1
|
Brent GA. A Historical Reflection on Scientific Advances in Understanding Thyroid Hormone Action. Thyroid 2023; 33:1140-1149. [PMID: 37594753 DOI: 10.1089/thy.2022.0636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/19/2023]
Abstract
Background: Thyroid hormone (TH) has actions in every tissue of the body and is essential for normal development, as well as having important actions in the adult. The earliest markers of TH action that were identified and monitored clinically, even before TH could be measured in serum, included oxygen consumption, basal metabolic rate, serum cholesterol, and deep tendon reflex time. Cellular, rodent, amphibian, zebrafish, and human models have been used to study TH action. Summary: Early studies of the mechanism of TH action focused on saturable-specific triiodothyronine (T3) nuclear binding and direct actions of T3 that altered protein expression. Additional effects of TH were recognized on mitochondria, stimulation of ion transport, especially the sodium potassium ATPase, augmentation of adrenergic signaling, role as a neurotransmitter, and direct plasma membrane effects. The cloning of the thyroid hormone receptor (THR) genes in 1986 and report of the THR crystal structure in 1995 produced rapid progress in understanding the mechanism of TH nuclear action, as well as the development of modified THR ligands. These findings revealed nuances of TH signaling, including the role of nuclear receptor coactivators and corepressors, repression of positively stimulated genes by the unliganded receptor, THR isoform-specific actions of TRα (THRA) and TRβ (THRB), and THR binding DNA as a heterodimer with retinoid-x-receptor (RXR) for genes positively regulated by TH. The identification of genetic disorders of TH transport and signaling, especially Resistance to Thyroid Hormone (RTH) and monocarboxylate transporter 8 (Mct8) defects, has been highly informative with respect to the mechanism of TH action. Conclusions: The impact of THR isoform, post-translational modifications, receptor cofactors, DNA response element, and selective TH tissue uptake, on TH action, have clinical implications for diagnosing and treating thyroid disease. Additionally, these findings have led to the development of novel TH and TH analogue therapies for metabolic, neurological, and cardiovascular diseases.
Collapse
Affiliation(s)
- Gregory A Brent
- Division of Endocrinology, Diabetes, and Metabolism, Departments of Medicine and Physiology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
- Department of Medicine, VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| |
Collapse
|
2
|
Prasad C, Sundar IK. Regulating the Clock: REV-ERB Agonists as Promising Therapeutic Agents. GEN BIOTECHNOLOGY 2023; 2:95-97. [PMID: 37928778 PMCID: PMC10623616 DOI: 10.1089/genbio.2023.0004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 03/03/2023] [Indexed: 11/07/2023]
Affiliation(s)
- Chandrashekhar Prasad
- Division of Pulmonary Critical Care and Sleep Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Isaac Kirubakaran Sundar
- Division of Pulmonary Critical Care and Sleep Medicine, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
3
|
Sun LY, Lyu YY, Zhang HY, Shen Z, Lin GQ, Geng N, Wang YL, Huang L, Feng ZH, Guo X, Lin N, Ding S, Yuan AC, Zhang L, Qian K, Pu J. Nuclear Receptor NR1D1 Regulates Abdominal Aortic Aneurysm Development by Targeting the Mitochondrial Tricarboxylic Acid Cycle Enzyme Aconitase-2. Circulation 2022; 146:1591-1609. [PMID: 35880522 PMCID: PMC9674448 DOI: 10.1161/circulationaha.121.057623] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
BACKGROUND Metabolic disorder increases the risk of abdominal aortic aneurysm (AAA). NRs (nuclear receptors) have been increasingly recognized as important regulators of cell metabolism. However, the role of NRs in AAA development remains largely unknown. METHODS We analyzed the expression profile of the NR superfamily in AAA tissues and identified NR1D1 (NR subfamily 1 group D member 1) as the most highly upregulated NR in AAA tissues. To examine the role of NR1D1 in AAA formation, we used vascular smooth muscle cell (VSMC)-specific, endothelial cell-specific, and myeloid cell-specific conditional Nr1d1 knockout mice in both AngII (angiotensin II)- and CaPO4-induced AAA models. RESULTS Nr1d1 gene expression exhibited the highest fold change among all 49 NRs in AAA tissues, and NR1D1 protein was upregulated in both human and murine VSMCs from AAA tissues. The knockout of Nr1d1 in VSMCs but not endothelial cells and myeloid cells inhibited AAA formation in both AngII- and CaPO4-induced AAA models. Mechanistic studies identified ACO2 (aconitase-2), a key enzyme of the mitochondrial tricarboxylic acid cycle, as a direct target trans-repressed by NR1D1 that mediated the regulatory effects of NR1D1 on mitochondrial metabolism. NR1D1 deficiency restored the ACO2 dysregulation and mitochondrial dysfunction at the early stage of AngII infusion before AAA formation. Supplementation with αKG (α-ketoglutarate, a downstream metabolite of ACO2) was beneficial in preventing and treating AAA in mice in a manner that required NR1D1 in VSMCs. CONCLUSIONS Our data define a previously unrecognized role of nuclear receptor NR1D1 in AAA pathogenesis and an undescribed NR1D1-ACO2 axis involved in regulating mitochondrial metabolism in VSMCs. It is important that our findings suggest αKG supplementation as an effective therapeutic approach for AAA treatment.
Collapse
Affiliation(s)
- Ling-Yue Sun
- State Key Laboratory for Oncogenes and Related Genes, Department of Cardiology (L.-Y.S., Y.-Y.L., H.-Y.Z., Z.S., G.-Q.L., N.G., Z.-H.F., X.G., N.L., S.D., A.-C.Y., J.P.), Shanghai Jiao Tong University, Shanghai, China
| | - Yu-Yan Lyu
- State Key Laboratory for Oncogenes and Related Genes, Department of Cardiology (L.-Y.S., Y.-Y.L., H.-Y.Z., Z.S., G.-Q.L., N.G., Z.-H.F., X.G., N.L., S.D., A.-C.Y., J.P.), Shanghai Jiao Tong University, Shanghai, China
| | - Heng-Yuan Zhang
- State Key Laboratory for Oncogenes and Related Genes, Department of Cardiology (L.-Y.S., Y.-Y.L., H.-Y.Z., Z.S., G.-Q.L., N.G., Z.-H.F., X.G., N.L., S.D., A.-C.Y., J.P.), Shanghai Jiao Tong University, Shanghai, China
| | - Zhi Shen
- State Key Laboratory for Oncogenes and Related Genes, Department of Cardiology (L.-Y.S., Y.-Y.L., H.-Y.Z., Z.S., G.-Q.L., N.G., Z.-H.F., X.G., N.L., S.D., A.-C.Y., J.P.), Shanghai Jiao Tong University, Shanghai, China
| | - Guan-Qiao Lin
- State Key Laboratory for Oncogenes and Related Genes, Department of Cardiology (L.-Y.S., Y.-Y.L., H.-Y.Z., Z.S., G.-Q.L., N.G., Z.-H.F., X.G., N.L., S.D., A.-C.Y., J.P.), Shanghai Jiao Tong University, Shanghai, China
| | - Na Geng
- State Key Laboratory for Oncogenes and Related Genes, Department of Cardiology (L.-Y.S., Y.-Y.L., H.-Y.Z., Z.S., G.-Q.L., N.G., Z.-H.F., X.G., N.L., S.D., A.-C.Y., J.P.), Shanghai Jiao Tong University, Shanghai, China
| | - Yu-Li Wang
- Department of Vascular Surgery (Y.-L.W., L.Z.), Shanghai Jiao Tong University, Shanghai, China
| | - Lin Huang
- Renji Hospital, School of Medicine, School of Biomedical Engineering and Med-X Research Institute (L.H., K.Q.), Shanghai Jiao Tong University, Shanghai, China
| | - Ze-Hao Feng
- State Key Laboratory for Oncogenes and Related Genes, Department of Cardiology (L.-Y.S., Y.-Y.L., H.-Y.Z., Z.S., G.-Q.L., N.G., Z.-H.F., X.G., N.L., S.D., A.-C.Y., J.P.), Shanghai Jiao Tong University, Shanghai, China
| | - Xiao Guo
- State Key Laboratory for Oncogenes and Related Genes, Department of Cardiology (L.-Y.S., Y.-Y.L., H.-Y.Z., Z.S., G.-Q.L., N.G., Z.-H.F., X.G., N.L., S.D., A.-C.Y., J.P.), Shanghai Jiao Tong University, Shanghai, China
| | - Nan Lin
- State Key Laboratory for Oncogenes and Related Genes, Department of Cardiology (L.-Y.S., Y.-Y.L., H.-Y.Z., Z.S., G.-Q.L., N.G., Z.-H.F., X.G., N.L., S.D., A.-C.Y., J.P.), Shanghai Jiao Tong University, Shanghai, China
| | - Song Ding
- State Key Laboratory for Oncogenes and Related Genes, Department of Cardiology (L.-Y.S., Y.-Y.L., H.-Y.Z., Z.S., G.-Q.L., N.G., Z.-H.F., X.G., N.L., S.D., A.-C.Y., J.P.), Shanghai Jiao Tong University, Shanghai, China
| | - An-Cai Yuan
- State Key Laboratory for Oncogenes and Related Genes, Department of Cardiology (L.-Y.S., Y.-Y.L., H.-Y.Z., Z.S., G.-Q.L., N.G., Z.-H.F., X.G., N.L., S.D., A.-C.Y., J.P.), Shanghai Jiao Tong University, Shanghai, China
| | - Lan Zhang
- Department of Vascular Surgery (Y.-L.W., L.Z.), Shanghai Jiao Tong University, Shanghai, China
| | - Kun Qian
- Renji Hospital, School of Medicine, School of Biomedical Engineering and Med-X Research Institute (L.H., K.Q.), Shanghai Jiao Tong University, Shanghai, China
| | - Jun Pu
- State Key Laboratory for Oncogenes and Related Genes, Department of Cardiology (L.-Y.S., Y.-Y.L., H.-Y.Z., Z.S., G.-Q.L., N.G., Z.-H.F., X.G., N.L., S.D., A.-C.Y., J.P.), Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
4
|
Hosokawa H, Akagi R, Watanabe S, Horii M, Shinohara M, Mikami Y, Toguchi K, Kimura S, Yamaguchi S, Ohtori S, Sasho T. Nuclear receptor subfamily 1 group D member 1 in the pathology of obesity-induced osteoarthritis progression. J Orthop Res 2022; 41:930-941. [PMID: 36102152 DOI: 10.1002/jor.25440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 07/28/2022] [Accepted: 09/02/2022] [Indexed: 02/04/2023]
Abstract
Mechanical overload and chemical factors are both related to obesity-induced progression of knee osteoarthritis. The circadian rhythm is related to the development of metabolic syndrome and the progression of osteoarthritis, and the core clock genes nuclear receptor subfamily 1 group D member 1 (NR1D1) and brain and muscle arnt-like protein 1 (BMAL1) are dysregulated in cartilage from patients with osteoarthritis. Here, we focused on NR1D1 and investigated osteoarthritis-related changes and gene expression in a mouse model of diet-induced obesity. A high-fat diet was provided to C57BL6/J mice, and changes in body weight, blood lipids, and gene expression were investigated. Destabilization of the medial meniscus or sham surgery was performed on mice fed a high-fat diet or normal diet, and histological osteoarthritis-related changes and NR1D1 expression were investigated. The effects of the NR1D1 agonist SR9009 were also assessed. Mice fed a high-fat diet developed significant obesity and dyslipidemia. Nr1d1 and Bmal1 gene expression levels decreased in the liver and knee joints. Moreover, increased osteoarthritis progression and decreased NR1D1 protein expression were observed in high-fat diet-fed mice after surgical osteoarthritis induction. SR9009 decreased the progression of obesity, dyslipidemia, and osteoarthritis. Overall, obesity and dyslipidemia induced by the high-fat diet led to osteoarthritis progression and decreased NR1D1 expression. Thus, NR1D1 may play an important role in obesity-induced osteoarthritis.
