonela Amoasii
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, and Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, Texas 7539-9148
| | - Eric N Olson
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, and Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, Texas 7539-9148
| | - Rhonda Bassel-Duby
- Department of Molecular Biology, Hamon Center for Regenerative Science and Medicine, and Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, Texas 7539-9148
| |
Collapse
|
26
|
Gautam S, Singh P, Singh M, Roy S, Rawat JK, Yadav RK, Devi U, Gupta PS, Saraf SA, Kaithwas G. Rifaximin, a pregnane X receptor (PXR) activator regulates apoptosis in a murine model of breast cancer. RSC Adv 2018; 8:3512-3521. [PMID: 35542911 PMCID: PMC9077680 DOI: 10.1039/c7ra09689e] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 01/05/2018] [Indexed: 12/29/2022] Open
Abstract
The present study was proposed to investigate the effect of rifaximin (RFX) on methyl nitrosourea (MNU) induced mammary gland carcinoma in albino wistar rats.
Collapse
|
27
|
Ning R, Zhan Y, He S, Hu J, Zhu Z, Hu G, Yan B, Yang J, Liu W. Interleukin-6 Induces DEC1, Promotes DEC1 Interaction with RXRα and Suppresses the Expression of PXR, CAR and Their Target Genes. Front Pharmacol 2017; 8:866. [PMID: 29234281 PMCID: PMC5712319 DOI: 10.3389/fphar.2017.00866] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Accepted: 11/09/2017] [Indexed: 12/17/2022] Open
Abstract
Inflammatory burden is a primary cellular event in many liver diseases, and the overall capacity of drug elimination is decreased. PXR (pregnane X receptor) and CAR (constitutive androstane receptor) are two master regulators of genes encoding drug-metabolizing enzymes and transporters. DEC1 (differentiated embryonic chondrocyte-expressed gene 1) is a ligand-independent transcription factor and reportedly is induced by many inflammatory cytokines including IL-6. In this study, we used primary hepatocytes (human and mouse) as well as HepG2 cell line and demonstrated that IL-6 increased DEC1 expression and decreased the expressions of PXR, CAR, and their target genes. Overexpression of DEC1 had similar effect as IL-6 on the expression of these genes, and knockdown of DEC1 reversed their downregulation by IL-6. Interestingly, neither IL-6 nor DEC1 altered the expression of RXRα, a common dimerization partner for many nuclear receptors including PXR and CAR. Instead, DEC1 was found to interact with RXRα and IL-6 enhanced the interaction. These results conclude that DEC1 uses diverse mechanisms of action and supports IL-6 downregulation of drug-elimination genes and their regulators.
Collapse
Affiliation(s)
- Rui Ning
- Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Yunran Zhan
- Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Shuangcheng He
- Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Jinhua Hu
- Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Zhu Zhu
- Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Gang Hu
- Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Bingfang Yan
- Pharmaceutical Sciences, Center for Pharmacogenomics and Molecular Therapy, University of Rhode Island, Kingston, RI, United States
| | - Jian Yang
- Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Wei Liu
- Department of Pharmacology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
28
|
Mansouri S, Naghavi-Al-Hosseini F, Farahmand L, Majidzadeh-A K. MED1 may explain the interaction between receptor tyrosine kinases and ERα66 in the complicated network of Tamoxifen resistance. Eur J Pharmacol 2017; 804:78-81. [PMID: 28322840 DOI: 10.1016/j.ejphar.2017.03.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 03/09/2017] [Accepted: 03/15/2017] [Indexed: 02/07/2023]
Abstract
According to the American Society of Clinical Oncology or ASCO's clinical practice guidelines, administration of Tamoxifen for hormone receptor positive patients improved outcomes. However, many studies have been conducted in this issue, with the rise of Tamoxifen resistance in recent decades. There are many alternative growth cascades that are activated in Tamoxifen resistant cells. The most common and well characterized components of such a resistant network are receptor tyrosine kinases, or RTKs, which can influence many other cellular processes. The interactions between estrogen dependent and independent pathways further complicate the networking. MED1, as a member of a mediator complex, which is activated by RTK growth pathways, plays role in co-activating ERα66 to transcribe genes and enhance cellular proliferation. Herein, we will discuss MED1, a novel biomarker which can explain how RTKs interact with ERα66 which results in Tamoxifen resistance.
