1
|
Huang L, Fu Y, Cao J, Zhai J. Clinical Value and Mechanism Exploration of Serum miR-379 in Obesity-Polycystic Ovary Syndrome. Int J Womens Health 2024; 16:1149-1157. [PMID: 38919684 PMCID: PMC11198001 DOI: 10.2147/ijwh.s427997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 10/23/2023] [Indexed: 06/27/2024] Open
Abstract
Objective As a common endocrine and metabolic disorder, polycystic ovary syndrome (PCOS) is mostly associated with an obese phenotype. The present research focuses on the clinical significance of miR-379 in obesity-PCOS and attempts to elucidate its potential mechanisms. Methods Healthy individuals (n = 46), obesity-PCOS (n = 92), and non-obesity PCOS (n = 31) subjects were enrolled. Quantitative real-time polymerase chain reaction (qRT-PCR) was conducted to examine the level of serum miR-379. The receiver operating characteristic (ROC) curve and logistic regressions were applied to reveal the diagnostic significance. Dual luciferase reporters were performed to validate the targeting relationships. And cell count kit (CCK-8) assay was used to detect cell proliferation. Results Serum miR-379 was highly expressed in PCOS patients (P < 0.05), in especially obesity-PCOS patients. Higher miR-379 was associated with greater body mass index (BMI), higher bioavailable testosterone (bT), and greater insulin resistance (IR). Additionally, miR-379 was an independent risk factor for the development of obesity-PCOS. The sensitivity of miR-379 in identifying patients with obesity-PCOS from healthy or non-obesity-PCSO patients was 81.52% and 72.83%, and the specificity was 86.96% and 80.65%. Semaphorin 3 A (SEMA3A) was identified as a target of miR-379 and was reduced in the patients with obesity PCOS (P < 0.05). Inhibition of miR-375 reduced KGN proliferation, but this reduction was partially restored by silencing of SEMA3A (P < 0.05). Conclusion Elevated miR-379 assists the diagnosis of obesity-PCOS and regulates the proliferation of KGN by targeting SEMA3A engaged in disease development.
Collapse
Affiliation(s)
- Lu Huang
- Department of Obstetrics and Gynecology, Beijing Tongren Hospital, Capital Medical University, Beijing100730, People’s Republic of China
| | - Yujing Fu
- Department of Obstetrics and Gynecology, Beijing Tongren Hospital, Capital Medical University, Beijing100730, People’s Republic of China
| | - Jinghong Cao
- Department of Obstetrics and Gynecology, Beijing Tongren Hospital, Capital Medical University, Beijing100730, People’s Republic of China
| | - Jianjun Zhai
- Department of Obstetrics and Gynecology, Beijing Tongren Hospital, Capital Medical University, Beijing100730, People’s Republic of China
| |
Collapse
|
2
|
Paes T, Feelders RA, Hofland LJ. Epigenetic Mechanisms Modulated by Glucocorticoids With a Focus on Cushing Syndrome. J Clin Endocrinol Metab 2024; 109:e1424-e1433. [PMID: 38517306 PMCID: PMC11099489 DOI: 10.1210/clinem/dgae151] [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: 08/22/2023] [Revised: 02/08/2024] [Accepted: 03/07/2024] [Indexed: 03/23/2024]
Abstract
In Cushing syndrome (CS), prolonged exposure to high cortisol levels results in a wide range of devastating effects causing multisystem morbidity. Despite the efficacy of treatment leading to disease remission and clinical improvement, hypercortisolism-induced complications may persist. Since glucocorticoids use the epigenetic machinery as a mechanism of action to modulate gene expression, the persistence of some comorbidities may be mediated by hypercortisolism-induced long-lasting epigenetic changes. Additionally, glucocorticoids influence microRNA expression, which is an important epigenetic regulator as it modulates gene expression without changing the DNA sequence. Evidence suggests that chronically elevated glucocorticoid levels may induce aberrant microRNA expression which may impact several cellular processes resulting in cardiometabolic disorders. The present article reviews the evidence on epigenetic changes induced by (long-term) glucocorticoid exposure. Key aspects of some glucocorticoid-target genes and their implications in the context of CS are described. Lastly, the effects of epigenetic drugs influencing glucocorticoid effects are discussed for their ability to be potentially used as adjunctive therapy in CS.
Collapse
Affiliation(s)
- Ticiana Paes
- Department of Internal Medicine, Division of Endocrinology, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital and Harvard Medical School, Boston 02115, MA, USA
| | - Richard A Feelders
- Department of Internal Medicine, Division of Endocrinology, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Leo J Hofland
- Department of Internal Medicine, Division of Endocrinology, Erasmus Medical Center, 3015 GD Rotterdam, The Netherlands
| |
Collapse
|
3
|
Sanceau J, Poupel L, Joubel C, Lagoutte I, Caruso S, Pinto S, Desbois-Mouthon C, Godard C, Hamimi A, Montmory E, Dulary C, Chantalat S, Roehrig A, Muret K, Saint-Pierre B, Deleuze JF, Mouillet-Richard S, Forné T, Grosset CF, Zucman-Rossi J, Colnot S, Gougelet A. DLK1/DIO3 locus upregulation by a β-catenin-dependent enhancer drives cell proliferation and liver tumorigenesis. Mol Ther 2024; 32:1125-1143. [PMID: 38311851 PMCID: PMC11163201 DOI: 10.1016/j.ymthe.2024.01.036] [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: 12/01/2023] [Revised: 01/05/2024] [Accepted: 01/31/2024] [Indexed: 02/06/2024] Open
Abstract
The CTNNB1 gene, encoding β-catenin, is frequently mutated in hepatocellular carcinoma (HCC, ∼30%) and in hepatoblastoma (HB, >80%), in which DLK1/DIO3 locus induction is correlated with CTNNB1 mutations. Here, we aim to decipher how sustained β-catenin activation regulates DLK1/DIO3 locus expression and the role this locus plays in HB and HCC development in mouse models deleted for Apc (ApcΔhep) or Ctnnb1-exon 3 (β-cateninΔExon3) and in human CTNNB1-mutated hepatic cancer cells. We identified an enhancer site bound by TCF-4/β-catenin complexes in an open conformation upon sustained β-catenin activation (DLK1-Wnt responsive element [WRE]) and increasing DLK1/DIO3 locus transcription in β-catenin-mutated human HB and mouse models. DLK1-WRE editing by CRISPR-Cas9 approach impaired DLK1/DIO3 locus expression and slowed tumor growth in subcutaneous CTNNB1-mutated tumor cell grafts, ApcΔhep HB and β-cateninΔExon3 HCC. Tumor growth inhibition resulted either from increased FADD expression and subsequent caspase-3 cleavage in the first case or from decreased expression of cell cycle actors regulated by FoxM1 in the others. Therefore, the DLK1/DIO3 locus is an essential determinant of FoxM1-dependent cell proliferation during β-catenin-driven liver tumorigenesis. Targeting the DLK1-WRE enhancer to silence the DLK1/DIO3 locus might thus represent an interesting therapeutic strategy to restrict tumor growth in primary liver cancers with CTNNB1 mutations.
Collapse
Affiliation(s)
- Julie Sanceau
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, F-75006 Paris, France; Team « Oncogenic functions of beta-catenin signaling in the liver », Équipe labellisée par la Ligue Nationale contre le Cancer, F-75013 Paris, France; APHP, Institut du Cancer Paris CARPEM, F-75015 Paris, France
| | - Lucie Poupel
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, F-75006 Paris, France; Team « Oncogenic functions of beta-catenin signaling in the liver », Équipe labellisée par la Ligue Nationale contre le Cancer, F-75013 Paris, France; APHP, Institut du Cancer Paris CARPEM, F-75015 Paris, France; Inovarion, F-75005 Paris, France
| | - Camille Joubel
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, F-75006 Paris, France; Team « Oncogenic functions of beta-catenin signaling in the liver », Équipe labellisée par la Ligue Nationale contre le Cancer, F-75013 Paris, France; APHP, Institut du Cancer Paris CARPEM, F-75015 Paris, France
| | - Isabelle Lagoutte
- University Paris Cité, Institut Cochin, INSERM, CNRS, F-75014 Paris, France
| | - Stefano Caruso
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, F-75006 Paris, France; APHP, Institut du Cancer Paris CARPEM, F-75015 Paris, France
| | - Sandra Pinto
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, F-75006 Paris, France; Team « Oncogenic functions of beta-catenin signaling in the liver », Équipe labellisée par la Ligue Nationale contre le Cancer, F-75013 Paris, France
| | - Christèle Desbois-Mouthon
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, F-75006 Paris, France; Team « Oncogenic functions of beta-catenin signaling in the liver », Équipe labellisée par la Ligue Nationale contre le Cancer, F-75013 Paris, France; APHP, Institut du Cancer Paris CARPEM, F-75015 Paris, France
| | - Cécile Godard
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, F-75006 Paris, France; Team « Oncogenic functions of beta-catenin signaling in the liver », Équipe labellisée par la Ligue Nationale contre le Cancer, F-75013 Paris, France; APHP, Institut du Cancer Paris CARPEM, F-75015 Paris, France
| | - Akila Hamimi
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, F-75006 Paris, France; Team « Oncogenic functions of beta-catenin signaling in the liver », Équipe labellisée par la Ligue Nationale contre le Cancer, F-75013 Paris, France; APHP, Institut du Cancer Paris CARPEM, F-75015 Paris, France
| | - Enzo Montmory
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, F-75006 Paris, France; Team « Oncogenic functions of beta-catenin signaling in the liver », Équipe labellisée par la Ligue Nationale contre le Cancer, F-75013 Paris, France; APHP, Institut du Cancer Paris CARPEM, F-75015 Paris, France
| | - Cécile Dulary
- Centre National de Génotypage, Institut de Génomique, CEA, F-91057 Evry, France
| | - Sophie Chantalat
- Centre National de Génotypage, Institut de Génomique, CEA, F-91057 Evry, France
| | - Amélie Roehrig
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, F-75006 Paris, France; APHP, Institut du Cancer Paris CARPEM, F-75015 Paris, France
| | - Kevin Muret
- Centre National de Génotypage, Institut de Génomique, CEA, F-91057 Evry, France
| | | | | | - Sophie Mouillet-Richard
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, F-75006 Paris, France; APHP, Institut du Cancer Paris CARPEM, F-75015 Paris, France
| | - Thierry Forné
- IGMM, University Montpellier, CNRS, F-34293 Montpellier, France
| | - Christophe F Grosset
- University Bordeaux, INSERM, Biotherapy of Genetic Diseases, Inflammatory Disorders and Cancer, BMGIC, U1035, MIRCADE team, F-33076 Bordeaux, France; University Bordeaux, INSERM, Bordeaux Institute in Oncology, BRIC, U1312, MIRCADE team, F-33076 Bordeaux, France
| | - Jessica Zucman-Rossi
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, F-75006 Paris, France; APHP, Institut du Cancer Paris CARPEM, F-75015 Paris, France
| | - Sabine Colnot
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, F-75006 Paris, France; Team « Oncogenic functions of beta-catenin signaling in the liver », Équipe labellisée par la Ligue Nationale contre le Cancer, F-75013 Paris, France; APHP, Institut du Cancer Paris CARPEM, F-75015 Paris, France
| | - Angélique Gougelet
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université Paris Cité, F-75006 Paris, France; Team « Oncogenic functions of beta-catenin signaling in the liver », Équipe labellisée par la Ligue Nationale contre le Cancer, F-75013 Paris, France; APHP, Institut du Cancer Paris CARPEM, F-75015 Paris, France.
| |
Collapse
|
4
|
Ma J, Bi J, Sun B, Li H, Li Y, Wang S. Zinc Improves Semen Parameters in High-Fat Diet-Induced Male Rats by Regulating the Expression of LncRNA in Testis Tissue. Biol Trace Elem Res 2023; 201:4793-4805. [PMID: 36600170 DOI: 10.1007/s12011-022-03550-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 12/28/2022] [Indexed: 01/06/2023]
Abstract
This study aimed to identify differentially expressed LncRNAs in testis tissue of male rats induced by high-fat diet and their changes after zinc supplementation, by constructing a high-fat feeding rat model, and then supplemented with zinc, and observed the expression of LncRNA in three groups of normal, high-fat fed, and zinc-intervened rats. Experimental studies show that the semen parameters of male rats with high-fat diet were decreased but recovered after zinc supplementation, and the related LncRNA also changed. Zinc may improve the high-fat diet-induced reduction of semen parameters by changing the expression of related LncRNA.