Collapse
Affiliation(s)
- Hiroaki Hosokawa
- Department of Orthopaedic Surgery, School of Medicine, Chiba University, Chiba, Japan.,Center for Preventive Medicine, Musculoskeletal Disease and Pain, Chiba University, Chiba, Japan
| | - Ryuichiro Akagi
- Department of Orthopaedic Surgery, School of Medicine, Chiba University, Chiba, Japan
| | - Shotaro Watanabe
- Department of Orthopaedic Surgery, School of Medicine, Chiba University, Chiba, Japan.,Center for Preventive Medicine, Musculoskeletal Disease and Pain, Chiba University, Chiba, Japan
| | - Manato Horii
- Department of Orthopaedic Surgery, School of Medicine, Chiba University, Chiba, Japan
| | - Masashi Shinohara
- Department of Orthopaedic Surgery, School of Medicine, Chiba University, Chiba, Japan
| | - Yukio Mikami
- Department of Orthopaedic Surgery, School of Medicine, Chiba University, Chiba, Japan
| | - Kaoru Toguchi
- Department of Orthopaedic Surgery, School of Medicine, Chiba University, Chiba, Japan
| | - Seiji Kimura
- Department of Orthopaedic Surgery, Center for Advanced Joint Function and Reconstructive Spine Surgery Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Satoshi Yamaguchi
- Graduate School of Global and Transdisciplinary Studies, Chiba University, Chiba, Japan
| | - Seiji Ohtori
- Department of Orthopaedic Surgery, School of Medicine, Chiba University, Chiba, Japan
| | - Takahisa Sasho
- Center for Preventive Medicine, Musculoskeletal Disease and Pain, Chiba University, Chiba, Japan
| |
Collapse
|
5
|
Transcriptional Regulation of Hepatic Autophagy by Nuclear Receptors. Cells 2022; 11:cells11040620. [PMID: 35203271 PMCID: PMC8869834 DOI: 10.3390/cells11040620] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 02/04/2023] Open
Abstract
Autophagy is an adaptive self-eating process involved in degradation of various cellular components such as carbohydrates, lipids, proteins, and organelles. Its activity plays an essential role in tissue homeostasis and systemic metabolism in response to diverse challenges, including nutrient depletion, pathogen invasion, and accumulations of toxic materials. Therefore, autophagy dysfunctions are intimately associated with many human diseases such as cancer, neurodegeneration, obesity, diabetes, infection, and aging. Although its acute post-translational regulation is well described, recent studies have also shown that autophagy can be controlled at the transcriptional and post-transcriptional levels. Nuclear receptors (NRs) are in general ligand-dependent transcription factors consisting of 48 members in humans. These receptors extensively control transcription of a variety of genes involved in development, metabolism, and inflammation. In this review, we discuss the roles and mechanisms of NRs in an aspect of transcriptional regulation of hepatic autophagy, and how the NR-driven autophagy pathway can be harnessed to treat various liver diseases.
Collapse
|
6
|
Adlanmerini M, Nguyen HC, Krusen BM, Teng CW, Geisler CE, Peed LC, Carpenter BJ, Hayes MR, Lazar MA. Hypothalamic REV-ERB nuclear receptors control diurnal food intake and leptin sensitivity in diet-induced obese mice. J Clin Invest 2021; 131:140424. [PMID: 33021965 DOI: 10.1172/jci140424] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 09/29/2020] [Indexed: 12/15/2022] Open
Abstract
Obesity occurs when energy expenditure is outweighed by energy intake. Tuberal hypothalamic nuclei, including the arcuate nucleus (ARC), ventromedial nucleus (VMH), and dorsomedial nucleus (DMH), control food intake and energy expenditure. Here we report that, in contrast with females, male mice lacking circadian nuclear receptors REV-ERBα and -β in the tuberal hypothalamus (HDKO mice) gained excessive weight on an obesogenic high-fat diet due to both decreased energy expenditure and increased food intake during the light phase. Moreover, rebound food intake after fasting was markedly increased in HDKO mice. Integrative transcriptomic and cistromic analyses revealed that such disruption in feeding behavior was due to perturbed REV-ERB-dependent leptin signaling in the ARC. Indeed, in vivo leptin sensitivity was impaired in HDKO mice on an obesogenic diet in a diurnal manner. Thus, REV-ERBs play a crucial role in hypothalamic control of food intake and diurnal leptin sensitivity in diet-induced obesity.
Collapse
Affiliation(s)
- Marine Adlanmerini
- Institute for Diabetes, Obesity, and Metabolism and Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine
| | - Hoang Cb Nguyen
- Institute for Diabetes, Obesity, and Metabolism and Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine
| | - Brianna M Krusen
- Institute for Diabetes, Obesity, and Metabolism and Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine
| | - Clare W Teng
- Institute for Diabetes, Obesity, and Metabolism and Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine
| | - Caroline E Geisler
- Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Lindsey C Peed
- Institute for Diabetes, Obesity, and Metabolism and Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine
| | - Bryce J Carpenter
- Institute for Diabetes, Obesity, and Metabolism and Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine
| | - Matthew R Hayes
- Institute for Diabetes, Obesity, and Metabolism and Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine.,Department of Psychiatry, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Mitchell A Lazar
- Institute for Diabetes, Obesity, and Metabolism and Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine
| |
Collapse
|
7
|
Ishii S. The Role of Histone Deacetylase 3 Complex in Nuclear Hormone Receptor Action. Int J Mol Sci 2021; 22:ijms22179138. [PMID: 34502048 PMCID: PMC8431225 DOI: 10.3390/ijms22179138] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 11/16/2022] Open
Abstract
Nuclear hormone receptors (NRs) regulate transcription of the target genes in a ligand-dependent manner in either a positive or negative direction, depending on the case. Deacetylation of histone tails is associated with transcriptional repression. A nuclear receptor corepressor (N-CoR) and a silencing mediator for retinoid and thyroid hormone receptors (SMRT) are the main corepressors responsible for gene suppression mediated by NRs. Among numerous histone deacetylases (HDACs), HDAC3 is the core component of the N-CoR/SMRT complex, and plays a central role in NR-dependent repression. Here, the roles of HDAC3 in ligand-independent repression, gene repression by orphan NRs, NRs antagonist action, ligand-induced repression, and the activation of a transcriptional coactivator are reviewed. In addition, some perspectives regarding the non-canonical mechanisms of HDAC3 action are discussed.
Collapse
Affiliation(s)
- Sumiyasu Ishii
- Department of Integrative Physiology, Gunma University Graduate School of Medicine, Maebashi 371-8501, Japan
| |
Collapse
|
8
|
Guan D, Lazar MA. Interconnections between circadian clocks and metabolism. J Clin Invest 2021; 131:e148278. [PMID: 34338232 DOI: 10.1172/jci148278] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Circadian rhythms evolved through adaptation to daily light/dark changes in the environment; they are believed to be regulated by the core circadian clock interlocking feedback loop. Recent studies indicate that each core component executes general and specific functions in metabolism. Here, we review the current understanding of the role of these core circadian clock genes in the regulation of metabolism using various genetically modified animal models. Additionally, emerging evidence shows that exposure to environmental stimuli, such as artificial light, unbalanced diet, mistimed eating, and exercise, remodels the circadian physiological processes and causes metabolic disorders. This Review summarizes the reciprocal regulation between the circadian clock and metabolism, highlights remaining gaps in knowledge about the regulation of circadian rhythms and metabolism, and examines potential applications to human health and disease.
Collapse
Affiliation(s)
- Dongyin Guan
- Institute for Diabetes, Obesity, and Metabolism.,Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, and
| | - Mitchell A Lazar
- Institute for Diabetes, Obesity, and Metabolism.,Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, and.,Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
9
|
Liu Q, Xu L, Wu M, Zhou Y, Yang J, Huang C, Xu T, Li J, Zhang L. Rev-erbα exacerbates hepatic steatosis in alcoholic liver diseases through regulating autophagy. Cell Biosci 2021; 11:129. [PMID: 34246287 PMCID: PMC8272374 DOI: 10.1186/s13578-021-00622-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 05/31/2021] [Indexed: 12/20/2022] Open
Abstract
Background and aims Alcoholic fatty liver (AFL) is a liver disease caused by long-term excessive drinking and is characterized by hepatic steatosis. Understanding the regulatory mechanism of steatosis is essential for the treatment of AFL. Rev-erbα is a member of the Rev-erbs family of nuclear receptors, playing an important role in regulating lipid metabolism. However, its functional role in AFL and its underlying mechanism remains unclear. Results Rev-erbα was upregulated in the liver of EtOH-fed mice and EtOH-treated L-02 cells. Further, Rev-erbα activation exacerbates steatosis in L-02 cells. Inhibition/downexpression of Rev-erbα improved steatosis. Mechanistically, autophagy activity was inhibited in vivo and vitro. Interestingly, inhibition/downexpression of Rev-erbα enhanced autophagy. Furthermore, silencing of Rev-erbα up-regulated the nuclear expression of Bmal1. Autophagy activity was inhibited and steatosis was deteriorated after EtOH-treated L-02 cells were cotransfected with Rev-erbα shRNA and Bmal1 siRNA. Conclusions Rev-erbα induces liver steatosis, which promotes the progression of AFL. Our study reveals a novel steatosis regulatory mechanism in AFL and suggest that Rev-erbα might be a potential therapeutic target for AFL. Supplementary Information The online version contains supplementary material available at 10.1186/s13578-021-00622-4.
Collapse
Affiliation(s)
- Qingxue Liu
- School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, 230032, Anhui, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, 230032, People's Republic of China.,The Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Lei Xu
- School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, 230032, Anhui, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, 230032, People's Republic of China.,The Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Meifei Wu
- School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, 230032, Anhui, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, 230032, People's Republic of China.,The Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Yiwen Zhou
- School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, 230032, Anhui, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, 230032, People's Republic of China.,The Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Junfa Yang
- School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, 230032, Anhui, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, 230032, People's Republic of China.,The Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Cheng Huang
- School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, 230032, Anhui, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, 230032, People's Republic of China.,The Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Tao Xu
- School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, 230032, Anhui, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, 230032, People's Republic of China.,The Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Jun Li
- School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, 230032, Anhui, China.,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, 230032, People's Republic of China.,The Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China
| | - Lei Zhang
- School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, 230032, Anhui, China. .,Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Hefei, 230032, People's Republic of China. .,The Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, Hefei, 230032, China.
| |
Collapse
|
10
|
Xiao Y, Kim M, Lazar MA. Nuclear receptors and transcriptional regulation in non-alcoholic fatty liver disease. Mol Metab 2020; 50:101119. [PMID: 33220489 PMCID: PMC8324695 DOI: 10.1016/j.molmet.2020.101119] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/13/2020] [Accepted: 11/16/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND As a result of a sedentary lifestyle and excess food consumption in modern society, non-alcoholic fatty liver disease (NAFLD) characterized by fat accumulation in the liver is becoming a major disease burden. Non-alcoholic steatohepatitis (NASH) is an advanced form of NAFLD characterized by inflammation and fibrosis that can lead to hepatocellular carcinoma and liver failure. Nuclear receptors (NRs) are a family of ligand-regulated transcription factors that closely control multiple aspects of metabolism. Their transcriptional activity is modulated by various ligands, including hormones and lipids. NRs serve as potential pharmacological targets for NAFLD/NASH and other metabolic diseases. SCOPE OF REVIEW In this review, we provide a comprehensive overview of NRs that have been studied in the context of NAFLD/NASH with a focus on their transcriptional regulation, function in preclinical models, and studies of their clinical utility. MAJOR CONCLUSIONS The transcriptional regulation of NRs is context-dependent. During the dynamic progression of NAFLD/NASH, NRs play diverse roles in multiple organs and different cell types in the liver, which highlights the necessity of targeting NRs in a stage-specific and cell-type-specific manner to enhance the efficacy and safety of treatment methods.