Collapse
Affiliation(s)
- Sepideh Mansouri
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Fateme Naghavi-Al-Hosseini
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Leila Farahmand
- Recombinant Proteins Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Keivan Majidzadeh-A
- Cancer Genetics Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran.
| |
Collapse
|
29
|
Regulation of metabolism by the Mediator complex. BIOPHYSICS REPORTS 2016; 2:69-77. [PMID: 28018965 PMCID: PMC5138257 DOI: 10.1007/s41048-016-0031-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 02/15/2016] [Indexed: 01/11/2023] Open
Abstract
The Mediator complex was originally discovered in yeast, but it is conserved in all eukaryotes. Its best-known function is to regulate RNA polymerase II-dependent gene transcription. Although the mechanisms by which the Mediator complex regulates transcription are often complicated by the context-dependent regulation, this transcription cofactor complex plays a pivotal role in numerous biological pathways. Biochemical, molecular, and physiological studies using cancer cell lines or model organisms have established the current paradigm of the Mediator functions. However, the physiological roles of the mammalian Mediator complex remain poorly defined, but have attracted a great interest in recent years. In this short review, we will summarize some of the reported functions of selective Mediator subunits in the regulation of metabolism. These intriguing findings suggest that the Mediator complex may be an important player in nutrient sensing and energy balance in mammals.
Collapse
|
30
|
Jia Y, Chang HC, Schipma MJ, Liu J, Shete V, Liu N, Sato T, Thorp EB, Barger PM, Zhu YJ, Viswakarma N, Kanwar YS, Ardehali H, Thimmapaya B, Reddy JK. Cardiomyocyte-Specific Ablation of Med1 Subunit of the Mediator Complex Causes Lethal Dilated Cardiomyopathy in Mice. PLoS One 2016; 11:e0160755. [PMID: 27548259 PMCID: PMC4993490 DOI: 10.1371/journal.pone.0160755] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 07/25/2016] [Indexed: 11/19/2022] Open
Abstract
Mediator, an evolutionarily conserved multi-protein complex consisting of about 30 subunits, is a key component of the polymerase II mediated gene transcription. Germline deletion of the Mediator subunit 1 (Med1) of the Mediator in mice results in mid-gestational embryonic lethality with developmental impairment of multiple organs including heart. Here we show that cardiomyocyte-specific deletion of Med1 in mice (csMed1-/-) during late gestational and early postnatal development by intercrossing Med1fl/fl mice to α-MyHC-Cre transgenic mice results in lethality within 10 days after weaning due to dilated cardiomyopathy-related ventricular dilation and heart failure. The csMed1-/- mouse heart manifests mitochondrial damage, increased apoptosis and interstitial fibrosis. Global gene expression analysis revealed that loss of Med1 in heart down-regulates more than 200 genes including Acadm, Cacna1s, Atp2a2, Ryr2, Pde1c, Pln, PGC1α, and PGC1β that are critical for calcium signaling, cardiac muscle contraction, arrhythmogenic right ventricular cardiomyopathy, dilated cardiomyopathy and peroxisome proliferator-activated receptor regulated energy metabolism. Many genes essential for oxidative phosphorylation and proper mitochondrial function such as genes coding for the succinate dehydrogenase subunits of the mitochondrial complex II are also down-regulated in csMed1-/- heart contributing to myocardial injury. Data also showed up-regulation of about 180 genes including Tgfb2, Ace, Atf3, Ctgf, Angpt14, Col9a2, Wisp2, Nppa, Nppb, and Actn1 that are linked to cardiac muscle contraction, cardiac hypertrophy, cardiac fibrosis and myocardial injury. Furthermore, we demonstrate that cardiac specific deletion of Med1 in adult mice using tamoxifen-inducible Cre approach (TmcsMed1-/-), results in rapid development of cardiomyopathy and death within 4 weeks. We found that the key findings of the csMed1-/- studies described above are highly reproducible in TmcsMed1-/- mouse heart. Collectively, these observations suggest that Med1 plays a critical role in the maintenance of heart function impacting on multiple metabolic, compensatory and reparative pathways with a likely therapeutic potential in the management of heart failure.