Collapse
Affiliation(s)
- Jing Ma
- Hebei Key Laboratory of Reproductive Medicine, Hebei Institute of Reproductive Health Science and Technology, No. 480 Heping Street, Shijiazhuang, 050071, Xinhua District, China
| | - Jiajie Bi
- Graduate School of Chengde Medical University, Chengde, 067000, China
| | - Bo Sun
- Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Huanhuan Li
- Hebei Key Laboratory of Reproductive Medicine, Hebei Institute of Reproductive Health Science and Technology, No. 480 Heping Street, Shijiazhuang, 050071, Xinhua District, China
| | - Yuejia Li
- Graduate School of Hebei Medical University, Shijiazhuang, 050017, China
| | - Shusong Wang
- Hebei Key Laboratory of Reproductive Medicine, Hebei Institute of Reproductive Health Science and Technology, No. 480 Heping Street, Shijiazhuang, 050071, Xinhua District, China.
- Graduate School of Chengde Medical University, Chengde, 067000, China.
- Graduate School of Hebei Medical University, Shijiazhuang, 050017, China.
| |
Collapse
|
5
|
Khalifa O, Ouararhni K, Errafii K, Alajez NM, Arredouani A. Targeted MicroRNA Profiling Reveals That Exendin-4 Modulates the Expression of Several MicroRNAs to Reduce Steatosis in HepG2 Cells. Int J Mol Sci 2023; 24:11606. [PMID: 37511368 PMCID: PMC10380891 DOI: 10.3390/ijms241411606] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 06/25/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023] Open
Abstract
Excess hepatic lipid accumulation is the hallmark of non-alcoholic fatty liver disease (NAFLD), for which no medication is currently approved. However, glucagon-like peptide-1 receptor agonists (GLP-1RAs), already approved for treating type 2 diabetes, have lately emerged as possible treatments. Herein we aim to investigate how the GLP-1RA exendin-4 (Ex-4) affects the microRNA (miRNAs) expression profile using an in vitro model of steatosis. Total RNA, including miRNAs, was isolated from control, steatotic, and Ex-4-treated steatotic cells and used for probing a panel of 799 highly curated miRNAs using NanoString technology. Enrichment pathway analysis was used to find the signaling pathways and cellular functions associated with the differentially expressed miRNAs. Our data shows that Ex-4 reversed the expression of a set of miRNAs. Functional enrichment analysis highlighted many relevant signaling pathways and cellular functions enriched in the differentially expressed miRNAs, including hepatic fibrosis, insulin receptor, PPAR, Wnt/β-Catenin, VEGF, and mTOR receptor signaling pathways, fibrosis of the liver, cirrhosis of the liver, proliferation of hepatic stellate cells, diabetes mellitus, glucose metabolism disorder and proliferation of liver cells. Our findings suggest that miRNAs may play essential roles in the processes driving steatosis reduction in response to GLP-1R agonists, which warrants further functional investigation.
Collapse
Affiliation(s)
- Olfa Khalifa
- Diabetes Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha P.O. Box 34110, Qatar
| | - Khalid Ouararhni
- Genomics Core Facility, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha P.O. Box 34110, Qatar
| | - Khaoula Errafii
- African Genome Center, Mohammed VI Polytechnic University (UM6P), Ben Guerir 43151, Morocco
| | - Nehad M Alajez
- Translational Cancer and Immunity Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha P.O. Box 34110, Qatar
- College of Health & Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, Doha P.O. Box 34110, Qatar
| | - Abdelilah Arredouani
- Diabetes Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha P.O. Box 34110, Qatar
- College of Health & Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, Doha P.O. Box 34110, Qatar
| |
Collapse
|
6
|
Demori I, Grasselli E. The Role of the Stress Response in Metabolic Dysfunction-Associated Fatty Liver Disease: A Psychoneuroendocrineimmunology-Based Perspective. Nutrients 2023; 15:nu15030795. [PMID: 36771500 PMCID: PMC9921904 DOI: 10.3390/nu15030795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/17/2023] [Accepted: 02/01/2023] [Indexed: 02/08/2023] Open
Abstract
The novel term metabolic dysfunction-associated fatty liver disease (MAFLD), which has been proposed to describe the major cause of hepatic disease, pinpoints the coexistence of multiple metabolic disturbances and liver steatosis, giving rise to different phenotypic manifestations. Within the psychoneuroendocrineimmunological (PNEI) network that regulates body-mind interactions, the stress response plays a pervasive role by affecting metabolic, hormonal, immune, and behavioral balance. In this perspective, we focus on chronic psychosocial stress and high levels of cortisol to highlight their role in MAFLD pathogenesis and worsening. From a PNEI perspective, considering the stress response as a therapeutic target in MAFLD allows for simultaneously influencing multiple pathways in the development of MAFLD, including dysmetabolism, inflammation, feeding behaviors, gut-liver axis, and dysbiosis, with the hope of better outcomes.
Collapse
|
7
|
Abdollahi M, Kato M, Lanting L, Tunduguru R, Wang M, Wang Y, Fueger PT, Wang Q, Huang W, Natarajan R. miR-379 mediates insulin resistance and obesity through impaired angiogenesis and adipogenesis regulated by ER stress. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 30:115-130. [PMID: 36250205 PMCID: PMC9535382 DOI: 10.1016/j.omtn.2022.09.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 09/15/2022] [Indexed: 01/29/2023]
Abstract
We investigated the role of microRNA (miR-379) in the pathogenesis of obesity, adipose tissue dysfunction, and insulin resistance (IR). We used miR-379 knockout (miR-379KO) mice to test whether loss of miR-379 affects high-fat diet (HFD)-induced obesity and IR via dysregulation of key miR-379 targets in adipose tissue. Increases in body weight, hyperinsulinemia, and IR in wild-type (WT)-HFD mice were significantly attenuated in miR-379KO-HFD mice with some sex differences. Relative to control chow-fed mice, in WT-HFD mice, expression of miR-379 and C/EBP homologous protein (Chop) (pro-endoplasmic reticulum [ER] stress) and inflammation in perigonadal white adipose tissue (gWAT) were increased, whereas adipogenic genes and miR-379 target genes (Vegfb and Edem3) were decreased. These changes, as well as key parameters of brown adipose tissue dysfunction (including mitochondrial defects), were significantly attenuated in miR-379KO-HFD mice. WAT from obese human subjects with and without type 2 diabetes showed increased miR-379 and decreased miR-379 target genes. In cultured 3T3L1 pre-adipocytes, miR-379 inhibitors increased miR-379 targets and adipogenic genes. These data suggest that miR-379 plays an important role in HFD-induced obesity through increased adipose inflammation, mitochondrial dysfunction, and ER stress as well as impaired adipogenesis and angiogenesis. miR-379 inhibitors may be developed as novel therapies for obesity and associated complications.
Collapse
Affiliation(s)
- Maryam Abdollahi
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Mitsuo Kato
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Linda Lanting
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Ragadeepthi Tunduguru
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Mei Wang
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Yangmeng Wang
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Patrick T. Fueger
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
- Comprehensive Metabolic Phenotyping Core, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Qiong Wang
- Department of Molecular and Cellular Endocrinology, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Wendong Huang
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Rama Natarajan
- Department of Diabetes Complications and Metabolism, Arthur Riggs Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
8
|
Dong Y, Yu C, Ma N, Xu X, Wu Q, Lu H, Gong L, Chen J, Ren J. MicroRNA-379-5p regulates free cholesterol accumulation and relieves diet induced-liver damage in db/db mice via STAT1/HMGCS1 axis. MOLECULAR BIOMEDICINE 2022; 3:25. [PMID: 35945406 PMCID: PMC9363541 DOI: 10.1186/s43556-022-00089-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 06/28/2022] [Indexed: 11/30/2022] Open
Abstract
Lipotoxicity induced by the overload of lipid in the liver, especially excess free cholesterol (FC), has been recognized as one of driving factors in the transition from non-alcoholic fatty liver (NAFL) to non-alcoholic steatohepatitis (NASH). MicroRNA (miR)-379-5p has been reported to play regulatory roles in hepatic triglyceride homeostasis, but the relationship of miR-379-5p and hepatic cholesterol homeostasis has never been touched. In the current study, we found that hepatic miR-379-5p levels were decreased obviously in NAFLD patients and model mice compared with their controls. Moreover, miR-379-5p was discovered to be able to inhibit intracellular FC accumulation and alleviate mitochondrial damage induced by palmitic acid (PA) in vitro. Furthermore, overexpression of miR-379-5p in HFHC-fed db/db mice could reduce the level of hepatic total cholesterol (TC) and FC, and ameliorate hepatic injury reflected by the lower serum alanine aminotransferase (ALT) and aspartate transaminase (AST). Subsequently, by combining spectrometry (MS) and luciferase assay, we identified miR-379-5p suppressed STAT1 through transcriptional and translational regulation. Finally, we confirmed that STAT1 was a transcriptional factor of HMGCS1. In conclusion, miR-379-5p inhibits STAT1 expression and regulates cholesterol metabolism through the STAT1/HMGCS1 axis, suggesting miR-379-5p might be applied to improve lipotoxicity in the future.