Collapse
Affiliation(s)
- Yang Xiao
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mindy Kim
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mitchell A Lazar
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
11
|
Kim YH, Lazar MA. Transcriptional Control of Circadian Rhythms and Metabolism: A Matter of Time and Space. Endocr Rev 2020; 41:5835826. [PMID: 32392281 PMCID: PMC7334005 DOI: 10.1210/endrev/bnaa014] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/04/2020] [Indexed: 02/07/2023]
Abstract
All biological processes, living organisms, and ecosystems have evolved with the Sun that confers a 24-hour periodicity to life on Earth. Circadian rhythms arose from evolutionary needs to maximize daily organismal fitness by enabling organisms to mount anticipatory and adaptive responses to recurrent light-dark cycles and associated environmental changes. The clock is a conserved feature in nearly all forms of life, ranging from prokaryotes to virtually every cell of multicellular eukaryotes. The mammalian clock comprises transcription factors interlocked in negative feedback loops, which generate circadian expression of genes that coordinate rhythmic physiology. In this review, we highlight previous and recent studies that have advanced our understanding of the transcriptional architecture of the mammalian clock, with a specific focus on epigenetic mechanisms, transcriptomics, and 3-dimensional chromatin architecture. In addition, we discuss reciprocal ways in which the clock and metabolism regulate each other to generate metabolic rhythms. We also highlight implications of circadian biology in human health, ranging from genetic and environment disruptions of the clock to novel therapeutic opportunities for circadian medicine. Finally, we explore remaining fundamental questions and future challenges to advancing the field forward.
Collapse
Affiliation(s)
- Yong Hoon Kim
- Institute for Diabetes, Obesity, and Metabolism, and Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Mitchell A Lazar
- Institute for Diabetes, Obesity, and Metabolism, and Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
12
|
Kim K, Kim JH, Kim I, Seong S, Kim N. Rev-erbα Negatively Regulates Osteoclast and Osteoblast Differentiation through p38 MAPK Signaling Pathway. Mol Cells 2020; 43:34-47. [PMID: 31896234 PMCID: PMC6999712 DOI: 10.14348/molcells.2019.0232] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 11/21/2019] [Accepted: 12/01/2019] [Indexed: 11/27/2022] Open
Abstract
The circadian clock regulates various physiological processes, including bone metabolism. The nuclear receptors Reverbs, comprising Rev-erbα and Rev-erbβ, play a key role as transcriptional regulators of the circadian clock. In this study, we demonstrate that Rev-erbs negatively regulate differentiation of osteoclasts and osteoblasts. The knockdown of Rev-erbα in osteoclast precursor cells enhanced receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclast formation, as well as expression of nuclear factor of activated T cells 1 (NFATc1), osteoclast-associated receptor (OSCAR), and tartrate-resistant acid phosphatase (TRAP). The overexpression of Rev-erbα leads to attenuation of the NFATc1 expression via inhibition of recruitment of c-Fos to the NFATc1 promoter. The overexpression of Rev-erbα in osteoblast precursors attenuated the expression of osteoblast marker genes including Runx2, alkaline phosphatase (ALP), bone sialoprotein (BSP), and osteocalcin (OC). Rev-erbα interfered with the recruitment of Runx2 to the promoter region of the target genes. Conversely, knockdown of Reverbα in the osteoblast precursors enhanced the osteoblast differentiation and function. In addition, Rev-erbα negatively regulated osteoclast and osteoblast differentiation by suppressing the p38 MAPK pathway. Furthermore, intraperitoneal administration of GSK4112, a Rev-erb agonist, protects RANKL-induced bone loss via inhibition of osteoclast differentiation in vivo . Taken together, our results demonstrate a molecular mechanism of Rev-erbs in the bone remodeling, and provide a molecular basis for a potential therapeutic target for treatment of bone disease characterized by excessive bone resorption.
Collapse
MESH Headings
- Animals
- Bone Remodeling
- Bone Resorption/genetics
- Bone Resorption/metabolism
- Cell Differentiation
- Cells, Cultured
- Circadian Clocks
- Disease Models, Animal
- Gain of Function Mutation/genetics
- Humans
- Male
- Mice
- Mice, Inbred ICR
- Nuclear Receptor Subfamily 1, Group D, Member 1/genetics
- Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism
- Osteoblasts/physiology
- Osteoclasts/physiology
- Osteogenesis/genetics
- RNA, Small Interfering/genetics
- Receptors, Cytoplasmic and Nuclear/genetics
- Receptors, Cytoplasmic and Nuclear/metabolism
- Repressor Proteins/genetics
- Repressor Proteins/metabolism
- Signal Transduction
- p38 Mitogen-Activated Protein Kinases/metabolism
Collapse
Affiliation(s)
- Kabsun Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 61469,
Korea
| | - Jung Ha Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 61469,
Korea
| | - Inyoung Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 61469,
Korea
| | - Semun Seong
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 61469,
Korea
- Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju 61469,
Korea
| | - Nacksung Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 61469,
Korea
- Department of Biomedical Sciences, Chonnam National University Medical School, Gwangju 61469,
Korea
| |
Collapse
|
13
|
M2 Macrophages as a Potential Target for Antiatherosclerosis Treatment. Neural Plast 2019; 2019:6724903. [PMID: 30923552 PMCID: PMC6409015 DOI: 10.1155/2019/6724903] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 11/06/2018] [Accepted: 11/28/2018] [Indexed: 12/24/2022] Open
Abstract
Atherosclerosis is a chronic progressive inflammation course, which could induce life-threatening diseases such as stroke and myocardial infarction. Optimal medical treatments for atherosclerotic risk factors with current antihypertensive and lipid-lowering drugs (for example, statins) are widely used in clinical practice. However, many patients with established disease still continue to have recurrent cardiovascular events in spite of treatment with a state-of-the-art therapy. Atherosclerotic cardiovascular disease (ASCVD) remains the leading cause of mortality worldwide. Hence, current treatment of atherosclerosis is still far from being satisfactory. Recently, M2 macrophages have been found associated with atherosclerosis regression. The M2 phenotype can secrete anti-inflammatory factors such as IL-10 and TGF-β, promote tissue remodeling and repairing through collagen formation, and clear dying cells and debris by efferocytosis. Therefore, modulators targeting macrophages' polarization to the M2 phenotype could be another promising treatment strategy for atherosclerosis. Two main signaling pathways, the Akt/mTORC/LXR pathway and the JAK/STAT6 pathway, are found playing important roles in M2 polarization. In addition, researchers have reported several potential approaches to modulate M2 polarization. Inhibiting or activating some kinds of enzymes, affecting transcription factors, or acting on several membrane receptors could regulate the polarization of the M2 phenotype. Besides, biomolecules, for example vitamin D, were found to affect the process of M2 polarization. Pomegranate juice could promote M2 polarization via unclear mechanism. In this review, we will discuss how M2 macrophages affect atherosclerosis regression, signal transduction in M2 polarization, and outline potential targets and compounds that affect M2 polarization, thus controlling the progress of atherosclerosis.
Collapse
|
14
|
Mazaira GI, Zgajnar NR, Lotufo CM, Daneri-Becerra C, Sivils JC, Soto OB, Cox MB, Galigniana MD. The Nuclear Receptor Field: A Historical Overview and Future Challenges. NUCLEAR RECEPTOR RESEARCH 2018; 5:101320. [PMID: 30148160 PMCID: PMC6108593 DOI: 10.11131/2018/101320] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In this article we summarize the birth of the field of nuclear receptors, the discovery of untransformed and transformed isoforms of ligand-binding macromolecules, the discovery of the three-domain structure of the receptors, and the properties of the Hsp90-based heterocomplex responsible for the overall structure of the oligomeric receptor and many aspects of the biological effects. The discovery and properties of the subfamily of receptors called orphan receptors is also outlined. Novel molecular aspects of the mechanism of action of nuclear receptors and challenges to resolve in the near future are discussed.
Collapse
Affiliation(s)
- Gisela I. Mazaira
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (1428), Argentina
| | - Nadia R. Zgajnar
- Instituto de Biología y Medicina Experimental- CONICET. Buenos Aires (1428), Argentina
| | - Cecilia M. Lotufo
- Instituto de Biología y Medicina Experimental- CONICET. Buenos Aires (1428), Argentina
| | | | - Jeffrey C. Sivils
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968, USA
| | - Olga B. Soto
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968, USA
| | - Marc B. Cox
- Department of Biological Sciences and Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968, USA
| | - Mario D. Galigniana
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (1428), Argentina
- Instituto de Biología y Medicina Experimental- CONICET. Buenos Aires (1428), Argentina
| |
Collapse
|
15
|
Benna C, Helfrich-Förster C, Rajendran S, Monticelli H, Pilati P, Nitti D, Mocellin S. Genetic variation of clock genes and cancer risk: a field synopsis and meta-analysis. Oncotarget 2017; 8:23978-23995. [PMID: 28177907 PMCID: PMC5410358 DOI: 10.18632/oncotarget.15074] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 12/27/2016] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The number of studies on the association between clock genes’ polymorphisms and cancer susceptibility has increased over the last years but the results are often conflicting and no comprehensive overview and quantitative summary of the evidence in this field is available. RESULTS Literature search identified 27 eligible studies comprising 96756 subjects (cases: 38231) and investigating 687 polymorphisms involving 14 clock genes. Overall, 1025 primary and subgroup meta-analyses on 366 gene variants were performed. Study distribution by tumor was as follows: breast cancer (n=15), prostate cancer (n=3), pancreatic cancer (n=2), non-Hodgkin's lymphoma (n=2), glioma (n=1), chronic lymphocytic leukemia (n=1), colorectal cancer (n=1), non-small cell lung cancer (n=1) and ovarian cancer (n=1). We identified 10 single nucleotide polymorphisms (SNPs) significantly associated with cancer risk: NPAS2 rs10165970 (mixed and breast cancer shiftworkers), rs895520 (mixed), rs17024869 (breast) and rs7581886 (breast); CLOCK rs3749474 (breast) and rs11943456 (breast); RORA rs7164773 (breast and breast cancer postmenopausal), rs10519097 (breast); RORB rs7867494 (breast cancer postmenopausal), PER3 rs1012477 (breast cancer subgroups) and assessed the level of quality evidence to be intermediate. We also identified polymorphisms with lower quality statistically significant associations (n=30). CONCLUSIONS Our work supports the hypothesis that genetic variation of clock genes might affect cancer risk. These findings also highlight the need for more efforts in this research field in order to fully establish the contribution of clock gene variants to the risk of developing cancer. METHODS We conducted a systematic review and meta-analysis of the evidence on the association between clock genes’ germline variants and the risk of developing cancer. To assess result credibility, summary evidence was graded according to the Venice criteria and false positive report probability (FPRP) was calculated to further validate result noteworthiness. Subgroup meta-analysis was also performed based on participant features and tumor type. The breast cancer subgroup was further stratified by work conditions, estrogen receptor/progesterone receptor status and menopausal status, conditions associated with the risk of breast cancer in different studies.