Collapse
MESH Headings
- Animals
- Apoptosis
- Cadherins/genetics
- Cadherins/metabolism
- Calcium Channels, L-Type/genetics
- Calcium Channels, L-Type/metabolism
- Calcium Signaling
- Cardiomyopathy, Dilated/genetics
- Cardiomyopathy, Dilated/metabolism
- Cardiomyopathy, Dilated/pathology
- Cyclic Nucleotide Phosphodiesterases, Type 1/genetics
- Cyclic Nucleotide Phosphodiesterases, Type 1/metabolism
- Embryo, Mammalian
- Energy Metabolism
- Female
- Gene Deletion
- Gene Expression Profiling
- Gene Expression Regulation
- Genes, Lethal
- Gestational Age
- Heart Failure/genetics
- Heart Failure/metabolism
- Heart Failure/pathology
- Mediator Complex Subunit 1/deficiency
- Mediator Complex Subunit 1/genetics
- Mice
- Mice, Knockout
- Mitochondria/metabolism
- Mitochondria/pathology
- Myocardial Contraction
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Peroxisome Proliferator-Activated Receptors/genetics
- Peroxisome Proliferator-Activated Receptors/metabolism
- Pregnancy
- Ryanodine Receptor Calcium Release Channel/genetics
- Ryanodine Receptor Calcium Release Channel/metabolism
- Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics
- Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism
Collapse
Affiliation(s)
- Yuzhi Jia
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Hsiang-Chun Chang
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Matthew J. Schipma
- Next Generation Sequencing Core Facility, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Jing Liu
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Varsha Shete
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Ning Liu
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Tatsuya Sato
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Edward B. Thorp
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Philip M. Barger
- Center for Cardiovascular Research, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Yi-Jun Zhu
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Navin Viswakarma
- Department of Surgery, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Yashpal S. Kanwar
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Hossein Ardehali
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Bayar Thimmapaya
- Department of Microbiology and Immunology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
- * E-mail: (JKR); (BT)
| | - Janardan K. Reddy
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
- * E-mail: (JKR); (BT)
| |
Collapse
|
31
|
Amoasii L, Holland W, Sanchez-Ortiz E, Baskin KK, Pearson M, Burgess SC, Nelson BR, Bassel-Duby R, Olson EN. A MED13-dependent skeletal muscle gene program controls systemic glucose homeostasis and hepatic metabolism. Genes Dev 2016; 30:434-46. [PMID: 26883362 PMCID: PMC4762428 DOI: 10.1101/gad.273128.115] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Amoasii et al. found that skeletal muscle-specific deletion of the Mediator subunit MED13 in mice conferred resistance to hepatic steatosis by activating a metabolic gene program that enhances muscle glucose uptake and storage as glycogen. MED13 suppressed expression of genes involved in glucose uptake and metabolism in skeletal muscle by inhibiting the nuclear receptor NURR1 and the MEF2 transcription factor. The Mediator complex governs gene expression by linking upstream signaling pathways with the basal transcriptional machinery. However, how individual Mediator subunits may function in different tissues remains to be investigated. Through skeletal muscle-specific deletion of the Mediator subunit MED13 in mice, we discovered a gene regulatory mechanism by which skeletal muscle modulates the response of the liver to a high-fat diet. Skeletal muscle-specific deletion of MED13 in mice conferred resistance to hepatic steatosis by activating a metabolic gene program that enhances muscle glucose uptake and storage as glycogen. The consequent insulin-sensitizing effect within skeletal muscle lowered systemic glucose and insulin levels independently of weight gain and adiposity and prevented hepatic lipid accumulation. MED13 suppressed the expression of genes involved in glucose uptake and metabolism in skeletal muscle by inhibiting the nuclear receptor NURR1 and the MEF2 transcription factor. These findings reveal a fundamental molecular mechanism for the governance of glucose metabolism and the control of hepatic lipid accumulation by skeletal muscle. Intriguingly, MED13 exerts opposing metabolic actions in skeletal muscle and the heart, highlighting the customized, tissue-specific functions of the Mediator complex.
