1
|
Champsi S, Hood DA. Sulforaphane treatment mimics contractile activity-induced mitochondrial adaptations in muscle myotubes. Am J Physiol Cell Physiol 2025; 328:C335-C354. [PMID: 39672545 DOI: 10.1152/ajpcell.00669.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/15/2024] [Accepted: 12/03/2024] [Indexed: 12/15/2024]
Abstract
Mitochondria are metabolic hubs that govern skeletal muscle health. Although exercise has been established as a powerful inducer of quality control processes that ultimately enhance mitochondrial function, there are currently limited pharmaceutical interventions available that emulate exercise-induced mitochondrial adaptations. To investigate a novel candidate for this role, we examined sulforaphane (SFN), a naturally occurring compound found in cruciferous vegetables. SFN has been documented as a potent antioxidant inducer through its activation of the nuclear factor erythroid 2-related factor 2 (Nrf-2) antioxidant response pathway. However, its effects on muscle health have been underexplored. To investigate the interplay between chronic exercise and SFN, C2C12 myotubes were electrically stimulated to model chronic contractile activity (CCA) in the presence or absence of SFN. SFN promoted Nrf-2 nuclear translocation, enhanced mitochondrial respiration, and upregulated key antioxidant proteins including catalase and glutathione reductase. These adaptations were accompanied by reductions in cellular and mitochondrial reactive oxygen species (ROS) emission. Signaling toward biogenesis was enhanced, demonstrated by increases in mitochondrial transcription factor A (TFAM), peroxisome proliferator-activated receptor-gamma coactivator (PGC)-1α nuclear translocation, PGC-1α promoter activity, mitochondrial content, and organelle branching, suggestive of a larger, more interconnected mitochondrial pool. These mitochondrial adaptations were accompanied by an increase in lysosomal proteins, suggesting coordinated regulation. There was no difference in mitochondrial and antioxidant-related proteins between CCA and non-CCA SFN-treated cells. Our data suggest that SFN activates signaling cascades that are common to those produced by contractile activity, indicating that SFN-centered therapeutic strategies may improve the mitochondrial phenotype in skeletal muscle.NEW & NOTEWORTHY Nrf-2 is a transcription factor that has been implicated in mitigating oxidative stress and regulating mitochondrial homeostasis. However, limited research has demonstrated how Nrf-2-mediated adaptations compare with those produced by exercise. To investigate this, we treated myotubes with Sulforaphane, a well-established Nrf-2 activator, and combined this with stimulation-induced chronic contractile activity to model exercise training. Our work is the first to establish that sulforaphane mimics training-induced mitochondrial adaptations, including enhancements in respiration, biogenesis, and dynamics.
Collapse
Affiliation(s)
- Sabrina Champsi
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| | - David A Hood
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| |
Collapse
|
2
|
Chen J, Wu Y, Hao W, You J, Wu L. Non-canonical hepatic androgen receptor mediates glucagon sensitivity in female mice through the PGC1α/ERRα/mitochondria axis. Cell Rep 2025; 44:115188. [PMID: 39792556 DOI: 10.1016/j.celrep.2024.115188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 10/27/2024] [Accepted: 12/19/2024] [Indexed: 01/12/2025] Open
Abstract
Glucagon has recently been found to modulate liver fat content, in addition to its role in regulating gluconeogenesis. However, the precise mechanisms by which glucagon signaling synchronizes glucose and lipid metabolism in the liver remain poorly understood. By employing chemical and genetic approaches, we demonstrate that inhibiting the androgen receptor (AR) impairs the ability of glucagon to stimulate gluconeogenesis and lipid catabolism in primary hepatocytes and female mice. Notably, AR expression in the liver of female mice is up to three times higher than that in their male littermates, accounting for the more pronounced response to glucagon in females. Mechanistically, hepatic AR promotes energy metabolism and enhances lipid breakdown for liver glucose production in response to glucagon treatment through the peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α)/estrogen-related receptor alpha (ERRα)-mitochondria axis. Overall, our findings highlight the crucial role of hepatic AR in mediating glucagon signaling and the sexual dimorphism in hepatic glucagon sensitivity.