Collapse
Affiliation(s)
- Clara Benna
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Charlotte Helfrich-Förster
- Neurobiology and Genetics, Theodor-Boveri Institute, Biocenter, University of Würzburg, Würzburg, Germany
| | - Senthilkumar Rajendran
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padova, Italy
| | | | | | - Donato Nitti
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padova, Italy.,Clinica Chirurgica I, Azienda Ospedaliera Padova, Padova, Italy
| | - Simone Mocellin
- Department of Surgery Oncology and Gastroenterology, University of Padova, Padova, Italy.,Istituto Oncologico Veneto, IOV-IRCSS, Padova, Italy
| |
Collapse
|
16
|
Yang SL, Ren QG, Wen L, Hu JL, Wang HY. Research progress on circadian clock genes in common abdominal malignant tumors. Oncol Lett 2017; 14:5091-5098. [PMID: 29113149 PMCID: PMC5661368 DOI: 10.3892/ol.2017.6856] [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: 01/30/2016] [Accepted: 07/03/2017] [Indexed: 02/01/2023] Open
Abstract
The circadian clock refers to the inherent biological rhythm of an organism, which, is accurately regulated by numerous clock genes. Studies in recent years have reported that the abnormal expression of clock genes is ubiquitous in common abdominal malignant tumors, including liver, colorectal, gastric and pancreatic cancer. In addition, the abnormal expression of certain clock genes is closely associated with clinical tumor parameters or patient prognosis. Studies in clock genes may expand the knowledge about the mechanism of occurrence and development of tumors, and may provide a new approach for tumor therapy. The present study summarizes the research progress in this field.
Collapse
Affiliation(s)
- Sheng-Li Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, P.R. China
| | - Quan-Guang Ren
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, P.R. China
| | - Lu Wen
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, P.R. China
| | - Jian-Li Hu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, P.R. China
| | - Heng-Yi Wang
- Department of Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| |
Collapse
|
17
|
Akagi R, Akatsu Y, Fisch KM, Alvarez-Garcia O, Teramura T, Muramatsu Y, Saito M, Sasho T, Su AI, Lotz MK. Dysregulated circadian rhythm pathway in human osteoarthritis: NR1D1 and BMAL1 suppression alters TGF-β signaling in chondrocytes. Osteoarthritis Cartilage 2017; 25:943-951. [PMID: 27884645 PMCID: PMC5438901 DOI: 10.1016/j.joca.2016.11.007] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 11/08/2016] [Accepted: 11/12/2016] [Indexed: 02/02/2023]
Abstract
OBJECTIVES Circadian rhythm (CR) was identified by RNA sequencing as the most dysregulated pathway in human osteoarthritis (OA) in articular cartilage. This study examined circadian rhythmicity in cultured chondrocytes and the role of the CR genes NR1D1 and BMAL1 in regulating chondrocyte functions. METHODS RNA was extracted from normal and OA-affected human knee cartilage (n = 14 each). Expression levels of NR1D1 and BMAL1 mRNA and protein were assessed by quantitative PCR and immunohistochemistry. Human chondrocytes were synchronized and harvested at regular intervals to examine circadian rhythmicity in RNA and protein expression. Chondrocytes were treated with small interfering RNA (siRNA) for NR1D1 or BMAL1, followed by RNA sequencing and analysis of the effects on the transforming growth factor beta (TGF-β) pathway. RESULTS NR1D1 and BMAL1 mRNA and protein levels were significantly reduced in OA compared to normal cartilage. In cultured human chondrocytes, a clear circadian rhythmicity was observed for NR1D1 and BMAL1. Increased BMAL1 expression was observed after knocking down NR1D1, and decreased NR1D1 levels were observed after knocking down BMAL1. Sequencing of RNA from chondrocytes treated with NR1D1 or BMAL1 siRNA identified 330 and 68 significantly different genes, respectively, and this predominantly affected the TGF-β signaling pathway. CONCLUSIONS The CR pathway is dysregulated in OA cartilage. Interference with circadian rhythmicity in cultured chondrocytes affects TGF-β signaling, which is a central pathway in cartilage homeostasis.
Collapse
Affiliation(s)
- R Akagi
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, USA; Department of Orthopaedic Surgery, School of Medicine, Chiba University, 1-8-1, Inohana, Chuou, Chiba, 260-8677, Japan
| | - Y Akatsu
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, USA; Department of Orthopaedic Surgery, School of Medicine, Chiba University, 1-8-1, Inohana, Chuou, Chiba, 260-8677, Japan
| | - K M Fisch
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, USA
| | - O Alvarez-Garcia
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, USA
| | - T Teramura
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, USA
| | - Y Muramatsu
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, USA
| | - M Saito
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, USA; Department of Orthopaedic Surgery, Toho University Sakura Medical Center, 564-1 Shimoshizu, Sakura, Chiba, 285-8741, Japan
| | - T Sasho
- Department of Orthopaedic Surgery, School of Medicine, Chiba University, 1-8-1, Inohana, Chuou, Chiba, 260-8677, Japan
| | - A I Su
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, USA
| | - M K Lotz
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, USA.
| |
Collapse
|
18
|
Savisaar R, Hurst LD. Estimating the prevalence of functional exonic splice regulatory information. Hum Genet 2017; 136:1059-1078. [PMID: 28405812 PMCID: PMC5602102 DOI: 10.1007/s00439-017-1798-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 04/04/2017] [Indexed: 12/14/2022]
Abstract
In addition to coding information, human exons contain sequences necessary for correct splicing. These elements are known to be under purifying selection and their disruption can cause disease. However, the density of functional exonic splicing information remains profoundly uncertain. Several groups have experimentally investigated how mutations at different exonic positions affect splicing. They have found splice information to be distributed widely in exons, with one estimate putting the proportion of splicing-relevant nucleotides at >90%. These results suggest that splicing could place a major pressure on exon evolution. However, analyses of sequence conservation have concluded that the need to preserve splice regulatory signals only slightly constrains exon evolution, with a resulting decrease in the average human rate of synonymous evolution of only 1–4%. Why do these two lines of research come to such different conclusions? Among other reasons, we suggest that the methods are measuring different things: one assays the density of sites that affect splicing, the other the density of sites whose effects on splicing are visible to selection. In addition, the experimental methods typically consider short exons, thereby enriching for nucleotides close to the splice junction, such sites being enriched for splice-control elements. By contrast, in part owing to correction for nucleotide composition biases and to the assumption that constraint only operates on exon ends, the conservation-based methods can be overly conservative.
Collapse
Affiliation(s)
- Rosina Savisaar
- The Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, Bath, BA2 7AY, UK.
| | - Laurence D Hurst
- The Milner Centre for Evolution, Department of Biology and Biochemistry, University of Bath, Bath, BA2 7AY, UK
| |
Collapse
|
19
|
Carter EL, Gupta N, Ragsdale SW. High Affinity Heme Binding to a Heme Regulatory Motif on the Nuclear Receptor Rev-erbβ Leads to Its Degradation and Indirectly Regulates Its Interaction with Nuclear Receptor Corepressor. J Biol Chem 2015; 291:2196-222. [PMID: 26670607 DOI: 10.1074/jbc.m115.670281] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Indexed: 01/11/2023] Open
Abstract
Rev-erbα and Rev-erbβ are heme-binding nuclear receptors (NR) that repress the transcription of genes involved in regulating metabolism, inflammation, and the circadian clock. Previous gene expression and co-immunoprecipitation studies led to a model in which heme binding to Rev-erbα recruits nuclear receptor corepressor 1 (NCoR1) into an active repressor complex. However, in contradiction, biochemical and crystallographic studies have shown that heme decreases the affinity of the ligand-binding domain of Rev-erb NRs for NCoR1 peptides. One explanation for this discrepancy is that the ligand-binding domain and NCoR1 peptides used for in vitro studies cannot replicate the key features of the full-length proteins used in cellular studies. However, the combined in vitro and cellular results described here demonstrate that heme does not directly promote interactions between full-length Rev-erbβ (FLRev-erbβ) and an NCoR1 construct encompassing all three NR interaction domains. NCoR1 tightly binds both apo- and heme-replete FLRev-erbβ·DNA complexes; furthermore, heme, at high concentrations, destabilizes the FLRev-erbβ·NCoR1 complex. The interaction between FLRev-erbβ and NCoR1 as well as Rev-erbβ repression at the Bmal1 promoter appear to be modulated by another cellular factor(s), at least one of which is related to the ubiquitin-proteasome pathway. Our studies suggest that heme is involved in regulating the degradation of Rev-erbβ in a manner consistent with its role in circadian rhythm maintenance. Finally, the very slow rate constant (10(-6) s(-1)) of heme dissociation from Rev-erbβ rules out a prior proposal that Rev-erbβ acts as an intracellular heme sensor.
Collapse
Affiliation(s)
- Eric L Carter
- From the Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan 48109
| | - Nirupama Gupta
- From the Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan 48109
| | - Stephen W Ragsdale
- From the Department of Biological Chemistry, University of Michigan, Ann Arbor, Michigan 48109
| |
Collapse
|
20
|
Munroe SH, Morales CH, Duyck TH, Waters PD. Evolution of the Antisense Overlap between Genes for Thyroid Hormone Receptor and Rev-erbα and Characterization of an Exonic G-Rich Element That Regulates Splicing of TRα2 mRNA. PLoS One 2015; 10:e0137893. [PMID: 26368571 PMCID: PMC4569393 DOI: 10.1371/journal.pone.0137893] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 08/24/2015] [Indexed: 11/18/2022] Open
Abstract
The α-thyroid hormone receptor gene (TRα) codes for two functionally distinct proteins: TRα1, the α-thyroid hormone receptor; and TRα2, a non-hormone-binding variant. The final exon of TRα2 mRNA overlaps the 3' end of Rev-erbα mRNA, which encodes another nuclear receptor on the opposite strand of DNA. To understand the evolution of this antisense overlap, we sequenced these genes and mRNAs in the platypus Orthorhynchus anatinus. Despite its strong homology with other mammals, the platypus TRα/Rev-erbα locus lacks elements essential for expression of TRα2. Comparative analysis suggests that alternative splicing of TRα2 mRNA expression evolved in a stepwise fashion before the divergence of eutherian and marsupial mammals. A short G-rich element (G30) located downstream of the alternative 3'splice site of TRα2 mRNA and antisense to the 3'UTR of Rev-erbα plays an important role in regulating TRα2 splicing. G30 is tightly conserved in eutherian mammals, but is absent in marsupials and monotremes. Systematic deletions and substitutions within G30 have dramatically different effects on TRα2 splicing, leading to either its inhibition or its enhancement. Mutations that disrupt one or more clusters of G residues enhance splicing two- to three-fold. These results suggest the G30 sequence can adopt a highly structured conformation, possibly a G-quadruplex, and that it is part of a complex splicing regulatory element which exerts both positive and negative effects on TRα2 expression. Since mutations that strongly enhance splicing in vivo have no effect on splicing in vitro, it is likely that the regulatory role of G30 is mediated through linkage of transcription and splicing.
Collapse
Affiliation(s)
- Stephen H. Munroe
- Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin, United States of America
- * E-mail:
| | - Christopher H. Morales
- Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin, United States of America
| | - Tessa H. Duyck
- Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin, United States of America
| | - Paul D. Waters
- School of Biotechnology and Biomolecular Sciences, Faculty of Science, UNSW Australia, Sydney, Australia
| |
Collapse
|
21
|
Abstract
Circadian rhythm, or daily oscillation, of behaviors and biological processes is a fundamental feature of mammalian physiology that has developed over hundreds of thousands of years under the continuous evolutionary pressure of energy conservation and efficiency. Evolution has fine-tuned the body's clock to anticipate and respond to numerous environmental cues in order to maintain homeostatic balance and promote survival. However, we now live in a society in which these classic circadian entrainment stimuli have been dramatically altered from the conditions under which the clock machinery was originally set. A bombardment of artificial lighting, heating, and cooling systems that maintain constant ambient temperature; sedentary lifestyle; and the availability of inexpensive, high-calorie foods has threatened even the most powerful and ancient circadian programming mechanisms. Such environmental changes have contributed to the recent staggering elevation in lifestyle-influenced pathologies, including cancer, cardiovascular disease, depression, obesity, and diabetes. This review scrutinizes the role of the body's internal clocks in the hard-wiring of circadian networks that have evolved to achieve energetic balance and adaptability, and it discusses potential therapeutic strategies to reset clock metabolic control to modern time for the benefit of human health.