Collapse
Affiliation(s)
- Leonela Amoasii
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA; Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - William Holland
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Efrain Sanchez-Ortiz
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA; Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Kedryn K Baskin
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA; Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Mackenzie Pearson
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Shawn C Burgess
- Advanced Imaging Research Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA; Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Benjamin R Nelson
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA; Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Rhonda Bassel-Duby
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA; Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| | - Eric N Olson
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA; Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA
| |
Collapse
|
32
|
Motojima K. [Lessons Learned after 25 Years of Studies on the Physiological Roles of the Peroxisome Proliferator-activated Receptor]. YAKUGAKU ZASSHI 2015; 135:1083-9. [PMID: 26423862 DOI: 10.1248/yakushi.15-00150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The peroxisome proliferator-activated receptor (PPAR) was discovered a quarter of a century ago. PPAR was soon recognized as a general transcriptional regulator of lipid homeostasis, and several hypolipidemic and antidiabetic agents were shown to be effective ligands for it. Since then, many attempts to develop more potent drugs have been made worldwide, but most were unsuccessful due to serious side effects. It appears that the PPAR boom has ended. This review summarizes the short history of PPAR studies, including our own results, and discusses the lessons learned from the rise and fall of studies in this field for next-generation basic studies and drug development research.
Collapse
|
33
|
Li G, L. Guo G. Farnesoid X receptor, the bile acid sensing nuclear receptor, in liver regeneration. Acta Pharm Sin B 2015; 5:93-8. [PMID: 26579433 PMCID: PMC4629218 DOI: 10.1016/j.apsb.2015.01.005] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 01/02/2015] [Accepted: 01/05/2015] [Indexed: 01/19/2023] Open
Abstract
The liver is unique in regenerative potential, which could recover the lost mass and function after injury from ischemia and resection. The underlying molecular mechanisms of liver regeneration have been extensively studied in the past using the partial hepatectomy (PH) model in rodents, where 2/3 PH is carried out by removing two lobes. The whole process of liver regeneration is complicated, orchestrated event involving a network of connected interactions, which still remain fully elusive. Bile acids (BAs) are ligands of farnesoid X receptor (FXR), a nuclear receptor of ligand-activated transcription factor. FXR has been shown to be highly involved in liver regeneration. BAs and FXR not only interact with each other but also regulate various downstream targets independently during liver regeneration. Moreover, recent findings suggest that tissue-specific FXR also contributes to liver regeneration significantly. These novel findings suggest that FXR has much broader role than regulating BA, cholesterol, lipid and glucose metabolism. Therefore, these researches highlight FXR as an important pharmaceutical target for potential use of FXR ligands to regulate liver regeneration in clinic. This review focuses on the roles of BAs and FXR in liver regeneration and the current underlying molecular mechanisms which contribute to liver regeneration.
Collapse
Key Words
- ABC, ATP-binding cassette
- AMPK, AMP-activated protein kinase
- BA, bile acid
- Bile acids
- C/EBPβ, CCAAT-enhancer binding protein β
- CA, cholic acid
- CDCA, chenodeoxycholic acid
- CTX, cerebrotendinous xanthomatosis
- CYP7A1, cholesterol 7alpha-hydroxylase
- CYP8B1, sterol 12α-hydroxylase
- Cyp27-KO, sterol 27-hydroxylase–knockout
- DDAH-1, dimethylarginineaminohydrolase-1
- ERK1/2, extracellular signal-regulated kinase 1/2
- FGF-15, fibroblast growth factor 15
- FGFR4, FGF receptor 4
- FOXM1b, forkhead boxm1b
- FXR, farnesoid X receptor
- Farnesoid X receptor
- Fibroblast growth factor 15
- Fxr-KO, Fxr-knockout
- GPBAR1 or TGR5, G protein-coupled BA receptor 1
- HEX, hematopoietically expressed homeobox
- JNK, c-Jun N-terminal kinase
- KC, Kupffer cells
- KO, knockout
- Liver regeneration
- Liver-intestine croass talk
- MAPK, mitogen-activated protein kinase
- MRP3, multidrug resistance associated protein 3
- NASH, nonalcoholic steatohepatitis
- NF-κB, nuclear factor-κB
- PH, partial hepatectomy
- Rb, retinoblastoma
- SHP, small heterodimer partner
- STAT3, signal transducer and activator of transcription 3
- TH, thyroid hormone
- THR, TH receptor
- Transmembrane G protein coupled receptor 5
- WT, wild type
- cAMP, cyclic adenosine monophosphate
- hepFxr-KO, hepatocyte-specific Fxr knockout
Collapse
|