Collapse
|
9
|
Zhu L, Liao R, Huang J, Yan H, Xiao C, Yang Y, Wang H, Yang C. The miR-216/miR-217 Cluster Regulates Lipid Metabolism in Laying Hens With Fatty Liver Syndrome via PPAR/SREBP Signaling Pathway. Front Vet Sci 2022; 9:913841. [PMID: 35711801 PMCID: PMC9195098 DOI: 10.3389/fvets.2022.913841] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 04/27/2022] [Indexed: 12/14/2022] Open
Abstract
Fatty liver syndrome (FLS), a common metabolic disease in laying hens, caused by excessive hepatic fat deposition is a bottleneck in the poultry industry. However, no specific therapeutic methods have been developed. Evidence suggests that microRNAs (miRNAs) are essential for liver lipid metabolism and homeostasis, providing strong evidence for targeting miRNAs as a potential treatment option for liver diseases. However, the roles of miRNAs in the pathogenesis of FLS remain unclear. In present study, RNA-sequencing was performed to discern the expression patterns of miRNAs in normal and fatty livers of laying hens. In total, 12 dysregulated miRNAs (2 down-regulated and 10 up-regulated) were detected between the normal and fatty livers. Functional enrichment analysis showed the potential impacts of the dysregulated miRNAs on lipid metabolism. Notably, miR-216a/b and miR-217-5p, which belong to the miR-216/miR-217 cluster, were up-regulated in the sera and livers of FLS chickens, as well as free fatty acid (FFA)-induced LMH cells. Oil-red O staining revealed that up-regulation of the miR-216/miR-217 cluster induced lipid accumulation in FFA-induced LMH cells. Furthermore, the dual luciferase gene reporter assay and RT-qPCR analysis demonstrated that 3-hydroxyacyl-CoA dehydratase 2, F-box protein 8, and transmembrane 9 superfamily member 3 (TM9SF3) were directly targeted by miR-216a/b and miR-217-5p, respectively, and suppressed in the fatty livers of laying hens. Moreover, overexpression of the miR-216/miR-217 cluster or reduction in TM9SF3 levels led to activation of the proliferator-activated receptor/sterol regulatory-element binding protein (PPAR/SREBP) pathway. Overall, these results demonstrate that the miR-216/miR-217 cluster regulates lipid metabolism in laying hens with FLS, which should prove helpful in the development of new interventional strategies.
Collapse
Affiliation(s)
- Lihui Zhu
- Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai, China
- National Poultry Research Center for Engineering and Technology, Shanghai, China
| | - Rongrong Liao
- Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai, China
| | - Jiwen Huang
- College of Animal Science and Technology, Zhejiang Agriculture and Forestry University, Hangzhou, China
| | - Huaxiang Yan
- Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai, China
- National Poultry Research Center for Engineering and Technology, Shanghai, China
| | - Changfeng Xiao
- Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai, China
- National Poultry Research Center for Engineering and Technology, Shanghai, China
| | - Yunzhou Yang
- Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai, China
| | - Huiying Wang
- Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai, China
| | - Changsuo Yang
- Institute of Animal Husbandry and Veterinary Science, Shanghai Academy of Agricultural Sciences, Shanghai, China
- National Poultry Research Center for Engineering and Technology, Shanghai, China
| |
Collapse
|
10
|
Nuclear Receptors in Energy Metabolism. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1390:61-82. [DOI: 10.1007/978-3-031-11836-4_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
11
|
Cao C, Duan P, Li W, Guo Y, Zhang J, Gui Y, Yuan S. Lack of miR-379/miR-544 Cluster Resists High-Fat Diet-Induced Obesity and Prevents Hepatic Triglyceride Accumulation in Mice. Front Cell Dev Biol 2021; 9:720900. [PMID: 34527673 PMCID: PMC8435714 DOI: 10.3389/fcell.2021.720900] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 08/10/2021] [Indexed: 12/21/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) affects obesity-associated metabolic syndrome, which exhibits hepatic steatosis, insulin insensitivity and glucose intolerance. Emerging evidence suggests that microRNAs (miRNAs) are essential for the metabolic homeostasis of liver tissues. Many hepatic miRNAs located in the miR-379/miR-544 cluster were significantly increased in leptin-receptor-deficient type 2 mice (db/db), a mouse model of diabetes. However, the function of the miR-379/miR-544 cluster in the process of hepatic steatosis remains unclear. Here, we report that the novel function of miR-379/miR-544 cluster in regulating obesity-mediated metabolic dysfunction. Genetical mutation of miR-379/miR-544 cluster in mice displayed resistance to high-fat diet (HFD)-induced obesity with moderate hepatic steatosis and hypertriglyceridemia. In vitro studies revealed that silencing of miR-379 in human hepatocellular carcinoma (HepG2) cells ameliorated palmitic acid-induced elevation of cellular triglycerides, and overexpression of miR-379 had the opposite effect. Moreover, Igf1r (Insulin-like growth factor 1 receptor) and Dlk1 (Delta-like homolog 1) were directly targeted by miR-379 and miR-329, respectively, and elevated in the livers of the miR-379/miR-544 cluster knockout mice fed on HFD. Further transcriptome analyses revealed that the hepatic gene expressions are dysregulated in miR-379/miR-544 knockout mice fed with HFD. Collectively, our findings identify the miR-379/miR-544 cluster as integral components of a regulatory circuit that functions under conditions of metabolic stress to control hepatic steatosis. Thus, this miRNA cluster provides potential targets for pharmacologic intervention in obesity and NAFLD.
Collapse
Affiliation(s)
- Congcong Cao
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen-Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Peng Duan
- Department of Obstetrics and Gynaecology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, China
| | - Wencun Li
- The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yang Guo
- College of Pharmacy, Hubei University of Medicine, Shiyan, China
| | - Jin Zhang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yaoting Gui
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen-Peking University - The Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Shuiqiao Yuan
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Shenzhen Huazhong University of Science and Technology Research Institute, Shenzhen, China
| |
Collapse
|
12
|
Corrales WA, Silva JP, Parra CS, Olave FA, Aguayo FI, Román-Albasini L, Aliaga E, Venegas-Zamora L, Avalos AM, Rojas PS, Maracaja-Coutinho V, Oakley RH, Cidlowski JA, Fiedler JL. Sex-Dependent Changes of miRNA Levels in the Hippocampus of Adrenalectomized Rats Following Acute Corticosterone Administration. ACS Chem Neurosci 2021; 12:2981-3001. [PMID: 34339164 DOI: 10.1021/acschemneuro.0c00762] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
We explored sex-biased effects of the primary stress glucocorticoid hormone corticosterone on the miRNA expression profile in the rat hippocampus. Adult adrenalectomized (ADX) female and male rats received a single corticosterone (10 mg/kg) or vehicle injection, and after 6 h, hippocampi were collected for miRNA, mRNA, and Western blot analyses. miRNA profiling microarrays showed a basal sex-biased miRNA profile in ADX rat hippocampi. Additionally, acute corticosterone administration triggered a sex-biased differential expression of miRNAs derived from genes located in several chromosomes and clusters on the X and 6 chromosomes. Putative promoter analysis unveiled that most corticosterone-responsive miRNA genes contained motifs for either direct or indirect glucocorticoid actions in both sexes. The evaluation of transcription factors indicated that almost 50% of miRNA genes sensitive to corticosterone in both sexes was under glucocorticoid receptor regulation. Transcription factor-miRNA regulatory network analyses identified several transcription factors that regulate, activate, or repress miRNA expression. Validated target mRNA analysis of corticosterone-responsive miRNAs showed a more complex miRNA-mRNA interaction network in males compared to females. Enrichment analysis revealed that several hippocampal-relevant pathways were affected in both sexes, such as neurogenesis and neurotrophin signaling. The evaluation of selected miRNA targets from these pathways displayed a strong sex difference in the hippocampus of ADX-vehicle rats. Corticosterone treatment did not change the levels of the miRNA targets and their corresponding tested proteins. Our data indicate that corticosterone exerts a sex-biased effect on hippocampal miRNA expression, which may engage in sculpting the basal sex differences observed at higher levels of hippocampal functioning.
Collapse
Affiliation(s)
- Wladimir A. Corrales
- Laboratory of Neuroplasticity and Neurogenetics, Faculty of Chemical and Pharmaceutical Sciences, Department of Biochemistry and Molecular Biology, Universidad de Chile, Independencia, Santiago 8380492, Chile
| | - Juan P. Silva
- Laboratory of Neuroplasticity and Neurogenetics, Faculty of Chemical and Pharmaceutical Sciences, Department of Biochemistry and Molecular Biology, Universidad de Chile, Independencia, Santiago 8380492, Chile
| | - Claudio S. Parra
- Laboratory of Neuroplasticity and Neurogenetics, Faculty of Chemical and Pharmaceutical Sciences, Department of Biochemistry and Molecular Biology, Universidad de Chile, Independencia, Santiago 8380492, Chile
| | - Felipe A. Olave
- Laboratory of Neuroplasticity and Neurogenetics, Faculty of Chemical and Pharmaceutical Sciences, Department of Biochemistry and Molecular Biology, Universidad de Chile, Independencia, Santiago 8380492, Chile
| | - Felipe I. Aguayo
- Laboratory of Neuroplasticity and Neurogenetics, Faculty of Chemical and Pharmaceutical Sciences, Department of Biochemistry and Molecular Biology, Universidad de Chile, Independencia, Santiago 8380492, Chile
| | - Luciano Román-Albasini
- Laboratory of Neuroplasticity and Neurogenetics, Faculty of Chemical and Pharmaceutical Sciences, Department of Biochemistry and Molecular Biology, Universidad de Chile, Independencia, Santiago 8380492, Chile
| | - Esteban Aliaga
- Department of Kinesiology and The Neuropsychology and Cognitive Neurosciences Research Center (CINPSI-Neurocog), Faculty of Health Sciences, Universidad Católica del Maule, Talca 3460000, Chile
| | - Leslye Venegas-Zamora
- Laboratory of Neuroplasticity and Neurogenetics, Faculty of Chemical and Pharmaceutical Sciences, Department of Biochemistry and Molecular Biology, Universidad de Chile, Independencia, Santiago 8380492, Chile
| | - Ana M. Avalos
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago 8910060, Chile
| | - Paulina S. Rojas
- Escuela de Química y Farmacia, Facultad de Medicina, Universidad Andres Bello, Santiago 8370149, Chile
| | - Vinicius Maracaja-Coutinho
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, Department of Biochemistry and Molecular Biology, Universidad de Chile, Independencia, Santiago 8380492, Chile
| | - Robert H. Oakley
- National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709, United States
| | - John A. Cidlowski
- National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709, United States
| | - Jenny L. Fiedler
- Laboratory of Neuroplasticity and Neurogenetics, Faculty of Chemical and Pharmaceutical Sciences, Department of Biochemistry and Molecular Biology, Universidad de Chile, Independencia, Santiago 8380492, Chile
| |
Collapse
|
13
|
Ghafouri-Fard S, Shaterabadi D, Abak A, Shoorei H, Bahroudi Z, Taheri M, Mousavinejad SA. An update on the role of miR-379 in human disorders. Biomed Pharmacother 2021; 139:111553. [PMID: 33845370 DOI: 10.1016/j.biopha.2021.111553] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 03/23/2021] [Accepted: 03/27/2021] [Indexed: 12/24/2022] Open
Abstract
miR-379 is a miRNA transcribed from the MIR379 locus on 14q32.31. This miRNA is located in an evolutionarily conserved miRNA cluster in an imprinted region that contains DLK1 and DIO3 genes. The mouse homolog of this miRNA has been shown to be under-expressed in response to glucocorticoid receptor deficiency. Moreover, miR-379 has a tumor-suppressive role in a wide variety of tissues including the brain, breast, lung, and liver. In addition to restraining cell proliferation and migration, miR-379 can suppress the epithelial-mesenchymal transition process. Abnormal expression of this miRNA implies the pathogenesis of Duchene muscular dystrophy, spinal cord injury, diabetic nephropathy, acute myocardial infarction, and premature ovarian failure. This review aims to the summarization of the role of miR-379 in neoplastic and non-neoplastic conditions.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Donya Shaterabadi
- Department of Medical Nanotechnology, Faculty of Advanced Medical Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Afete Abak
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Shoorei
- Department of Anatomical Sciences, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Bahroudi
- Department of Anatomical Sciences, Faculty of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohammad Taheri
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Seyed Ali Mousavinejad
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
14
|
Wu J, Nagy LE, Liangpunsakul S, Wang L. Non-coding RNA crosstalk with nuclear receptors in liver disease. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166083. [PMID: 33497819 PMCID: PMC7987766 DOI: 10.1016/j.bbadis.2021.166083] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 12/28/2020] [Accepted: 01/16/2021] [Indexed: 02/06/2023]
Abstract
The dysregulation of nuclear receptors (NRs) underlies the pathogenesis of a variety of liver disorders. Non-coding RNAs (ncRNAs) are defined as RNA molecules transcribed from DNA but not translated into proteins. MicroRNAs (miRNAs) and long non-coding RNAs (lncRNAs) are two types of ncRNAs that have been extensively studied for regulating gene expression during diverse cellular processes. NRs as therapeutic targets in liver disease have been exemplified by the successful application of their pharmacological ligands in clinics. MiRNA-based reagents or drugs are emerging as flagship products in clinical trials. Advancing our understanding of the crosstalk between NRs and ncRNAs is critical to the development of diagnostic and therapeutic strategies. This review summarizes recent findings on the reciprocal regulation between NRs and ncRNAs (mainly on miRNAs and lncRNAs) and their implication in liver pathophysiology, which might be informative to the translational medicine of targeting NRs and ncRNAs in liver disease.