Collapse
Affiliation(s)
- Zachary Gerhart-Hines
- Section for Metabolic Receptology (Z.G.-H.), Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; and Division of Endocrinology, Diabetes, and Metabolism (M.A.L.), Department of Medicine, Department of Genetics, and The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Mitchell A Lazar
- Section for Metabolic Receptology (Z.G.-H.), Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; and Division of Endocrinology, Diabetes, and Metabolism (M.A.L.), Department of Medicine, Department of Genetics, and The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104
| |
Collapse
|
22
|
Everett LJ, Lazar MA. Nuclear receptor Rev-erbα: up, down, and all around. Trends Endocrinol Metab 2014; 25:586-92. [PMID: 25066191 PMCID: PMC4252361 DOI: 10.1016/j.tem.2014.06.011] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Revised: 06/23/2014] [Accepted: 06/27/2014] [Indexed: 02/08/2023]
Abstract
Rev-erbα is a nuclear receptor that links circadian rhythms to transcriptional control of metabolic pathways. Rev-erbα is a potent transcriptional repressor and plays an important role in the core mammalian molecular clock while also serving as a key regulator of clock output in metabolic tissues including liver and brown adipose tissue (BAT). Recent findings have shed new light on the role of Rev-erbα and its paralog Rev-erbβ in rhythm generation, as well as additional regulatory roles for Rev-erbα in other tissues that contribute to energy expenditure, inflammation, and behavior. This review highlights physiological functions of Rev-erbα and β in multiple tissues and discusses the therapeutic potential and challenges of targeting these pathways in human disease.
Collapse
Affiliation(s)
- Logan J Everett
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Department of Genetics, and The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mitchell A Lazar
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Department of Genetics, and The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
23
|
Matta-Camacho E, Banerjee S, Hughes TS, Solt LA, Wang Y, Burris TP, Kojetin DJ. Structure of REV-ERBβ ligand-binding domain bound to a porphyrin antagonist. J Biol Chem 2014; 289:20054-66. [PMID: 24872411 DOI: 10.1074/jbc.m113.545111] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
REV-ERBα and REV-ERBβ are members of the nuclear receptor (NR) superfamily of ligand-regulated transcription factors that play important roles in the regulation of circadian physiology, metabolism, and immune function. Although the REV-ERBs were originally characterized as orphan receptors, recent studies have demonstrated that they function as receptors for heme. Here, we demonstrate that cobalt protoporphyrin IX (CoPP) and zinc protoporphyrin IX (ZnPP) are ligands that bind directly to the REV-ERBs. However, instead of mimicking the agonist action of heme, CoPP and ZnPP function as antagonists of REV-ERB function. This was unexpected because the only distinction between these ligands is the metal ion that is coordinated. To understand the structural basis by which REV-ERBβ can differentiate between a porphyrin agonist and antagonist, we characterized the interaction between REV-ERBβ with heme, CoPP, and ZnPP using biochemical and structural approaches, including x-ray crystallography and NMR. The crystal structure of CoPP-bound REV-ERBβ indicates only minor conformational changes induced by CoPP compared with heme, including the porphyrin ring of CoPP, which adopts a planar conformation as opposed to the puckered conformation observed in the heme-bound REV-ERBβ crystal structure. Thus, subtle changes in the porphyrin metal center and ring conformation may influence the agonist versus antagonist action of porphyrins and when considered with other studies suggest that gas binding to the iron metal center heme may drive alterations in REV-ERB activity.
Collapse
Affiliation(s)
- Edna Matta-Camacho
- From the Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, Florida 33418 and
| | - Subhashis Banerjee
- From the Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, Florida 33418 and
| | - Travis S Hughes
- From the Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, Florida 33418 and
| | - Laura A Solt
- From the Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, Florida 33418 and
| | - Yongjun Wang
- From the Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, Florida 33418 and the Department of Pharmacological and Physiological Sciences, St. Louis University School of Medicine, St. Louis, Missouri 63103
| | - Thomas P Burris
- From the Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, Florida 33418 and the Department of Pharmacological and Physiological Sciences, St. Louis University School of Medicine, St. Louis, Missouri 63103
| | - Douglas J Kojetin
- From the Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, Florida 33418 and
| |
Collapse
|
24
|
Abstract
The nuclear receptors REV-ERB (consisting of REV-ERBα and REV-ERBβ) and retinoic acid receptor-related orphan receptors (RORs; consisting of RORα, RORβ and RORγ) are involved in many physiological processes, including regulation of metabolism, development and immunity as well as the circadian rhythm. The recent characterization of endogenous ligands for these former orphan nuclear receptors has stimulated the development of synthetic ligands and opened up the possibility of targeting these receptors to treat several diseases, including diabetes, atherosclerosis, autoimmunity and cancer. This Review focuses on the latest developments in ROR and REV-ERB pharmacology indicating that these nuclear receptors are druggable targets and that ligands targeting these receptors may be useful in the treatment of several disorders.
Collapse
|
25
|
Abstract
The nuclear receptor superfamily includes many receptors, identified based on their similarity to steroid hormone receptors but without a known ligand. The study of how these receptors are diversely regulated to interact with genomic regions to control a plethora of biological processes has provided critical insight into development, physiology, and the molecular pathology of disease. Here we provide a compendium of these so-called orphan receptors and focus on what has been learned about their modes of action, physiological functions, and therapeutic promise.
Collapse
Affiliation(s)
- Shannon E Mullican
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, and The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | |
Collapse
|
26
|
Ma H, Zhong W, Jiang Y, Fontaine C, Li S, Fu J, Olkkonen VM, Staels B, Yan D. Increased atherosclerotic lesions in LDL receptor deficient mice with hematopoietic nuclear receptor Rev-erbα knock- down. J Am Heart Assoc 2013; 2:e000235. [PMID: 23963755 PMCID: PMC3828791 DOI: 10.1161/jaha.113.000235] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Nuclear receptor Rev-erbα plays important roles in circadian clock timing, lipid metabolism, adipogenesis, and vascular inflammation. However, the role of Rev-erbα in atherosclerotic lesion development has not been assessed in vivo. METHODS AND RESULTS The nuclear receptor Rev-erbα was knocked down in mouse haematopoietic cells by means of shRNA-lentiviral transduction, followed by bone marrow transplantation into LDL receptor knockout mice. The Rev-erbα protein in peripheral macrophage was reduced by 70% as compared to control mice injected with nontargeting shRNA lentivirus-transduced bone marrow. A significant increase in atherosclerotic lesions was observed around the aorta valves as well as upon en face aorta analysis of Rev-erbα knock-down bone marrow recipients (P<0.01) as compared to the control mice, while plasma cholesterol, phospholipid, and triacylglycerol levels were not affected. Overexpression of Rev-erbα in bone marrow mononuclear cells decreased inflammatory M1 while increasing M2 macrophage markers, while Rev-erbα knock down increased the macrophage inflammatory phenotype in vitro and in vivo. Furthermore, treatment of differentiating macrophages with the Rev-erbα ligand heme promoted expression of antiinflammatory M2 markers. CONCLUSIONS These observations identify hematopoietic cell Rev-erbα as a new modulator of atherogenesis in mice.
Collapse
Affiliation(s)
- Hongling Ma
- Department of Biotechnology, Jinan University, Guangzhou, China
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Chandra V, Mahajan S, Saini A, Dkhar HK, Nanduri R, Raj EB, Kumar A, Gupta P. Human IL10 gene repression by Rev-erbα ameliorates Mycobacterium tuberculosis clearance. J Biol Chem 2013; 288:10692-702. [PMID: 23449984 PMCID: PMC3624449 DOI: 10.1074/jbc.m113.455915] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Revised: 02/28/2013] [Indexed: 12/12/2022] Open
Abstract
Nuclear receptors modulate macrophage effector functions, which are imperative for clearance or survival of mycobacterial infection. The adopted orphan nuclear receptor Rev-erbα is a constitutive transcriptional repressor as it lacks AF2 domain and was earlier shown to be present in macrophages. In the present study, we highlight the differences in the relative subcellular localization of Rev-erbα in monocytes and macrophages. The nuclear localization of Rev-erbα in macrophages is subsequent to monocyte differentiation. Expression analysis of Rev-erbα elucidated it to be considerably more expressed in M1 phenotype in comparison with M2. Rev-erbα overexpression augments antimycobacterial properties of macrophage by keeping IL10 in a basal repressed state. Further, promoter analysis revealed that IL10 promoter harbors a Rev-erbα binding site exclusive to humans and higher order primates and not mouse, demonstrating a species barrier in its functionality. This direct gene repression is mediated by recruitment of co-repressors NCoR and HDAC3. In addition, our data elucidate that its overexpression reduced the survival of intracellular pathogen Mycobacterium tuberculosis by enhancing phagosome lysosome maturation, an event resulting from IL10 repression. Thus, these findings suggest that Rev-erbα bestows protection against mycobacterial infection by direct gene repression of IL10 and thus provide a novel target in modulating macrophage microbicidal properties.
Collapse
Affiliation(s)
- Vemika Chandra
- From the Department of Protein Science and Molecular Biology, Institute of Microbial Technology/Council of Scientific and Industrial Research (CSIR), Sector 39 A, Chandigarh 160036, India
| | - Sahil Mahajan
- From the Department of Protein Science and Molecular Biology, Institute of Microbial Technology/Council of Scientific and Industrial Research (CSIR), Sector 39 A, Chandigarh 160036, India
| | - Ankita Saini
- From the Department of Protein Science and Molecular Biology, Institute of Microbial Technology/Council of Scientific and Industrial Research (CSIR), Sector 39 A, Chandigarh 160036, India
| | - Hedwin K. Dkhar
- From the Department of Protein Science and Molecular Biology, Institute of Microbial Technology/Council of Scientific and Industrial Research (CSIR), Sector 39 A, Chandigarh 160036, India
| | - Ravikanth Nanduri
- From the Department of Protein Science and Molecular Biology, Institute of Microbial Technology/Council of Scientific and Industrial Research (CSIR), Sector 39 A, Chandigarh 160036, India
| | - Ella B. Raj
- From the Department of Protein Science and Molecular Biology, Institute of Microbial Technology/Council of Scientific and Industrial Research (CSIR), Sector 39 A, Chandigarh 160036, India
| | - Ashwani Kumar
- From the Department of Protein Science and Molecular Biology, Institute of Microbial Technology/Council of Scientific and Industrial Research (CSIR), Sector 39 A, Chandigarh 160036, India
| | - Pawan Gupta
- From the Department of Protein Science and Molecular Biology, Institute of Microbial Technology/Council of Scientific and Industrial Research (CSIR), Sector 39 A, Chandigarh 160036, India
| |
Collapse
|
28
|
Abstract
Long noncoding RNAs (lncRNAs) have gained widespread attention in recent years as a potentially new and crucial layer of biological regulation. lncRNAs of all kinds have been implicated in a range of developmental processes and diseases, but knowledge of the mechanisms by which they act is still surprisingly limited, and claims that almost the entirety of the mammalian genome is transcribed into functional noncoding transcripts remain controversial. At the same time, a small number of well-studied lncRNAs have given us important clues about the biology of these molecules, and a few key functional and mechanistic themes have begun to emerge, although the robustness of these models and classification schemes remains to be seen. Here, we review the current state of knowledge of the lncRNA field, discussing what is known about the genomic contexts, biological functions, and mechanisms of action of lncRNAs. We also reflect on how the recent interest in lncRNAs is deeply rooted in biology's longstanding concern with the evolution and function of genomes.