Collapse
Affiliation(s)
- Jie Chen
- Fudan University, Shanghai, China; Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China; Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China
| | - Yuanyuan Wu
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Wenyuan Secondary School Affiliated to Xuejun High School, Hangzhou, Zhejiang, China
| | - Wanyu Hao
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Westlake Pharmaceuticals, Hangzhou, Zhejiang, China
| | - Jia You
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China; Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China.
| | - Lianfeng Wu
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China; Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China; Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China.
| |
Collapse
|
3
|
Rambout X, Maquat LE. Nuclear mRNA decay: regulatory networks that control gene expression. Nat Rev Genet 2024; 25:679-697. [PMID: 38637632 PMCID: PMC11408106 DOI: 10.1038/s41576-024-00712-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/19/2024] [Indexed: 04/20/2024]
Abstract
Proper regulation of mRNA production in the nucleus is critical for the maintenance of cellular homoeostasis during adaptation to internal and environmental cues. Over the past 25 years, it has become clear that the nuclear machineries governing gene transcription, pre-mRNA processing, pre-mRNA and mRNA decay, and mRNA export to the cytoplasm are inextricably linked to control the quality and quantity of mRNAs available for translation. More recently, an ever-expanding diversity of new mechanisms by which nuclear RNA decay factors finely tune the expression of protein-encoding genes have been uncovered. Here, we review the current understanding of how mammalian cells shape their protein-encoding potential by regulating the decay of pre-mRNAs and mRNAs in the nucleus.
Collapse
Affiliation(s)
- Xavier Rambout
- Department of Biochemistry and Biophysics, School of Medicine and Dentistry, University of Rochester, Rochester, NY, USA.
- Center for RNA Biology, University of Rochester, Rochester, NY, USA.
| | - Lynne E Maquat
- Department of Biochemistry and Biophysics, School of Medicine and Dentistry, University of Rochester, Rochester, NY, USA.
- Center for RNA Biology, University of Rochester, Rochester, NY, USA.
| |
Collapse
|
4
|
Tian Z, Li J, Tang H, Liu W, Hou H, Wang C, Li D, Chen G, Xia T, Wang A. ZLN005 alleviates PBDE-47 induced impairment of mitochondrial translation and neurotoxicity through PGC-1α/ERRα axis. JOURNAL OF HAZARDOUS MATERIALS 2024; 471:134331. [PMID: 38677116 DOI: 10.1016/j.jhazmat.2024.134331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 03/28/2024] [Accepted: 04/15/2024] [Indexed: 04/29/2024]
Abstract
Recent studies are identified the mitochondria as critical targets of 2, 2', 4, 4'-tetrabromodiphenyl ether (PBDE-47) induced neurotoxicity. This study aimed at examining the impact of PBDE-47 exposure on mitochondrial translation, and its subsequent effect on PBDE-47 neurotoxicity. The Sprague-Dawley (SD) rat model and neuroendocrine pheochromocytoma (PC12) cells were adopted for the measurements of mitochondrial ATP levels, mitochondrial translation products, and expressions of important mitochondrial regulators, such as required meiotic nuclear division 1 (RMND1), estrogen-related receptor α (ERRα), and peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC-1α). To delve into the role of PGC-1α/ERRα axis in mitochondrial translation, 2-(4-tert-butylphenyl) benzimidazole (ZLN005) was employed. Both cellular and animal model results shown that PBDE-47 impeded PGC-1α/ERRα axis and mitochondrial translation. PBDE-47 suppressed mitochondrial function in rat hippocampus and PC12 cells by decreasing relative mitochondrial DNA (mtDNA) content, mitochondrial translation products, and mitochondrial ATP levels. Particularly, ZLN005 reversed PBDE-47 neurotoxicity by enhancing mitochondrial translation through activation of PGC-1α/ERRα axis, yet suppressing PGC-1α with siRNA attenuates its neuroprotective effect in vitro. In conclusion, this work highlights the importance of mitochondrial translation in PBDE-47 neurotoxicity by presenting results from cellular and animal models and suggests a potential therapeutic approach through activation of PGC-1α/ERRα axis. ENVIRONMENTAL IMPLICATION: PBDEs have attracted extensive attention because of their high lipophilicity, persistence, and detection levels in various environmental media. Increasing evidence has shown that neurodevelopmental disorders in children are associated with PBDE exposure. Several studies have also found that perinatal PBDE exposure can cause long-lasting neurobehavioral abnormalities in experimental animals. Our recent studies have also demonstrated the impact of PBDE-47 exposure on mitochondrial biogenesis and dynamics, leading to memory and neurobehavioral deficits. Therefore, we explore whether the pathological mechanism of PBDE-47-induced neurotoxicity involves the regulation of mitochondrial translation through the PGC-1α/ERRα axis.