Collapse
Affiliation(s)
- Jianguo Wu
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States of America; Department of Molecular Medicine, Case Western Reserve University, Cleveland, OH, United States of America.
| | - Laura E Nagy
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States of America; Department of Gastroenterology and Hepatology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States of America; Department of Molecular Medicine, Case Western Reserve University, Cleveland, OH, United States of America
| | - Suthat Liangpunsakul
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States of America; Roudebush Veterans Administration Medical Center, Indianapolis, IN, United States of America; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Li Wang
- Department of Internal Medicine, Section of Digestive Diseases, Yale University, New Haven, CT, United States of America
| |
Collapse
|
15
|
Ghafouri-Fard S, Taheri M. The expression profile and role of non-coding RNAs in obesity. Eur J Pharmacol 2020; 892:173809. [PMID: 33345852 DOI: 10.1016/j.ejphar.2020.173809] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/07/2020] [Accepted: 12/11/2020] [Indexed: 02/06/2023]
Abstract
Latest years have experienced a dramatic upsurge in the knowledge about the function of non-coding transcripts in the determination of diverse human phenotypes including obesity. Several miRNAs and lncRNAs participate in the regulation of metabolic pathways leading to obesity. Several lncRNAs such as Mist, lincIRS2, lncRNA-p5549, H19, GAS5 and SNHG9 have been shown to be down-regulated in adipose tissues or other biological samples in the obese human or animal subjects. On the other hand, Meg3, Plnc1, Blnc1, AC092834.1, TINCR and PVT1 are among up-regulated lncRNAs in the obese subjects. Tens of miRNAs have differential expression between obese and non-obese subjects or between mature adipocytes and pre-adipocytes. Understanding the molecular mechanism of involvement of non-coding RNAs in the pathobiology of obesity would simplify design of therapeutic choices for protecting against obesity and its related comorbidities. We explain the available literature on the function of these transcripts in the pathobiology of obesity.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
16
|
Differential expression profile of microRNA in yak skeletal muscle and adipose tissue during development. Genes Genomics 2020; 42:1347-1359. [PMID: 32996042 DOI: 10.1007/s13258-020-00988-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 08/13/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND miRNAs play an important role in regulating normal animal development. Muscle tissue and fat metabolism are important for maintaining energy balance in animals. Yak has important agricultural and economic importance as it provides milk, meat, and hair. It is used for transportation as well. However, the miRNA expression profiles of their muscle and adipose tissue are currently unknown. OBJECTIVE To explore the regulatory roles of miRNAs in the skeletal muscle and adipose tissues of yak. METHODS A total of 12 small RNA libraries were constructed from the skeletal muscle and adipose samples from yak aged 0.5, 2.5, 4.5, and 7.5 years. High-throughput sequencing and bioinformatics analysis were used to determine the dynamic expression profile of miRNA, and a miRNA regulatory network related to muscle and adipose tissue development was established. RESULTS miR-1-3p and miR-143-3p showed the highest expression during yak skeletal muscle and fat development, respectively. The MAPK and Ras signaling pathways were the pivotal pathways. miR-181-5p, miR-542-3p, and miR-424-5p may have key roles in skeletal muscle development, and CREBRF, GRB10, CDK1, RFX3, and EPC2 were the core target genes. While miR-127-5p, miR-379-3p, and miR-494-3p may play important regulatory roles in adipose deposition, and ETV1, XPO7, and C5AR2 were the core target genes. CONCLUSION This study provides valuable resources for further study of the molecular mechanisms underlying yak skeletal muscle and adipose tissue development, and also a basis for studying the interactions between genes and miRNAs.
Collapse
|
17
|
de Guia RM. Stress, glucocorticoid signaling pathway, and metabolic disorders. Diabetes Metab Syndr 2020; 14:1273-1280. [PMID: 32755820 DOI: 10.1016/j.dsx.2020.06.038] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 06/15/2020] [Accepted: 06/16/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND AIMS Glucocorticoids and the GR serve as an essential molecular mediator of stress and different physiologic processes. This review summarizes main findings from studies on the role of the GC/GR signaling in the modulation of genes for nutrient processing by the different organs involved in metabolic diseases. METHODS Descriptive review of relevant papers known to the author was conducted. RESULTS Several high-throughput screenings in the past 15 years have identified potential GR DNA-binding regions in different cell types with genes that are annotated to be important for the control of metabolism. Transcriptional regulation of these GC-responsive genes provides links between the hypothalamic-pituitary-adrenal axis (HPA) and systemic energy homeostasis in both physiological and pathophysiological states. Future studies must reconsider the use of agonist, the utilization of animal models of stress and metabolic disorders, and validation in humans. CONCLUSION This review recapitulates the significant role of the GC/GR signaling in molecular metabolic control and metabolic disorders. Potential future research focus and optimizations have also been identified.
Collapse
Affiliation(s)
- Roldan M de Guia
- Joint Division Molecular Metabolic Control, DKFZ-ZMBH Alliance and Network Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany; Keio Global Research Institute (KGRI) and Department of Pharmacology, Keio University School of Medicine, Tokyo, Japan; Czech Centre for Phenogenomics (CCP), Institute of Molecular Genetics of the Czech Academy of Sciences, Vestec, Czech Republic.
| |
Collapse
|
18
|
Sanson M, Vu Hong A, Massourides E, Bourg N, Suel L, Amor F, Corre G, Bénit P, Barthélémy I, Blot S, Bigot A, Pinset C, Rustin P, Servais L, Voit T, Richard I, Israeli D. miR-379 links glucocorticoid treatment with mitochondrial response in Duchenne muscular dystrophy. Sci Rep 2020; 10:9139. [PMID: 32499563 PMCID: PMC7272451 DOI: 10.1038/s41598-020-66016-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 05/11/2020] [Indexed: 12/12/2022] Open
Abstract
Duchenne Muscular Dystrophy (DMD) is a lethal muscle disorder, caused by mutations in the DMD gene and affects approximately 1:5000-6000 male births. In this report, we identified dysregulation of members of the Dlk1-Dio3 miRNA cluster in muscle biopsies of the GRMD dog model. Of these, we selected miR-379 for a detailed investigation because its expression is high in the muscle, and is known to be responsive to glucocorticoid, a class of anti-inflammatory drugs commonly used in DMD patients. Bioinformatics analysis predicts that miR-379 targets EIF4G2, a translational factor, which is involved in the control of mitochondrial metabolic maturation. We confirmed in myoblasts that EIF4G2 is a direct target of miR-379, and identified the DAPIT mitochondrial protein as a translational target of EIF4G2. Knocking down DAPIT in skeletal myotubes resulted in reduced ATP synthesis and myogenic differentiation. We also demonstrated that this pathway is GC-responsive since treating mice with dexamethasone resulted in reduced muscle expression of miR-379 and increased expression of EIF4G2 and DAPIT. Furthermore, miR-379 seric level, which is also elevated in the plasma of DMD patients in comparison with age-matched controls, is reduced by GC treatment. Thus, this newly identified pathway may link GC treatment to a mitochondrial response in DMD.