Collapse
Affiliation(s)
- Johnny T Y Kung
- Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02114, USA
| | | | | |
Collapse
|
29
|
Bugge A, Feng D, Everett LJ, Briggs ER, Mullican SE, Wang F, Jager J, Lazar MA. Rev-erbα and Rev-erbβ coordinately protect the circadian clock and normal metabolic function. Genes Dev 2012; 26:657-67. [PMID: 22474260 DOI: 10.1101/gad.186858.112] [Citation(s) in RCA: 391] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The nuclear receptor Rev-erbα regulates circadian rhythm and metabolism, but its effects are modest and it has been considered to be a secondary regulator of the cell-autonomous clock. Here we report that depletion of Rev-erbα together with closely related Rev-erbβ has dramatic effects on the cell-autonomous clock as well as hepatic lipid metabolism. Mouse embryonic fibroblasts were rendered arrhythmic by depletion of both Rev-erbs. In mouse livers, Rev-erbβ mRNA and protein levels oscillate with a diurnal pattern similar to that of Rev-erbα, and both Rev-erbs are recruited to a remarkably similar set of binding sites across the genome, enriched near metabolic genes. Depletion of both Rev-erbs in liver synergistically derepresses several metabolic genes as well as genes that control the positive limb of the molecular clock. Moreover, deficiency of both Rev-erbs causes marked hepatic steatosis, in contrast to relatively subtle changes upon loss of either subtype alone. These findings establish the two Rev-erbs as major regulators of both clock function and metabolism, displaying a level of subtype collaboration that is unusual among nuclear receptors but common among core clock proteins, protecting the organism from major perturbations in circadian and metabolic physiology.
Collapse
Affiliation(s)
- Anne Bugge
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Department of Genetics, The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Shin Y, Noel R, Banerjee S, Kojetin D, Song X, He Y, Lin L, Cameron MD, Burris TP, Kamenecka TM. Small molecule tertiary amines as agonists of the nuclear hormone receptor Rev-erbα. Bioorg Med Chem Lett 2012; 22:4413-7. [PMID: 22633688 DOI: 10.1016/j.bmcl.2012.04.126] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2012] [Revised: 04/25/2012] [Accepted: 04/27/2012] [Indexed: 10/28/2022]
Abstract
The structure-activity relationship study of a small molecule Rev-erbα agonist is reported. The potency and efficacy of the agonists in a cell-based assay were optimized as compared to the initial lead. Modest mouse pharmacokinetics coupled with an improved in vitro profile make 12e a suitable in vivo probe to interrogate the functions of Rev-erbα in animal models of disease.
Collapse
Affiliation(s)
- Youseung Shin
- Department of Molecular Therapeutics and Translational Research Institute, The Scripps Research Institute, Scripps Florida, 130 Scripps Way #A2A, Jupiter, FL 33458, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
|
32
|
Chan MC, Spieth PM, Quinn K, Parotto M, Zhang H, Slutsky AS. Circadian rhythms: from basic mechanisms to the intensive care unit. Crit Care Med 2012; 40:246-53. [PMID: 21926587 DOI: 10.1097/ccm.0b013e31822f0abe] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE : Circadian rhythms are intrinsic timekeeping mechanisms that allow for adaptation to cyclic environmental changes. Increasing evidence suggests that circadian rhythms may influence progression of a variety of diseases as well as effectiveness and toxicity of drugs commonly used in the intensive care unit. In this perspective, we provide a brief review of the molecular mechanisms of circadian rhythms and its relevance to critical care. DATA SOURCES, STUDY SELECTION, DATA EXTRACTION, AND DATA SYNTHESIS:: Articles related to circadian rhythms and organ systems in normal and disease conditions were searched through the PubMed library with the goal of providing a concise review. CONCLUSIONS : Critically ill patients may be highly vulnerable to disruption of circadian rhythms as a result of the severity of their underlying diseases as well as the intensive care unit environment where noise and frequent therapeutic/diagnostic interventions take place. Further basic and clinical research addressing the importance of circadian rhythms in the context of critical care is warranted to develop a better understanding of the complex pathophysiology of critically ill patients as well as to identify novel therapeutic approaches for these patients.
Collapse
Affiliation(s)
- Ming-Cheng Chan
- Keenan Research Centre at the Li Ka Shing Knowledge Institute of St Michael's Hospital, Toronto, Ontario, Canada
| | | | | | | | | | | |
Collapse
|
33
|
Nuclear receptor Rev-erb alpha (Nr1d1) functions in concert with Nr2e3 to regulate transcriptional networks in the retina. PLoS One 2011; 6:e17494. [PMID: 21408158 PMCID: PMC3050883 DOI: 10.1371/journal.pone.0017494] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2010] [Accepted: 02/07/2011] [Indexed: 12/23/2022] Open
Abstract
The majority of diseases in the retina are caused by genetic mutations affecting the development and function of photoreceptor cells. The transcriptional networks directing these processes are regulated by genes such as nuclear hormone receptors. The nuclear hormone receptor gene Rev-erb alpha/Nr1d1 has been widely studied for its role in the circadian cycle and cell metabolism, however its role in the retina is unknown. In order to understand the role of Rev-erb alpha/Nr1d1 in the retina, we evaluated the effects of loss of Nr1d1 to the developing retina and its co-regulation with the photoreceptor-specific nuclear receptor gene Nr2e3 in the developing and mature retina. Knock-down of Nr1d1 expression in the developing retina results in pan-retinal spotting and reduced retinal function by electroretinogram. Our studies show that NR1D1 protein is co-expressed with NR2E3 in the outer neuroblastic layer of the developing mouse retina. In the adult retina, NR1D1 is expressed in the ganglion cell layer and is co-expressed with NR2E3 in the outer nuclear layer, within rods and cones. Several genes co-targeted by NR2E3 and NR1D1 were identified that include: Nr2c1, Recoverin, Rgr, Rarres2, Pde8a, and Nupr1. We examined the cyclic expression of Nr1d1 and Nr2e3 over a twenty-four hour period and observed that both nuclear receptors cycle in a similar manner. Taken together, these studies reveal a novel role for Nr1d1, in conjunction with its cofactor Nr2e3, in regulating transcriptional networks critical for photoreceptor development and function.
Collapse
|
34
|
Rindfleisch BC, Brown MS, VandeBerg JL, Munroe SH. Structure and expression of two nuclear receptor genes in marsupials: insights into the evolution of the antisense overlap between the α-thyroid hormone receptor and Rev-erbα. BMC Mol Biol 2010; 11:97. [PMID: 21143985 PMCID: PMC3047299 DOI: 10.1186/1471-2199-11-97] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2010] [Accepted: 12/10/2010] [Indexed: 11/30/2022] Open
Abstract
Background Alternative processing of α-thyroid hormone receptor (TRα, NR1A1) mRNAs gives rise to two functionally antagonistic nuclear receptors: TRα1, the α-type receptor, and TRα2, a non-hormone binding variant that is found only in mammals. TRα2 shares an unusual antisense coding overlap with mRNA for Rev-erbα (NR1D1), another nuclear receptor protein. In this study we examine the structure and expression of these genes in the gray short-tailed opossum, Monodelphis domestica, in comparison with that of eutherian mammals and three other marsupial species, Didelphis virginiana, Potorous tridactylus and Macropus eugenii, in order to understand the evolution and regulatory role of this antisense overlap. Results The sequence, expression and genomic organization of mRNAs encoding TRα1 and Rev-erbα are very similar in the opossum and eutherian mammals. However, the sequence corresponding to the TRα2 coding region appears truncated by almost 100 amino acids. While expression of TRα1 and Rev-erbα was readily detected in all tissues of M. domestica ages 0 days to 18 weeks, TRα2 mRNA was not detected in any tissue or stage examined. These results contrast with the widespread and abundant expression of TRα2 in rodents and other eutherian mammals. To examine requirements for alternative splicing of TRα mRNAs, a series of chimeric minigenes was constructed. Results show that the opossum TRα2-specific 5' splice site sequence is fully competent for splicing but the sequence homologous to the TRα2 3' splice site is not, even though the marsupial sequences are remarkably similar to core splice site elements in rat. Conclusions Our results strongly suggest that the variant nuclear receptor isoform, TRα2, is not expressed in marsupials and that the antisense overlap between TRα and Rev-erbα thus is unique to eutherian mammals. Further investigation of the TRα and Rev-erbα genes in marsupial and eutherian species promises to yield additional insight into the physiological function of TRα2 and the role of the associated antisense overlap with Rev-erbα in regulating expression of these genes.
Collapse
|
35
|
Schupp M, Lazar MA. Endogenous ligands for nuclear receptors: digging deeper. J Biol Chem 2010; 285:40409-15. [PMID: 20956526 DOI: 10.1074/jbc.r110.182451] [Citation(s) in RCA: 130] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Nuclear receptors (NRs) are hormone-sensing transcription factors that translate dietary or endocrine signals into changes in gene expression. Therefore, the adoption of orphan NRs through the identification of their endogenous ligands is a key element for our understanding of their biology. In this minireview, we give an update on recent progress in regard to endogenous ligands for a cluster of NRs with high sequence homology, namely peroxisome proliferator-activated receptors α and γ, Rev-erbα, and related receptors. This knowledge about the nature and physiology of these ligands may create new opportunities for therapeutic drug development.
Collapse
Affiliation(s)
- Michael Schupp
- Department of Endocrinology, Diabetes, and Nutrition and Center for Cardiovascular Research, Institute of Pharmacology, Charité-University Medicine Berlin, 10115 Berlin, Germany.
| | | |
Collapse
|
36
|
Yin L, Wu N, Lazar MA. Nuclear receptor Rev-erbalpha: a heme receptor that coordinates circadian rhythm and metabolism. NUCLEAR RECEPTOR SIGNALING 2010; 8:e001. [PMID: 20414452 PMCID: PMC2858265 DOI: 10.1621/nrs.08001] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2009] [Accepted: 02/24/2010] [Indexed: 12/22/2022]
Abstract
Nuclear receptor Rev-erbα (NR1D1), previously considered to be an orphan nuclear receptor, is a receptor for heme, which promotes transcriptional repression via recruitment of the NCoR-HDAC3 corepressor complex. Rev-erbα gene regulation is circadian, and Rev-erbα comprises a critical negative limb of the core circadian clock by directly repressing the expression of the positive clock component, Bmal1. Rev-erbα also regulates the metabolic gene pathway, thus serving as a heme sensor for coordination of circadian and metabolic pathways.
Collapse
Affiliation(s)
- Lei Yin
- University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | | | | |
Collapse
|
37
|
Salato VK, Rediske NW, Zhang C, Hastings ML, Munroe SH. An exonic splicing enhancer within a bidirectional coding sequence regulates alternative splicing of an antisense mRNA. RNA Biol 2010; 7:179-90. [PMID: 20200494 DOI: 10.4161/rna.7.2.11182] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The discovery of increasing numbers of genes with overlapping sequences highlights the problem of expression in the context of constraining regulatory elements from more than one gene. This study identifies regulatory sequences encompassed within two genes that overlap in an antisense orientation at their 3' ends. The genes encode the alpha-thyroid hormone receptor gene (TRalpha or NR1A1) and Rev-erbalpha (NR1D1). In mammals TRalpha pre-mRNAs are alternatively spliced to yield mRNAs encoding functionally antagonistic proteins: TRalpha1, an authentic thyroid hormone receptor; and TRalpha2, a non-hormone-binding variant that acts as a repressor. TRalpha2-specific splicing requires two regulatory elements that overlap with Rev-erbalpha sequences. Functional mapping of these elements reveals minimal splicing enhancer elements that have evolved within the constraints of the overlapping Rev-erbalpha sequence. These results provide insight into the evolution of regulatory elements within the context of bidirectional coding sequences. They also demonstrate the ability of the genetic code to accommodate multiple layers of information within a given sequence, an important property of the code recently suggested on theoretical grounds.