Collapse
Affiliation(s)
- Mathilde Sanson
- Généthon INSERM, UMR_S951, INTEGRARE research unit, Evry, 91000, France
| | - Ai Vu Hong
- Généthon INSERM, UMR_S951, INTEGRARE research unit, Evry, 91000, France
| | | | - Nathalie Bourg
- Généthon INSERM, UMR_S951, INTEGRARE research unit, Evry, 91000, France
| | - Laurence Suel
- Généthon INSERM, UMR_S951, INTEGRARE research unit, Evry, 91000, France
| | - Fatima Amor
- Généthon INSERM, UMR_S951, INTEGRARE research unit, Evry, 91000, France
| | - Guillaume Corre
- Généthon INSERM, UMR_S951, INTEGRARE research unit, Evry, 91000, France
| | - Paule Bénit
- INSERM, UMR S1141, Hôpital Robert Debré, Paris, France
| | - Inès Barthélémy
- Inserm U955-E10, IMRB, Université Paris Est, Ecole nationale vétérinaire d'Alfort, 94700, Maisons-Alfort, France
| | - Stephane Blot
- Inserm U955-E10, IMRB, Université Paris Est, Ecole nationale vétérinaire d'Alfort, 94700, Maisons-Alfort, France
| | - Anne Bigot
- Center for Research in Myology UMRS974, Sorbonne Université, INSERM, Myology Institute, Paris, France
| | | | - Pierre Rustin
- INSERM, UMR S1141, Hôpital Robert Debré, Paris, France
| | - Laurent Servais
- MDUK Oxford Neuromuscular Centre, Department of Paediatrics, University of Oxford, Oxford, UK
- Division of Child Neurology, Centre de Références des Maladies Neuromusculaires, Department of Pediatrics, University Hospital Liège & University of Liège, Liège, Belgium
| | - Thomas Voit
- NIHR Great Ormond Street Hospital Biomedical Research Centre and Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Isabelle Richard
- Généthon INSERM, UMR_S951, INTEGRARE research unit, Evry, 91000, France
| | - David Israeli
- Généthon INSERM, UMR_S951, INTEGRARE research unit, Evry, 91000, France.
| |
Collapse
|
19
|
Okamoto K, Koda M, Okamoto T, Onoyama T, Miyoshi K, Kishina M, Matono T, Kato J, Tokunaga S, Sugihara T, Hiramatsu A, Hyogo H, Tobita H, Sato S, Kawanaka M, Hara Y, Hino K, Chayama K, Murawaki Y, Isomoto H. Serum miR-379 expression is related to the development and progression of hypercholesterolemia in non-alcoholic fatty liver disease. PLoS One 2020; 15:e0219412. [PMID: 32106257 PMCID: PMC7046274 DOI: 10.1371/journal.pone.0219412] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 02/10/2020] [Indexed: 12/15/2022] Open
Abstract
Introduction Non-alcoholic fatty liver disease (NAFLD) has a wide spectrum, eventually leading to cirrhosis and hepatic carcinogenesis. We previously reported that a series of microRNAs (miRNAs) mapped in the 14q32.2 maternally imprinted gene region (Dlk1-Dio3 mat) are related to NAFLD development and progression in a mouse model. We examined the suitability of miR-379, a circulating Dlk1-Dio3 mat miRNA, as a human NAFLD biomarker. Methods Eighty NAFLD patients were recruited for this study. miR-379 was selected from the putative Dlk1-Dio3 mat miRNA cluster because it exhibited the greatest expression difference between NAFLD and non-alcoholic steatohepatitis in our preliminary study. Real-time PCR was used to examine the expression levels of miR-379 and miR-16 as an internal control. One patient was excluded due to low RT-PCR signal. Results Compared to normal controls, serum miR-379 expression was significantly up-regulated in NAFLD patients. Receiver operating characteristic curve analysis suggested that miR-379 is a suitable marker for discriminating NAFLD patients from controls, with an area under the curve value of 0.72. Serum miR-379 exhibited positive correlations with alkaline phosphatase, total cholesterol, low-density-lipoprotein cholesterol and non-high-density-lipoprotein cholesterol levels in patients with early stage NAFLD (Brunt fibrosis stage 0 to 1). The correlation between serum miR-379 and cholesterol levels was lost in early stage NAFLD patients treated with statins. Software-based predictions indicated that various energy metabolism–related genes, including insulin-like growth factor-1 (IGF-1) and IGF-1 receptor, are potential targets of miR-379. Conclusions Serum miR-379 exhibits high potential as a biomarker for NAFLD. miR-379 appears to increase cholesterol lipotoxicity, leading to the development and progression of NAFLD, via interference with the expression of target genes, including those related to the IGF-1 signaling pathway. Our results could facilitate future research into the pathogenesis, diagnosis, and treatment of NAFLD.
Collapse
Affiliation(s)
- Kinya Okamoto
- Second Department of Internal Medicine, Tottori University School of Medicine, Yonago, Tottori, Japan
- * E-mail:
| | - Masahiko Koda
- Second Department of Internal Medicine, Tottori University School of Medicine, Yonago, Tottori, Japan
| | - Toshiaki Okamoto
- Second Department of Internal Medicine, Tottori University School of Medicine, Yonago, Tottori, Japan
| | - Takumi Onoyama
- Second Department of Internal Medicine, Tottori University School of Medicine, Yonago, Tottori, Japan
| | - Kenichi Miyoshi
- Second Department of Internal Medicine, Tottori University School of Medicine, Yonago, Tottori, Japan
| | - Manabu Kishina
- Second Department of Internal Medicine, Tottori University School of Medicine, Yonago, Tottori, Japan
| | - Tomomitsu Matono
- Second Department of Internal Medicine, Tottori University School of Medicine, Yonago, Tottori, Japan
| | - Jun Kato
- Second Department of Internal Medicine, Tottori University School of Medicine, Yonago, Tottori, Japan
| | - Shiho Tokunaga
- Second Department of Internal Medicine, Tottori University School of Medicine, Yonago, Tottori, Japan
| | - Takaaki Sugihara
- Second Department of Internal Medicine, Tottori University School of Medicine, Yonago, Tottori, Japan
| | - Akira Hiramatsu
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima, Japan
| | - Hideyuki Hyogo
- Department of Gastroenterology and Hepatology, JA Hiroshima General Hospital, Hatsukaichi, Hiroshima, Japan
| | - Hiroshi Tobita
- Department of Gastroenterology and Hepatology, Shimane University School of Medicine, Izumo, Shimane, Japan
| | - Shuichi Sato
- Department of Gastroenterology and Hepatology, Shimane University School of Medicine, Izumo, Shimane, Japan
| | - Miwa Kawanaka
- Department of General Internal Medicine 2, General Medical Center, Kawasaki Medical School, Okayama, Okayama, Japan
| | - Yuichi Hara
- Department of Hepatology and Pancreatology, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Keisuke Hino
- Department of Hepatology and Pancreatology, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Kazuaki Chayama
- Department of Gastroenterology and Metabolism, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Hiroshima, Japan
| | - Yoshikazu Murawaki
- Second Department of Internal Medicine, Tottori University School of Medicine, Yonago, Tottori, Japan
| | - Hajime Isomoto
- Second Department of Internal Medicine, Tottori University School of Medicine, Yonago, Tottori, Japan
| |
Collapse
|
20
|
Glantschnig C, Mattijssen F, Vogl ES, Ali Khan A, Rios Garcia M, Fischer K, Müller T, Uhlenhaut H, Nawroth P, Scheideler M, Rose AJ, Pellegata N, Herzig S. The glucocorticoid receptor in brown adipocytes is dispensable for control of energy homeostasis. EMBO Rep 2019; 20:e48552. [PMID: 31559673 PMCID: PMC6832000 DOI: 10.15252/embr.201948552] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 09/02/2019] [Accepted: 09/09/2019] [Indexed: 12/14/2022] Open
Abstract
Aberrant activity of the glucocorticoid (GC)/glucocorticoid receptor (GR) endocrine system has been linked to obesity-related metabolic dysfunction. Traditionally, the GC/GR axis has been believed to play a crucial role in adipose tissue formation and function in both, white (WAT) and brown adipose tissue (BAT). While recent studies have challenged this notion for WAT, the contribution of GC/GR signaling to BAT-dependent energy homeostasis remained unknown. Here, we have generated and characterized a BAT-specific GR-knockout mouse (GRBATKO ), for the first time allowing to genetically interrogate the metabolic impact of BAT-GR. The HPA axis in GRBATKO mice was intact, as was the ability of mice to adapt to cold. BAT-GR was dispensable for the adaptation to fasting-feeding cycles and the development of diet-induced obesity. In obesity, glucose and lipid metabolism, insulin sensitivity, and food intake remained unchanged, aligning with the absence of changes in thermogenic gene expression. Together, we demonstrate that the GR in UCP1-positive BAT adipocytes plays a negligible role in systemic metabolism and BAT function, thereby opposing a long-standing paradigm in the field.
Collapse
|
21
|
Liu F, Chen Q, Chen F, Wang J, Gong R, He B. The lncRNA ENST00000608794 acts as a competing endogenous RNA to regulate PDK4 expression by sponging miR-15b-5p in dexamethasone induced steatosis. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:1449-1457. [PMID: 31330194 DOI: 10.1016/j.bbalip.2019.07.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 06/30/2019] [Accepted: 07/09/2019] [Indexed: 12/31/2022]
Abstract
The contribution of ncRNAs, especially long non-coding RNAs (lncRNAs) to drug induced steatosis remains largely unknown. The aim of this study was to investigate the role of lncRNA ENST00000608794 in dexamethasone induced steatosis. We found that ENST00000608794 is expressed at higher levels in dexamethasone treated HepG2 cell, and ENST00000608794 can bind and be regulated by miR-15b-5p. Ectopic expression of ENST00000608794 enhanced steatosis and the protein expression of PDK4 which is a critical gene in lipid metabolism and also is a target of miR-15b-5p. However, the differentiated PDK4 expression between control and ectopic expression of ENST00000608794 was absence in the presence of miR-15b-5p inhibitor. Moreover, in dexamethasone treated HepG2 cell lines, ENST00000608794 increased whether with miR-15b-5p inhibitor treatment or not, while increase of PDK4 expression by dexamethasone was greatly compromised in the presence of miR-15b-5p mimic. Meanwhile, dexamethasone induced steatosis could be ameliorated by silencing ENST00000608794 or expressing miR-15b-5p. Taken together, the results suggested that ENST00000608794 plays an important role in dexamethasone induced steatosis, which was partly mediated by derepressing of PDK4 through competitively binding to miR-15b-5p.
Collapse
Affiliation(s)
- Fengqiong Liu
- Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fujian, China
| | - Qing Chen
- Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fujian, China
| | - Fa Chen
- Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fujian, China
| | - Jing Wang
- Laboratory Center, The Major Subject of Environment and Health of Fujian Key Universities, School of Public Health, Fujian Medical University, Fujian, China
| | - Ruijie Gong
- Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fujian, China
| | - Baochang He
- Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fujian, China.
| |
Collapse
|
22
|
Challa TD, Wueest S, Lucchini FC, Dedual M, Modica S, Borsigova M, Wolfrum C, Blüher M, Konrad D. Liver ASK1 protects from non-alcoholic fatty liver disease and fibrosis. EMBO Mol Med 2019; 11:e10124. [PMID: 31595673 PMCID: PMC6783644 DOI: 10.15252/emmm.201810124] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 04/30/2019] [Accepted: 05/03/2019] [Indexed: 12/15/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is strongly associated with obesity and may progress to non-alcoholic steatohepatitis (NASH) and liver fibrosis. The deficit of pharmacological therapies for the latter mainly results from an incomplete understanding of involved pathological mechanisms. Herein, we identify apoptosis signal-regulating kinase 1 (ASK1) as a suppressor of NASH and fibrosis formation. High-fat diet-fed and aged chow-fed liver-specific ASK1-knockout mice develop a higher degree of hepatic steatosis, inflammation, and fibrosis compared to controls. In addition, pharmacological inhibition of ASK1 increased hepatic lipid accumulation in wild-type mice. In line, liver-specific ASK1 overexpression protected mice from the development of high-fat diet-induced hepatic steatosis and carbon tetrachloride-induced fibrosis. Mechanistically, ASK1 depletion blunts autophagy, thereby enhancing lipid droplet accumulation and liver fibrosis. In human livers of lean and obese subjects, ASK1 expression correlated negatively with liver fat content and NASH scores, but positively with markers for autophagy. Taken together, ASK1 may be a novel therapeutic target to tackle NAFLD and liver fibrosis.
Collapse
Affiliation(s)
- Tenagne D Challa
- Division of Pediatric Endocrinology and DiabetologyUniversity Children's HospitalZurichSwitzerland
- Children's Research CenterUniversity Children's HospitalZurichSwitzerland
| | - Stephan Wueest
- Division of Pediatric Endocrinology and DiabetologyUniversity Children's HospitalZurichSwitzerland
- Children's Research CenterUniversity Children's HospitalZurichSwitzerland
| | - Fabrizio C Lucchini
- Division of Pediatric Endocrinology and DiabetologyUniversity Children's HospitalZurichSwitzerland
- Children's Research CenterUniversity Children's HospitalZurichSwitzerland
- Zurich Center for Integrative Human PhysiologyUniversity of ZurichZurichSwitzerland
| | - Mara Dedual
- Division of Pediatric Endocrinology and DiabetologyUniversity Children's HospitalZurichSwitzerland
- Children's Research CenterUniversity Children's HospitalZurichSwitzerland
- Zurich Center for Integrative Human PhysiologyUniversity of ZurichZurichSwitzerland
| | - Salvatore Modica
- Institute of Food, Nutrition and HealthETH ZurichSchwerzenbachSwitzerland
| | - Marcela Borsigova
- Division of Pediatric Endocrinology and DiabetologyUniversity Children's HospitalZurichSwitzerland
- Children's Research CenterUniversity Children's HospitalZurichSwitzerland
| | - Christian Wolfrum
- Institute of Food, Nutrition and HealthETH ZurichSchwerzenbachSwitzerland
| | | | - Daniel Konrad
- Division of Pediatric Endocrinology and DiabetologyUniversity Children's HospitalZurichSwitzerland
- Children's Research CenterUniversity Children's HospitalZurichSwitzerland
- Zurich Center for Integrative Human PhysiologyUniversity of ZurichZurichSwitzerland
| |
Collapse
|
23
|
Choi H, Koh HWL, Zhou L, Cheng H, Loh TP, Parvaresh Rizi E, Toh SA, Ronnett GV, Huang BE, Khoo CM. Plasma Protein and MicroRNA Biomarkers of Insulin Resistance: A Network-Based Integrative -Omics Analysis. Front Physiol 2019; 10:379. [PMID: 31024340 PMCID: PMC6460474 DOI: 10.3389/fphys.2019.00379] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 03/19/2019] [Indexed: 12/20/2022] Open
Abstract
Although insulin resistance (IR) is a key pathophysiologic condition underlying various metabolic disorders, impaired cellular glucose uptake is one of many manifestations of metabolic derangements in the human body. To study the systems-wide molecular changes associated with obesity-dependent IR, we integrated information on plasma proteins and microRNAs in eight obese insulin-resistant (OIR, HOMA-IR > 2.5) and nine lean insulin-sensitive (LIS, HOMA-IR < 1.0) normoglycemic males. Of 374 circulating miRNAs we profiled, 65 species increased and 73 species decreased in the OIR compared to the LIS subjects, suggesting that the overall balance of the miRNA secretome is shifted in the OIR subjects. We also observed that 40 plasma proteins increased and 4 plasma proteins decreased in the OIR subjects compared to the LIS subjects, and most proteins are involved in metabolic and endocytic functions. We used an integrative -omics analysis framework called iOmicsPASS to link differentially regulated miRNAs with their target genes on the TargetScan map and the human protein interactome. Combined with tissue of origin information, the integrative analysis allowed us to nominate obesity-dependent and obesity-independent protein markers, along with potential sites of post-transcriptional regulation by some of the miRNAs. We also observed the changes in each -omics platform that are not linked by the TargetScan map, suggesting that proteins and microRNAs provide orthogonal information for the progression of OIR. In summary, our integrative analysis provides a network of elevated plasma markers of OIR and a global shift of microRNA secretome composition in the blood plasma.
Collapse
Affiliation(s)
- Hyungwon Choi
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore.,Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research, Singapore, Singapore
| | - Hiromi W L Koh
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| | | | - He Cheng
- MiRXES, Pte. Ltd., Singapore, Singapore
| | - Tze Ping Loh
- Department of Laboratory Medicine, National University Hospital, Singapore, Singapore
| | - Ehsan Parvaresh Rizi
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Sue Anne Toh
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Gabriele V Ronnett
- Janssen Research & Development US, World Without Disease Accelerator, Spring House, NJ, United States
| | - Bevan E Huang
- Janssen Research & Development US, South San Francisco, CA, United States
| | - Chin Meng Khoo
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
24
|
Mousovich‐Neto F, Matos MS, Costa ACR, Melo Reis RA, Atella GC, Miranda‐Alves L, Carvalho DP, Ketzer LA, Corrêa da Costa VM. Brown adipose tissue remodelling induced by corticosterone in male Wistar rats. Exp Physiol 2019; 104:514-528. [DOI: 10.1113/ep087332] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 01/15/2019] [Indexed: 12/16/2022]
Affiliation(s)
- Felippe Mousovich‐Neto
- Laboratório de Fisiologia Endócrina Doris Rosenthal Universidade Federal do Rio de Janeiro Rio de Janeiro Brasil
| | - Marina Souza Matos
- Laboratório de Fisiologia Endócrina Doris Rosenthal Universidade Federal do Rio de Janeiro Rio de Janeiro Brasil
| | - Anna Carolina Rego Costa
- Laboratório de Neuroquímica Instituto de Biofísica Carlos Chagas Filho Universidade Federal do Rio de Janeiro Rio de Janeiro Brasil
| | - Ricardo Augusto Melo Reis
- Laboratório de Neuroquímica Instituto de Biofísica Carlos Chagas Filho Universidade Federal do Rio de Janeiro Rio de Janeiro Brasil
| | - Georgia Correa Atella
- Laboratório de Bioquímica de Lipídeos e Lipoproteínas Instituto de Bioquímica Médica Leopoldo de Meis Universidade Federal do Rio de Janeiro Rio de Janeiro Brasil
| | - Leandro Miranda‐Alves
- Instituto de Ciências Biomédicas Universidade Federal do Rio de Janeiro Rio de Janeiro Brasil
| | - Denise P. Carvalho
- Laboratório de Fisiologia Endócrina Doris Rosenthal Universidade Federal do Rio de Janeiro Rio de Janeiro Brasil
| | - Luisa Andrea Ketzer
- Núcleo Multidisciplinar de Pesquisa UFRJ‐Xerém em Biologia Campus Duque de Caxias Universidade Federal do Rio de Janeiro Rio de Janeiro Brasil
| | - Vânia Maria Corrêa da Costa
- Laboratório de Fisiologia Endócrina Doris Rosenthal Universidade Federal do Rio de Janeiro Rio de Janeiro Brasil
| |
Collapse
|
25
|
Glantschnig C, Koenen M, Gil‐Lozano M, Karbiener M, Pickrahn I, Williams‐Dautovich J, Patel R, Cummins CL, Giroud M, Hartleben G, Vogl E, Blüher M, Tuckermann J, Uhlenhaut H, Herzig S, Scheideler M. A miR‐29a‐driven negative feedback loop regulates peripheral glucocorticoid receptor signaling. FASEB J 2019; 33:5924-5941. [DOI: 10.1096/fj.201801385rr] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Christina Glantschnig
- Institute for Diabetes and Cancer (IDC)Helmholtz Center Munich Neuherberg Germany
- Joint Heidelberg‐IDC, Inner Medicine 1Heidelberg University Hospital Heidelberg Germany
- German Center for Diabetes Research (DZD) Neuherberg Germany
| | - Mascha Koenen
- Institute of Comparative Molecular EndocrinologyUlm University Ulm Germany
| | - Manuel Gil‐Lozano
- Institute for Diabetes and Cancer (IDC)Helmholtz Center Munich Neuherberg Germany
- Joint Heidelberg‐IDC, Inner Medicine 1Heidelberg University Hospital Heidelberg Germany
- German Center for Diabetes Research (DZD) Neuherberg Germany
| | - Michael Karbiener
- Division of Phoniatrics, Speech, and SwallowingDepartment of OtorhinolaryngologyUniversity HospitalMedical University of Graz Graz Austria
| | - Ines Pickrahn
- Department of Legal MedicineUniversity of Salzburg Salzburg Austria
| | | | - Rucha Patel
- Department of Pharmaceutical SciencesUniversity of Toronto Toronto Ontario Canada
| | - Carolyn L. Cummins
- Department of Pharmaceutical SciencesUniversity of Toronto Toronto Ontario Canada
| | - Maude Giroud
- Institute for Diabetes and Cancer (IDC)Helmholtz Center Munich Neuherberg Germany
- Joint Heidelberg‐IDC, Inner Medicine 1Heidelberg University Hospital Heidelberg Germany
- German Center for Diabetes Research (DZD) Neuherberg Germany
| | - Götz Hartleben
- Institute for Diabetes and Cancer (IDC)Helmholtz Center Munich Neuherberg Germany
- Joint Heidelberg‐IDC, Inner Medicine 1Heidelberg University Hospital Heidelberg Germany
- German Center for Diabetes Research (DZD) Neuherberg Germany
| | - Elena Vogl
- Institute for Diabetes and Cancer (IDC)Helmholtz Center Munich Neuherberg Germany
- Joint Heidelberg‐IDC, Inner Medicine 1Heidelberg University Hospital Heidelberg Germany
- German Center for Diabetes Research (DZD) Neuherberg Germany
| | - Matthias Blüher
- Clinic for Endocrinology and NephrologyMedical Research Center Leipzig Germany
| | - Jan Tuckermann
- Institute of Comparative Molecular EndocrinologyUlm University Ulm Germany
| | - Henriette Uhlenhaut
- Research Group Molecular EndocrinologyHelmholtz Center Munich Neuherberg Germany
| | - Stephan Herzig
- Institute for Diabetes and Cancer (IDC)Helmholtz Center Munich Neuherberg Germany
- Joint Heidelberg‐IDC, Inner Medicine 1Heidelberg University Hospital Heidelberg Germany
- German Center for Diabetes Research (DZD) Neuherberg Germany
- School of MedicineTechnical University Munich Munich Germany
| | - Marcel Scheideler
- Institute for Diabetes and Cancer (IDC)Helmholtz Center Munich Neuherberg Germany
- Joint Heidelberg‐IDC, Inner Medicine 1Heidelberg University Hospital Heidelberg Germany
- German Center for Diabetes Research (DZD) Neuherberg Germany
| |
Collapse
|
26
|
Yang F, Dai Y, Min C, Li X. Neonatal overfeeding induced glucocorticoid overexposure accelerates hepatic lipogenesis in male rats. Nutr Metab (Lond) 2018; 15:30. [PMID: 29743929 PMCID: PMC5930793 DOI: 10.1186/s12986-018-0272-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 04/18/2018] [Indexed: 02/07/2023] Open
Abstract
Background Postnatal overfeeding activates tissue glucocorticoid (GC) activity by up-regulating 11β-hydroxysteroid dehydrogenase 1 (11β-HSD1) and increasing sensitivity to high-fat (HF) diet-induced non-alcoholic fatty liver disease (NAFLD). The present study aimed to evaluate the effects of postnatal overfeeding on GC regulation and lipogenesis in the liver and to observe the impact of GC on hepatocyte lipid metabolism. Methods In vivo, Male Sprague-Dawley rat pup litters were adjusted to litter sizes of three (small litter, SL) or ten (normal litter, NL) on postnatal day 3 and then given standard chow from postnatal week 3 (W3) to W13. In vitro, HepG2 cells were stimulated by GC, mifepristone (Mi) or GC + Mi within 48 h, followed by sodium oleate (OA) intervention (or not) for 24 h. Intracellular lipid droplets, triglyceride (TG) concentrations and gene expression related to lipid metabolism were measured in hepatic tissues or HepG2 cells. Results In vivo, weight gain in the body and liver and TG concentrations in the liver were significantly increased in the SL rats compared to the NL rats at W3 and W13 (p < 0.05); mRNA expression of hepatic 11β-HSD1, acetyl-CoA carboxylase 1 (ACC), stearoyl-CoA desaturase-1 (SCD1), fatty acid synthase (FASN) and their nuclear transcription factor, sterol regulatory element binding protein-1c (SREBP-1c) (p < 0.05), was also increased. In vitro, intracellular lipid droplets and TG content in HepG2 cells increased under stimulation with GC or OA (p < 0.05); the increase was more significant following treatment with GC and OA together (p < 0.05). The ACC, SCD1, FASN and SREBP-1c mRNA expression changes were highly similar to the changes in TG content in cells. All the changes induced by GC disappeared when the glucocorticoid receptor (GR) was blocked by Mi. Conclusions Postnatal overfeeding induced GC overexposure through 11β-HSD1 up-regulation in the liver. GC activated hepatic de novo lipogenesis (DNL) via GR and led to hepatic lipid accumulation, which increased the risk of NAFLD during adulthood.