Collapse
Affiliation(s)
- Valerie K Salato
- Department of Biological Sciences, Marquette University, Milwaukee, WI, USA
| | | | | | | | | |
Collapse
|
38
|
Song G, Wang L. A conserved gene structure and expression regulation of miR-433 and miR-127 in mammals. PLoS One 2009; 4:e7829. [PMID: 19946636 PMCID: PMC2778354 DOI: 10.1371/journal.pone.0007829] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2009] [Accepted: 10/19/2009] [Indexed: 11/19/2022] Open
Abstract
MicroRNAs play essential roles in many cellular processes. However, limited information is available regarding the gene structure and transcriptional regulation of miRNAs. We explored the gene cluster encoding miR-433/127 in mammalian species using bioinformatics and in vitro "gene" expression approaches. Multiple sequence alignments (MSA) showed that the precursors of miR-433 and of miR-127 exhibited 95% and 100% similarity, respectively, in human, chimpanzee, horse, dog, monkey, rat, cow, and mouse. MSA of the promoter sequences of miR-433 and of miR-127 revealed lower sequence similarity among these mammalian species. However, the distance between miR-433 and miR-127 was strikingly similar, which was between 986 and 1007 bp and the position of transcription factor (TF) binding motifs, including estrogen related receptor response element (ERRE), was well conserved. Transient transfection assays showed that promoters of miR-433 and of miR-127 from human, rat, and dog were activated by estrogen related receptor gamma (ERRgamma) and inhibited by small heterodimer partner (SHP). ChIP assays confirmed the physical association of ERRgamma with the endogenous promoters of miR-433 and miR-127. In vitro over-expression of the human, rat, or dog miR-433/127 loci in cells, using an expression vector containing miR-433/127 and their promoter regions, markedly induced a differential expression of both primary and mature miR-433 and miR-127, indicating that miR-433 and miR-127 were possessed from their independent promoters. Our studies for the first time demonstrate a conserved gene structure and transcriptional regulation of miR-433 and miR-127 in mammals. The data suggest that the miR-433/127 loci may have evolved from a common gene of origin.
Collapse
Affiliation(s)
- Guisheng Song
- Departments of Medicine and Oncological Sciences, Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Li Wang
- Departments of Medicine and Oncological Sciences, Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
- * E-mail:
| |
Collapse
|
39
|
Vasu VT, Cross CE, Gohil K. Nr1d1, an important circadian pathway regulatory gene, is suppressed by cigarette smoke in murine lungs. Integr Cancer Ther 2009; 8:321-8. [PMID: 19926613 DOI: 10.1177/1534735409352027] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Nuclear receptor subfamily 1, group D member 1 (Nr1d1), also known as Rev-erb-alpha, belongs to the family of "orphan receptors" and functions as a member of clock gene family. In addition to being an important member of clock circuitry, Nr1d1, also regulates cell proliferation, lipid metabolism, and inflammation and is also touted as a tumor suppressor. Our focus on Nr1d1 was stimulated by data from a genome-wide search for mRNA correlates of cigarette smoke (CS) sensitive--whole smoke (WS) and filtered smoke (FS)--lung transcriptomes in tumor-resistant C57BL6 and tumor-susceptible AJ mice strains. Differential analysis of approximately 15,000 genes using Affymetrix 430A 2.0 high-density oligonucleotide arrays identified modulation of genes related to circadian pathways by CS in lungs of both mouse strains. Nr1d1 expression was downregulated by both WS and FS irrespective of mouse strain as compared to respective air-breathing controls. WS was more effective than FS on decreasing Nr1d1 expression. The present data suggest that transcriptional regulation of Nr1d1 by CS may affect circadian rhythmicity and thus may play a complementary role in CS-induced lung respiratory tract pathobiology and/or lung tumorigenesis.
Collapse
Affiliation(s)
- Vihas T Vasu
- Department of Internal Medicine, Centre for Comparative Respiratory Biology and Medicine, University of California, Davis, CA 95616, USA.
| | | | | |
Collapse
|
40
|
Marvin KA, Reinking JL, Lee AJ, Pardee K, Krause HM, Burstyn JN. Nuclear receptors homo sapiens Rev-erbbeta and Drosophila melanogaster E75 are thiolate-ligated heme proteins which undergo redox-mediated ligand switching and bind CO and NO. Biochemistry 2009; 48:7056-71. [PMID: 19405475 DOI: 10.1021/bi900697c] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Nuclear receptors E75, which regulates development in Drosophila melanogaster, and Rev-erbbeta, which regulates circadian rhythm in humans, bind heme within their ligand binding domains (LBD). The heme-bound ligand binding domains of E75 and Rev-erbbeta were studied using electronic absorption, MCD, resonance Raman, and EPR spectroscopies. Both proteins undergo redox-dependent ligand switching and CO- and NO-induced ligand displacement. In the Fe(III) oxidation state, the nuclear receptor hemes are low spin and 6-coordinate with cysteine(thiolate) as one of the two axial heme ligands. The sixth ligand is a neutral donor, presumably histidine. When the heme is reduced to the Fe(II) oxidation state, the cysteine(thiolate) is replaced by a different neutral donor ligand, whose identity is not known. CO binds to the Fe(II) heme in both E75(LBD) and Rev-erbbeta(LBD) opposite a sixth neutral ligand, plausibly the same histidine that served as the sixth ligand in the Fe(III) state. NO binds to the heme of both proteins; however, the NO-heme is 5-coordinate in E75 and 6-coordinate in Rev-erbbeta. These nuclear receptors exhibit coordination characteristics that are similar to other known redox and gas sensors, suggesting that E75 and Rev-erbbeta may function in heme-, redox-, or gas-regulated control of cellular function.
Collapse
Affiliation(s)
- Katherine A Marvin
- Department of Chemistry, University of Wisconsin-Madison, 1101 University Avenue, Madison, Wisconsin 53706, USA
| | | | | | | | | | | |
Collapse
|
41
|
Rambaud J, Triqueneaux G, Masse I, Staels B, Laudet V, Benoit G. Rev-erbalpha2 mRNA encodes a stable protein with a potential role in circadian clock regulation. Mol Endocrinol 2009; 23:630-9. [PMID: 19228794 DOI: 10.1210/me.2008-0395] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Circadian rhythms are observed in nearly all aspects of physiology and behavior. In mammals, such biological rhythms are supported by a complex network of self-sustained transcriptional loops and posttranslational modifications, which regulate timely controlled production and degradation of critical factors on a 24-h basis. Among these factors, the orphan nuclear receptor rev-erbalpha plays an essential role by linking together positive and negative regulatory loops. As an essential part of the circadian core clock mechanism, REV-ERBalpha expression shows a precisely scheduled oscillation reflecting the tight control of its production and degradation. In previous studies, we identified two alternative transcripts encoding two protein variants referred to as REV-ERBalpha1 and -alpha2. Interestingly, recent work identified structural elements present only in REV-ERBalpha1 that controls its turnover and thereby influences circadian oscillations. In the present work, we comparatively analyze the two variants and show that REV-ERBalpha2 exhibits a half-life incompatible with a circadian function, suggesting that this variant exerts different biological functions. However, our comparative study clearly indicates undistinguishable DNA-binding properties and transcriptional repression activity as well as a similar regulation mechanism. The only consistent difference appears to be the relative expression level of the two transcripts, rev-erbalpha1 being one to 100 times more expressed than alpha2 depending on tissue and circadian time. Taking this finding into consideration, we reassessed REV-ERBalpha2 turnover and were able to show that this variant exhibits a reduced half-life when coexpressed with REV-ERBalpha1. We propose that the relative expression levels of the two REV-ERBalpha variants fine-tune the circadian period length by regulating REV-ERBalpha half-life.
Collapse
Affiliation(s)
- Juliette Rambaud
- Molecular Zoology Group, Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, CNRS, INRA, Université Claude Bernard Lyon 1, Ecole Normale Supérieure de Lyon, Lyon, France
| | | | | | | | | | | |
Collapse
|
42
|
Teboul M, Guillaumond F, Gréchez-Cassiau A, Delaunay F. The nuclear hormone receptor family round the clock. Mol Endocrinol 2008; 22:2573-82. [PMID: 18653780 DOI: 10.1210/me.2007-0521] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Daily rhythms in behavior and physiology are observed in most organisms. These rhythms are controlled by internal self-sustained circadian ( approximately 24 h) clocks, which are present in virtually all cells. The 24-h oscillations are generated by a molecular mechanism entrained by external or internal time cues and which, in turn, regulate rhythmic outputs. In mammals, the circadian system comprises a master clock located in the hypothalamus that is directly entrained by the light-dark cycle and which coordinates the phases of local clocks in the periphery in order to ensure optimal timing of the physiology. Nuclear receptors (NRs) form a large family of transcription factors that include both ligand-inducible and orphan receptors. These NRs are key regulators of major biological processes such as reproduction, development, cell growth and death, inflammation, immunity, and metabolic homeostasis. Recent observations indicate that several NR signaling pathways play a critical role in central and peripheral circadian clocks. The REV-ERB/retinoid-related orphan receptor orphan NR subfamily regulates the expression of core clock genes and contributes to the robustness of the clock mechanism. Glucocorticoid and retinoic acid receptors are involved in the resetting of peripheral clocks. Several other NRs such as peroxisome proliferator-activated receptor-alpha, short heterodimer partner, and constitutive androstane receptor act as molecular links between clock genes and specific rhythmic metabolic outputs. The expanding functional links between NRs and circadian clocks open novel perspectives for understanding the hormonal regulation of the mammalian circadian system as well as for exploring the role of circadian clocks in the pathogenesis of NR-related diseases such as cancer and metabolic syndrome.
Collapse
Affiliation(s)
- Michèle Teboul
- Laboratoire de Biologie et Physiopathologie des Systèmes Intégrés, Université de Nice Sophia Antipolis, Centre National de la Recherche Scientifique, Nice, France
| | | | | | | |
Collapse
|
43
|
Fontaine C, Rigamonti E, Pourcet B, Duez H, Duhem C, Fruchart JC, Chinetti-Gbaguidi G, Staels B. The nuclear receptor Rev-erbalpha is a liver X receptor (LXR) target gene driving a negative feedback loop on select LXR-induced pathways in human macrophages. Mol Endocrinol 2008; 22:1797-811. [PMID: 18511497 DOI: 10.1210/me.2007-0439] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
A role of the nuclear receptor Rev-erbalpha in the regulation of transcription pathways involving other nuclear receptors is emerging. Indeed, Rev-erbalpha is a negative regulator of transcription by binding to overlapping response elements shared with various nuclear receptors, including the peroxisome proliferator-activated receptors and the retinoid-related orphan receptor alpha (RORalpha). Here, we show that Rev-erbalpha is expressed in primary human macrophages and that its expression is induced by synthetic ligands for the liver X receptors (LXRs), which control cholesterol homeostasis, inflammation, and the immune response in macrophages. LXRalpha binds to a specific response element in the human Rev-erbalpha promoter, thus inducing Rev-erbalpha transcriptional expression. Interestingly, Rev-erbalpha does not influence basal or LXR-regulated cholesterol homeostasis. However, Rev-erbalpha overexpression represses the induction of toll-like receptor (TLR)-4 by LXR agonists, whereas Rev-erbalpha silencing by short interfering RNA results in enhanced TLR-4 expression upon LXR activation. Electrophoretic mobility shift, chromatin immunoprecipitation, and transient transfection experiments demonstrate that Rev-erbalpha represses human TLR-4 promoter activity by binding as a monomer to a RevRE site overlapping with the LXR response element site in the TLR-4 promoter. These data identify Rev-erbalpha as a new LXR target gene, inhibiting LXR-induction of TLR-4 in a negative transcriptional feedback loop, but not cholesterol homeostasis gene expression.