Collapse
Affiliation(s)
- Fan Yang
- 1Department of Child Health Care, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, China
| | - Yanyan Dai
- 1Department of Child Health Care, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, China
| | - Cuiting Min
- 1Department of Child Health Care, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, China
| | - Xiaonan Li
- 1Department of Child Health Care, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing, China.,2Institute of Paediatric Research, Nanjing Medical University, 140 Hanzhong Road, Nanjing, China
| |
Collapse
|
27
|
Geisler CE, Renquist BJ. Hepatic lipid accumulation: cause and consequence of dysregulated glucoregulatory hormones. J Endocrinol 2017; 234:R1-R21. [PMID: 28428362 DOI: 10.1530/joe-16-0513] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 04/20/2017] [Indexed: 12/11/2022]
Abstract
Fatty liver can be diet, endocrine, drug, virus or genetically induced. Independent of cause, hepatic lipid accumulation promotes systemic metabolic dysfunction. By acting as peroxisome proliferator-activated receptor (PPAR) ligands, hepatic non-esterified fatty acids upregulate expression of gluconeogenic, beta-oxidative, lipogenic and ketogenic genes, promoting hyperglycemia, hyperlipidemia and ketosis. The typical hormonal environment in fatty liver disease consists of hyperinsulinemia, hyperglucagonemia, hypercortisolemia, growth hormone deficiency and elevated sympathetic tone. These endocrine and metabolic changes further encourage hepatic steatosis by regulating adipose tissue lipolysis, liver lipid uptake, de novo lipogenesis (DNL), beta-oxidation, ketogenesis and lipid export. Hepatic lipid accumulation may be induced by 4 separate mechanisms: (1) increased hepatic uptake of circulating fatty acids, (2) increased hepatic de novo fatty acid synthesis, (3) decreased hepatic beta-oxidation and (4) decreased hepatic lipid export. This review will discuss the hormonal regulation of each mechanism comparing multiple physiological models of hepatic lipid accumulation. Nonalcoholic fatty liver disease (NAFLD) is typified by increased hepatic lipid uptake, synthesis, oxidation and export. Chronic hepatic lipid signaling through PPARgamma results in gene expression changes that allow concurrent activity of DNL and beta-oxidation. The importance of hepatic steatosis in driving systemic metabolic dysfunction is highlighted by the common endocrine and metabolic disturbances across many conditions that result in fatty liver. Understanding the mechanisms underlying the metabolic dysfunction that develops as a consequence of hepatic lipid accumulation is critical to identifying points of intervention in this increasingly prevalent disease state.
Collapse
Affiliation(s)
- Caroline E Geisler
- School of Animal and Comparative Biomedical SciencesUniversity of Arizona, Tucson, Arizona, USA
| | - Benjamin J Renquist
- School of Animal and Comparative Biomedical SciencesUniversity of Arizona, Tucson, Arizona, USA
| |
Collapse
|
28
|
Scaroni C, Zilio M, Foti M, Boscaro M. Glucose Metabolism Abnormalities in Cushing Syndrome: From Molecular Basis to Clinical Management. Endocr Rev 2017; 38:189-219. [PMID: 28368467 DOI: 10.1210/er.2016-1105] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 03/15/2017] [Indexed: 12/13/2022]
Abstract
An impaired glucose metabolism, which often leads to the onset of diabetes mellitus (DM), is a common complication of chronic exposure to exogenous and endogenous glucocorticoid (GC) excess and plays an important part in contributing to morbidity and mortality in patients with Cushing syndrome (CS). This article reviews the pathogenesis, epidemiology, diagnosis, and management of changes in glucose metabolism associated with hypercortisolism, addressing both the pathophysiological aspects and the clinical and therapeutic implications. Chronic hypercortisolism may have pleiotropic effects on all major peripheral tissues governing glucose homeostasis. Adding further complexity, both genomic and nongenomic mechanisms are directly induced by GCs in a context-specific and cell-/organ-dependent manner. In this paper, the discussion focuses on established and potential pathologic molecular mechanisms that are induced by chronically excessive circulating levels of GCs and affect glucose homeostasis in various tissues. The management of patients with CS and DM includes treating their hyperglycemia and correcting their GC excess. The effects on glycemic control of various medical therapies for CS are reviewed in this paper. The association between DM and subclinical CS and the role of screening for CS in diabetic patients are also discussed.
Collapse
Affiliation(s)
- Carla Scaroni
- Endocrinology Unit, Department of Medicine, DIMED, University of Padova, Via Ospedale 105, 35128 Padua, Italy
| | - Marialuisa Zilio
- Endocrinology Unit, Department of Medicine, DIMED, University of Padova, Via Ospedale 105, 35128 Padua, Italy
| | - Michelangelo Foti
- Department of Cell Physiology & Metabolism, Centre Médical Universitaire, 1 Rue Michel Servet, 1211 Genèva, Switzerland
| | - Marco Boscaro
- Endocrinology Unit, Department of Medicine, DIMED, University of Padova, Via Ospedale 105, 35128 Padua, Italy
| |
Collapse
|
29
|
Zhang Y, Sun X, Icli B, Feinberg MW. Emerging Roles for MicroRNAs in Diabetic Microvascular Disease: Novel Targets for Therapy. Endocr Rev 2017. [DOI: 10.1210/er.2016-1122.2017.1.test] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
30
|
Zhang Y, Sun X, Icli B, Feinberg MW. Emerging Roles for MicroRNAs in Diabetic Microvascular Disease: Novel Targets for Therapy. Endocr Rev 2017; 38:145-168. [PMID: 28323921 PMCID: PMC5460677 DOI: 10.1210/er.2016-1122] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 02/13/2017] [Indexed: 12/11/2022]
Abstract
Chronic, low-grade systemic inflammation and impaired microvascular function are critical hallmarks in the development of insulin resistance. Accordingly, insulin resistance is a major risk factor for type 2 diabetes and cardiovascular disease. Accumulating studies demonstrate that restoration of impaired function of the diabetic macro- and microvasculature may ameliorate a range of cardiovascular disease states and diabetes-associated complications. In this review, we focus on the emerging role of microRNAs (miRNAs), noncoding RNAs that fine-tune target gene expression and signaling pathways, in insulin-responsive tissues and cell types important for maintaining optimal vascular homeostasis and preventing the sequelae of diabetes-induced end organ injury. We highlight current pathophysiological paradigms of miRNAs and their targets involved in regulating the diabetic microvasculature in a range of diabetes-associated complications such as retinopathy, nephropathy, wound healing, and myocardial injury. We provide an update of the potential use of circulating miRNAs diagnostically in type I or type II diabetes. Finally, we discuss emerging delivery platforms for manipulating miRNA expression or function as the next frontier in therapeutic intervention to improve diabetes-associated microvascular dysfunction and its attendant clinical consequences.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Medicine, Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
- Department of Pharmacology and Pharmacy, University of Hong Kong, Pokfulam, Hong Kong SAR, China, and
| | - Xinghui Sun
- Department of Medicine, Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, Nebraska 68588
| | - Basak Icli
- Department of Medicine, Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Mark W. Feinberg
- Department of Medicine, Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
31
|
Abstract
MicroRNAs (miRNAs) are small, non-coding, RNA molecules that regulate gene expression. They have a long evolutionary history and are found in plants, viruses, and animals. Although initially discovered in 1993 in Caenorhabditis elegans, they were not appreciated as widespread and abundant gene regulators until the early 2000s. Studies in the last decade have found that miRNAs confer phenotypic robustness in the face of environmental perturbation, may serve as diagnostic and prognostic indicators of disease, underlie the pathobiology of a wide array of complex disorders, and represent compelling therapeutic targets. Pre-clinical studies in animal models have demonstrated that pharmacologic manipulation of miRNAs, mostly in the liver, can modulate metabolic phenotypes and even reverse the course of insulin resistance and diabetes. There is cautious optimism in the field about miRNA-based therapies for diabetes, several of which are already in various stages of clinical trials. This review will highlight both the promise and the most pressing challenges of therapeutic miRNA silencing in diabetes and related conditions.
Collapse
Affiliation(s)
- Praveen Sethupathy
- Department of Genetics, School of Medicine, UNC Chapel Hill, Chapel Hill, NC, 27599, USA.
- Lineberger Comprehensive Cancer Center, School of Medicine, UNC Chapel Hill, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
32
|
Abstract
In response to stress, the central nervous system initiates a signaling cascade, which leads to the production of glucocorticoids (GCs). GCs act through the glucocorticoid receptor (GR) to coordinate the appropriate cellular response with the primary goal of mobilizing the storage forms of carbon precursors to generate a continuous glucose supply for the brain. Although GCs are critical for maintaining energy homeostasis, excessive GC stimulation leads to a number of undesirable side effects, including hyperglycemia, insulin resistance, fatty liver, obesity, and muscle wasting leading to severe metabolic dysfunction. Summarized below are the diverse metabolic roles of glucocorticoids in energy homeostasis and dysregulation, focusing specifically on glucose, lipid, and protein metabolism.
Collapse
Affiliation(s)
- Lilia Magomedova
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, M5S 3M2, Canada
| | - Carolyn L Cummins
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, M5S 3M2, Canada.
| |
Collapse
|
33
|
Wueest S, Item F, Lucchini FC, Challa TD, Müller W, Blüher M, Konrad D. Mesenteric Fat Lipolysis Mediates Obesity-Associated Hepatic Steatosis and Insulin Resistance. Diabetes 2016; 65:140-8. [PMID: 26384383 DOI: 10.2337/db15-0941] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 09/08/2015] [Indexed: 11/13/2022]
Abstract
Hepatic steatosis and insulin resistance are among the most prevalent metabolic disorders and are tightly associated with obesity and type 2 diabetes. However, the underlying mechanisms linking obesity to hepatic lipid accumulation and insulin resistance are incompletely understood. Glycoprotein 130 (gp130) is the common signal transducer of all interleukin 6 (IL-6) cytokines. We provide evidence that gp130-mediated adipose tissue lipolysis promotes hepatic steatosis and insulin resistance. In obese mice, adipocyte-specific gp130 deletion reduced basal lipolysis and enhanced insulin's ability to suppress lipolysis from mesenteric but not epididymal adipocytes. Consistently, free fatty acid levels were reduced in portal but not in systemic circulation of obese knockout mice. Of note, adipocyte-specific gp130 knockout mice were protected from high-fat diet-induced hepatic steatosis as well as from insulin resistance. In humans, omental but not subcutaneous IL-6 mRNA expression correlated positively with liver lipid accumulation (r = 0.31, P < 0.05) and negatively with hyperinsulinemic-euglycemic clamp glucose infusion rate (r = -0.28, P < 0.05). The results show that IL-6 cytokine-induced lipolysis may be restricted to mesenteric white adipose tissue and that it contributes to hepatic insulin resistance and steatosis. Therefore, blocking IL-6 cytokine signaling in (mesenteric) adipocytes may be a novel approach to blunting detrimental fat-liver crosstalk in obesity.
Collapse
Affiliation(s)
- Stephan Wueest
- Division of Pediatric Endocrinology and Diabetology, University Children's Hospital, Zurich, Switzerland Children's Research Centre, University Children's Hospital, Zurich, Switzerland
| | - Flurin Item
- Division of Pediatric Endocrinology and Diabetology, University Children's Hospital, Zurich, Switzerland Children's Research Centre, University Children's Hospital, Zurich, Switzerland
| | - Fabrizio C Lucchini
- Division of Pediatric Endocrinology and Diabetology, University Children's Hospital, Zurich, Switzerland Children's Research Centre, University Children's Hospital, Zurich, Switzerland Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Tenagne D Challa
- Division of Pediatric Endocrinology and Diabetology, University Children's Hospital, Zurich, Switzerland Children's Research Centre, University Children's Hospital, Zurich, Switzerland
| | - Werner Müller
- Faculty of Life Sciences, University of Manchester, Manchester, U.K
| | - Matthias Blüher
- Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Daniel Konrad
- Division of Pediatric Endocrinology and Diabetology, University Children's Hospital, Zurich, Switzerland Children's Research Centre, University Children's Hospital, Zurich, Switzerland Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
34
|
De Bosscher K, Beck IM, Ratman D, Berghe WV, Libert C. Activation of the Glucocorticoid Receptor in Acute Inflammation: the SEDIGRAM Concept. Trends Pharmacol Sci 2016; 37:4-16. [DOI: 10.1016/j.tips.2015.09.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 09/14/2015] [Accepted: 09/14/2015] [Indexed: 12/21/2022]
|
35
|
Woods CP, Hazlehurst JM, Tomlinson JW. Glucocorticoids and non-alcoholic fatty liver disease. J Steroid Biochem Mol Biol 2015; 154:94-103. [PMID: 26241028 DOI: 10.1016/j.jsbmb.2015.07.020] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 07/28/2015] [Accepted: 07/29/2015] [Indexed: 12/12/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the hepatic manifestation of the global obesity and metabolic disease epidemic and is rapidly becoming the leading cause of liver cirrhosis and indication for liver transplantation worldwide. The hallmark pathological finding in NAFLD is excess lipid accumulation within hepatocytes, but it is a spectrum of disease ranging from benign hepatic steatosis to steatohepatitis through to fibrosis, cirrhosis and risk of hepatocellular carcinoma. The exact pathophysiology remains unclear with a multi-hit hypothesis generally accepted as being required for inflammation and fibrosis to develop after initial steatosis. Glucocorticoids have been implicated in the pathogenesis of NAFLD across all stages. They have a diverse array of metabolic functions that have the potential to drive NAFLD acting on both liver and adipose tissue. In the fasting state, they are able to mobilize lipid, increasing fatty acid delivery and in the fed state can promote lipid accumulation. Their action is controlled at multiple levels and in this review will outline the evidence base for the role of GCs in the pathogenesis of NAFLD from cell systems, rodent models and clinical studies and describe interventional strategies that have been employed to modulate glucocorticoid action as a potential therapeutic strategy.
Collapse
Affiliation(s)
- Conor P Woods
- Oxford Centre for Diabetes Endocrinology & Metabolism (OCDEM), Churchill Hospital, Headington, Oxford, OX3 7LJ, UK
| | - Jonathon M Hazlehurst
- Oxford Centre for Diabetes Endocrinology & Metabolism (OCDEM), Churchill Hospital, Headington, Oxford, OX3 7LJ, UK
| | - Jeremy W Tomlinson
- Oxford Centre for Diabetes Endocrinology & Metabolism (OCDEM), Churchill Hospital, Headington, Oxford, OX3 7LJ, UK.
| |
Collapse
|
36
|
Okun JG, Conway S, Schmidt KV, Schumacher J, Wang X, de Guia R, Zota A, Klement J, Seibert O, Peters A, Maida A, Herzig S, Rose AJ. Molecular regulation of urea cycle function by the liver glucocorticoid receptor. Mol Metab 2015; 4:732-40. [PMID: 26500844 PMCID: PMC4588454 DOI: 10.1016/j.molmet.2015.07.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 07/20/2015] [Accepted: 07/23/2015] [Indexed: 11/21/2022] Open
Abstract
Objective One of the major side effects of glucocorticoid (GC) treatment is lean tissue wasting, indicating a prominent role in systemic amino acid metabolism. In order to uncover a novel aspect of GCs and their intracellular-receptor, the glucocorticoid receptor (GR), on metabolic control, we conducted amino acid and acylcarnitine profiling in human and mouse models of GC/GR gain- and loss-of-function. Methods Blood serum and tissue metabolite levels were determined in Human Addison's disease (AD) patients as well as in mouse models of systemic and liver-specific GR loss-of-function (AAV-miR-GR) with or without dexamethasone (DEX) treatments. Body composition and neuromuscular and metabolic function tests were conducted in vivo and ex vivo, the latter using precision cut liver slices. Results A serum metabolite signature of impaired urea cycle function (i.e. higher [ARG]:[ORN + CIT]) was observed in human (CTRL: 0.45 ± 0.03, AD: 1.29 ± 0.04; p < 0.001) and mouse (AAV-miR-NC: 0.97 ± 0.13, AAV-miR-GR: 2.20 ± 0.19; p < 0.001) GC/GR loss-of-function, with similar patterns also observed in liver. Serum urea levels were consistently affected by GC/GR gain- (∼+32%) and loss (∼−30%) -of-function. Combined liver-specific GR loss-of-function with DEX treatment revealed a tissue-autonomous role for the GR to coordinate an upregulation of liver urea production rate in vivo and ex vivo, and prevent hyperammonaemia and associated neuromuscular dysfunction in vivo. Liver mRNA expression profiling and GR-cistrome mining identified Arginase I (ARG1) a urea cycle gene targeted by the liver GR. Conclusions The liver GR controls systemic and liver urea cycle function by transcriptional regulation of ARG1 expression. Metabolite profiling revealed a role for the HPA-axis-liver GR in regulating urea cycle function in mouse and humans. The liver GR controls enhanced urea cycle function during chronic glucocorticoid exposure. Liver Arginase I is a key urea cycle transcript regulated by the GR.
Collapse
Affiliation(s)
- Jürgen G Okun
- Division of Neuropediatrics and Metabolic Medicine, University Children's Hospital, Heidelberg, Germany
| | - Sean Conway
- Joint Research Division Molecular Metabolic Control, German Cancer Research Center, Center for Molecular Biology, Heidelberg University and Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Kathrin V Schmidt
- Division of Neuropediatrics and Metabolic Medicine, University Children's Hospital, Heidelberg, Germany
| | - Jonas Schumacher
- Joint Research Division Molecular Metabolic Control, German Cancer Research Center, Center for Molecular Biology, Heidelberg University and Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Xiaoyue Wang
- Joint Research Division Molecular Metabolic Control, German Cancer Research Center, Center for Molecular Biology, Heidelberg University and Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Roldan de Guia
- Joint Research Division Molecular Metabolic Control, German Cancer Research Center, Center for Molecular Biology, Heidelberg University and Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Annika Zota
- Joint Research Division Molecular Metabolic Control, German Cancer Research Center, Center for Molecular Biology, Heidelberg University and Heidelberg University Hospital, 69120 Heidelberg, Germany ; Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, and Joint Heidelberg-IDC Translational Diabetes Program, 85764 Neuherberg, Germany
| | - Johanna Klement
- Department of Internal Medicine, University of Lübeck, 23538 Lübeck, Germany
| | - Oksana Seibert
- Joint Research Division Molecular Metabolic Control, German Cancer Research Center, Center for Molecular Biology, Heidelberg University and Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Achim Peters
- Department of Internal Medicine, University of Lübeck, 23538 Lübeck, Germany
| | - Adriano Maida
- Joint Research Division Molecular Metabolic Control, German Cancer Research Center, Center for Molecular Biology, Heidelberg University and Heidelberg University Hospital, 69120 Heidelberg, Germany ; Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, and Joint Heidelberg-IDC Translational Diabetes Program, 85764 Neuherberg, Germany
| | - Stephan Herzig
- Joint Research Division Molecular Metabolic Control, German Cancer Research Center, Center for Molecular Biology, Heidelberg University and Heidelberg University Hospital, 69120 Heidelberg, Germany ; Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, and Joint Heidelberg-IDC Translational Diabetes Program, 85764 Neuherberg, Germany
| | - Adam J Rose
- Joint Research Division Molecular Metabolic Control, German Cancer Research Center, Center for Molecular Biology, Heidelberg University and Heidelberg University Hospital, 69120 Heidelberg, Germany
| |
Collapse
|