Collapse
Affiliation(s)
- Coralie Fontaine
- Institut National de la Santé et de la Recherche Médicale, Unité 545, Institut Pasteur de Lille, 1, rue du Professeur Calmette, Boite Postale 245, Lille 59019, France
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Wang J, Lazar MA. Bifunctional role of Rev-erbalpha in adipocyte differentiation. Mol Cell Biol 2008; 28:2213-20. [PMID: 18227153 PMCID: PMC2268425 DOI: 10.1128/mcb.01608-07] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2007] [Revised: 10/09/2007] [Accepted: 01/20/2008] [Indexed: 12/22/2022] Open
Abstract
The nuclear receptor Rev-erbalpha is a potent transcriptional repressor that regulates circadian rhythm and metabolism. Here we demonstrate a dissociation between Rev-erbalpha mRNA and protein levels that profoundly influences adipocyte differentiation. During adipogenesis, Rev-erbalpha gene expression initially declines and subsequently increases. Remarkably, Rev-erbalpha protein levels are nearly the opposite, increasing early in adipogenesis and then markedly decreasing in adipocytes. The Rev-erbalpha protein is necessary for the early mitotic events that are required for adipogenesis. The subsequent reduction in Rev-erbalpha protein, due to increased degradation via the 26S proteasome, is also required for adipocyte differentiation because Rev-erbalpha represses the expression of PPARgamma2, the master transcriptional regulator of adipogenesis. Thus, opposite to what might be predicted from Rev-erbalpha gene expression, Rev-erbalpha protein levels must rise and then fall for adipocyte differentiation to occur.
Collapse
Affiliation(s)
- Jing Wang
- University of Pennsylvania School of Medicine, 700 CRB, 415 Curie Blvd., Philadelphia, PA 19104-6149, USA
| | | |
Collapse
|
45
|
Hummasti S, Tontonoz P. Adopting new orphans into the family of metabolic regulators. Mol Endocrinol 2008; 22:1743-53. [PMID: 18258684 DOI: 10.1210/me.2007-0566] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The importance of the adopted metabolite receptors, such as peroxisome proliferator-activated receptor, liver X receptor, and farnesoid X receptor, in transcriptional control of metabolic pathways has been appreciated for many years. However, it is becoming increasingly clear that the number of nuclear receptors with roles in metabolism is much larger than initially suspected. Recent years have brought an intense effort to define the biological functions of the most enigmatic group of the nuclear receptor superfamily, the true orphan receptors, including nuclear receptor 4As, estrogen-related receptors, retinoid-related orphan receptors, and Rev-erbs. Unexpectedly, several of these receptors also turn out to have important functions in various aspects of metabolic control.
Collapse
Affiliation(s)
- Sarah Hummasti
- Howard Hughes Medical Institute, University of California Los Angeles School of Medicine, Box 951662, Los Angeles, California 90095-1662, USA
| | | |
Collapse
|
46
|
Burris TP. Nuclear hormone receptors for heme: REV-ERBalpha and REV-ERBbeta are ligand-regulated components of the mammalian clock. Mol Endocrinol 2008; 22:1509-20. [PMID: 18218725 DOI: 10.1210/me.2007-0519] [Citation(s) in RCA: 125] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The nuclear hormone receptors (NHRs), REV-ERBalpha and REV-ERBbeta, regulate a number of physiological functions including the circadian rhythm, lipid metabolism, and cellular differentiation. These two receptors lack the activation function-2 region that is associated with the ability of NHRs to recruit coactivators and activate target gene transcription. These NHRs have been characterized as constitutive repressors of transcription due to their lack of an identified ligand and their strong ability to recruit the corepressor, nuclear receptor corepressor. Recently, the porphyrin heme was demonstrated to function as a ligand for both REV-ERBs. Heme binds directly to the ligand-binding domain and regulates the ability of these NHRs to recruit nuclear receptor corepressor to target gene promoters. This review focuses on the physiological roles that these two receptors play and the implications of heme functioning as their ligand. The prospect that these NHRs, now known to be regulated by small molecule ligands, may be targets for development of drugs for treatment of diseases associated with aberrant circadian rhythms including metabolic and psychiatric disorders as well as cancer is also addressed.
Collapse
Affiliation(s)
- Thomas P Burris
- Nuclear Receptor Biology Laboratory, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA 70808, USA.
| |
Collapse
|
47
|
Identification of heme as the ligand for the orphan nuclear receptors REV-ERBalpha and REV-ERBbeta. Nat Struct Mol Biol 2007; 14:1207-13. [PMID: 18037887 DOI: 10.1038/nsmb1344] [Citation(s) in RCA: 444] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2007] [Accepted: 10/30/2007] [Indexed: 11/08/2022]
Abstract
The nuclear receptors REV-ERBalpha (encoded by NR1D1) and REV-ERBbeta (NR1D2) have remained orphans owing to the lack of identified physiological ligands. Here we show that heme is a physiological ligand of both receptors. Heme associates with the ligand-binding domains of the REV-ERB receptors with a 1:1 stoichiometry and enhances the thermal stability of the proteins. Results from experiments of heme depletion in mammalian cells indicate that heme binding to REV-ERB causes the recruitment of the co-repressor NCoR, leading to repression of target genes including BMAL1 (official symbol ARNTL), an essential component of the circadian oscillator. Heme extends the known types of ligands used by the human nuclear receptor family beyond the endocrine hormones and dietary lipids described so far. Our results further indicate that heme regulation of REV-ERBs may link the control of metabolism and the mammalian clock.
Collapse
|
48
|
Ramakrishnan SN, Muscat GE. The orphan Rev-erb nuclear receptors: a link between metabolism, circadian rhythm and inflammation? NUCLEAR RECEPTOR SIGNALING 2006; 4:e009. [PMID: 16741567 PMCID: PMC1472670 DOI: 10.1621/nrs.04009] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/03/2005] [Accepted: 03/10/2006] [Indexed: 12/12/2022]
Abstract
Nuclear hormone receptors (NRs) function as ligand dependent DNA binding proteins that translate physiological/nutritional signals into gene regulation. Dysfunctional NR signaling leads to many disorders in reproduction, inflammation, and metabolism. The opportunity to identify novel regulatory pathways in the context of human health and disease drives the challenge to unravel the biological function of the “orphan nuclear hormone receptors”. For example, the Rev-erb (NR1D) subgroup (Rev-erbα/NR1D1 and Rev-erbβ/NR1D2) of orphan NRs are transcriptional silencers and negative regulators of RORα mediated trans-activation. The NR1D subgroup is highly enriched in peripheral tissues with onerous energy demands including skeletal muscle, brown and white adipose, brain, liver and kidney. This alludes to the involvement of this subgroup in metabolism. In this context, Rev-erbα-/- mice have a dyslipidemic phenotype. Recent studies in vascular smooth and skeletal muscle cells also suggest that the NR1D subgroup modulates inflammation by regulating IκBα/NFκB dependent gene expression. Rev-erbα has been identified as a critical regulator (and target) of circadian rhythm, a factor in blood pressure control and inflammation. Finally, two recent reports have demonstrated: (i) lithium mediated regulation of Rev-erbα stability and (ii) E75 (the Drosophila orthologue of human Rev-erbα) is tightly bound by heme, and functions as a “gas sensor” through interaction with CO/NO and interferes with the repression of DHR3 (the Drosophila orthologue of human RORα). In conclusion, the role of these receptors at the cross-roads of metabolism, inflammation, and circadian cycling underscores the importance of understanding the organ-specific function of the NR1D subgroup in homeostasis.
Collapse
|
49
|
Li YY, Qin L, Guo ZM, Liu L, Xu H, Hao P, Su J, Shi Y, He WZ, Li YX. In silico discovery of human natural antisense transcripts. BMC Bioinformatics 2006; 7:18. [PMID: 16409644 PMCID: PMC1369008 DOI: 10.1186/1471-2105-7-18] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2005] [Accepted: 01/13/2006] [Indexed: 12/03/2022] Open
Abstract
Background Several high-throughput searches for ppotential natural antisense transcripts (NATs) have been performed recently, but most of the reports were focused on cis type. A thorough in silico analysis of human transcripts will help expand our knowledge of NATs. Results We have identified 568 NATs from human RefSeq RNA sequences. Among them, 403 NATs are reported for the first time, and at least 157 novel NATs are trans type. According to the pairing region of a sense and antisense RNA pair, hNATs are divided into 6 classes, of which about 87% involve 5' or 3' UTR sequences, supporting the regulatory role of UTRs. Among a total of 535 NAT pairs related with splice variants, 77.4% (414/535) have their pairing regions affected or completely eliminated by alternative splicing, suggesting significant relationship of alternative splicing and antisense-directed regulation. The extensive occurrence of splice variants in hNATs and other multiple pairing patterns results in a one-to-many relationship, allowing the formation of complex regulation networks. Based on microarray data from Stanford Microarray Database, two hNAT pairs were found to display significant inverse expression patterns before and after insulin injection. Conclusion NATs might carry out more extensive and complex functions than previously thought. Combined with endogenous micro RNAs, hNATs could be regarded as a special group of transcripts contributing to the complex regulation networks.
Collapse
Affiliation(s)
- Yuan-Yuan Li
- Shanghai Center for Bioinformation Technology, Shanghai 200235, China
| | - Lei Qin
- Shanghai Center for Bioinformation Technology, Shanghai 200235, China
| | - Zong-Ming Guo
- Shanghai Center for Bioinformation Technology, Shanghai 200235, China
| | - Lei Liu
- The W. M. Keck Center for Comparative and Functional Genomics, University of Illinois at Urbana-Champaign, 1201 W. Gregory Dr., Urbana, Illinois 61801, USA
| | - Hao Xu
- Shanghai Center for Bioinformation Technology, Shanghai 200235, China
| | - Pei Hao
- Shanghai Center for Bioinformation Technology, Shanghai 200235, China
| | - Jiong Su
- Shanghai Center for Bioinformation Technology, Shanghai 200235, China
| | - Yixiang Shi
- Shanghai Center for Bioinformation Technology, Shanghai 200235, China
| | - Wei-Zhong He
- Shanghai Center for Bioinformation Technology, Shanghai 200235, China
| | - Yi-Xue Li
- Shanghai Center for Bioinformation Technology, Shanghai 200235, China
| |
Collapse
|
50
|
Triqueneaux G, Thenot S, Kakizawa T, Antoch MP, Safi R, Takahashi JS, Delaunay F, Laudet V. The orphan receptor Rev-erbalpha gene is a target of the circadian clock pacemaker. J Mol Endocrinol 2004; 33:585-608. [PMID: 15591021 PMCID: PMC3770723 DOI: 10.1677/jme.1.01554] [Citation(s) in RCA: 127] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Rev-erbalpha is a ubiquitously expressed orphan nuclear receptor which functions as a constitutive transcriptional repressor and is expressed in vertebrates according to a robust circadian rhythm. We report here that two Rev-erbalpha mRNA isoforms, namely Rev-erbalpha1 and Rev-erbalpha 2, are generated through alternative promoter usage and that both show a circadian expression pattern in an in vitro system using serum-shocked fibroblasts. Both promoter regions P1 (Rev-erbalpha1) and P2 (Rev-erbalpha2) contain several E-box DNA sequences which function as response elements for the core circadian-clock components: CLOCK and BMAL1. The CLOCK-BMAL1 heterodimer stimulates the activity of both P1 and P2 promoters in transient transfection assay by 3-6-fold. This activation was inhibited by the overexpression of CRY1, a component of the negative limb of the circadian transcriptional loop. Critical E-box elements were mapped within both promoters. This regulation is conserved in vertebrates since we found that the CLOCK-BMAL1 heterodimer also regulates the zebrafish Rev-erbalpha gene. In line with these data Rev-erbalpha circadian expression was strongly impaired in the livers of Clock mutant mice and in the pineal glands of zebrafish embryos treated with Clock and Bmal1 antisense oligonucleotides. Together these data demonstrate that CLOCK is a critical regulator of Rev-erbalpha circadian gene expression in evolutionarily distant vertebrates and suggest a role for Rev-erbalpha in the circadian clock output.
Collapse
Affiliation(s)
- Gérard Triqueneaux
- Laboratoire de Biologie Moléculaire et Cellulaire, CNRS UMR 5161, Ecole Normale Supérieur de Lyon, 46 allée d'Italie, 69364 Lyon cedex, France
| | | | | | | | | | | | | | | |
Collapse
|