Collapse
Affiliation(s)
- Zhiyuan Tian
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Jing Li
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Huayang Tang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Wenhui Liu
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Haoqi Hou
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Chenxi Wang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Dongjie Li
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Gaoshuai Chen
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Tao Xia
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China.
| | - Aiguo Wang
- Department of Occupational and Environmental Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China; Key Laboratory of Environment and Health, Ministry of Education & Ministry of Environmental Protection, State Key Laboratory of Environmental health (incubating), School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China.
| |
Collapse
|
5
|
de Smalen LM, Börsch A, Leuchtmann AB, Gill JF, Ritz D, Zavolan M, Handschin C. Impaired age-associated mitochondrial translation is mitigated by exercise and PGC-1α. Proc Natl Acad Sci U S A 2023; 120:e2302360120. [PMID: 37639610 PMCID: PMC10483666 DOI: 10.1073/pnas.2302360120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 07/24/2023] [Indexed: 08/31/2023] Open
Abstract
Sarcopenia, the age-related loss of skeletal muscle mass and function, can dramatically impinge on quality of life and mortality. While mitochondrial dysfunction and imbalanced proteostasis are recognized as hallmarks of sarcopenia, the regulatory and functional link between these processes is underappreciated and unresolved. We therefore investigated how mitochondrial proteostasis, a crucial process that coordinates the expression of nuclear- and mitochondrial-encoded mitochondrial proteins with supercomplex formation and respiratory activity, is affected in skeletal muscle aging. Intriguingly, a robust mitochondrial translation impairment was observed in sarcopenic muscle, which is regulated by the peroxisome proliferator-activated receptor γ coactivator 1 α (PGC-1α) with the estrogen-related receptor α (ERRα). Exercise, a potent inducer of PGC-1α activity, rectifies age-related reduction in mitochondrial translation, in conjunction with quality control pathways. These results highlight the importance of mitochondrial proteostasis in muscle aging, and elucidate regulatory interactions that underlie the powerful benefits of physical activity in this context.
Collapse
Affiliation(s)
| | | | | | | | - Danilo Ritz
- Biozentrum, University of Basel, BaselCH-4056, Switzerland
| | | | | |
Collapse
|
6
|
Billon C, Sitaula S, Banerjee S, Welch R, Elgendy B, Hegazy L, Oh TG, Kazantzis M, Chatterjee A, Chrivia J, Hayes ME, Xu W, Hamilton A, Huss JM, Zhang L, Walker JK, Downes M, Evans RM, Burris TP. Synthetic ERRα/β/γ Agonist Induces an ERRα-Dependent Acute Aerobic Exercise Response and Enhances Exercise Capacity. ACS Chem Biol 2023; 18:756-771. [PMID: 36988910 PMCID: PMC11584170 DOI: 10.1021/acschembio.2c00720] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Repetitive physical exercise induces physiological adaptations in skeletal muscle that improves exercise performance and is effective for the prevention and treatment of several diseases. Genetic evidence indicates that the orphan nuclear receptors estrogen receptor-related receptors (ERRs) play an important role in skeletal muscle exercise capacity. Three ERR subtypes exist (ERRα, β, and γ), and although ERRβ/γ agonists have been designed, there have been significant difficulties in designing compounds with ERRα agonist activity. Additionally, there are limited synthetic agonists that can be used to target ERRs in vivo. Here, we report the identification of a synthetic ERR pan agonist, SLU-PP-332, that targets all three ERRs but has the highest potency for ERRα. Additionally, SLU-PP-332 has sufficient pharmacokinetic properties to be used as an in vivo chemical tool. SLU-PP-332 increases mitochondrial function and cellular respiration in a skeletal muscle cell line. When administered to mice, SLU-PP-332 increased the type IIa oxidative skeletal muscle fibers and enhanced exercise endurance. We also observed that SLU-PP-332 induced an ERRα-specific acute aerobic exercise genetic program, and the ERRα activation was critical for enhancing exercise endurance in mice. These data indicate the feasibility of targeting ERRα for the development of compounds that act as exercise mimetics that may be effective in the treatment of numerous metabolic disorders and to improve muscle function in the aging.
Collapse
Affiliation(s)
- Cyrielle Billon
- Center for Clinical Pharmacology, Washington University School of Medicine and St. Louis College of Pharmacy, St. Louis, Missouri 63110, United States
| | - Sadichha Sitaula
- Center for Clinical Pharmacology, Washington University School of Medicine and St. Louis College of Pharmacy, St. Louis, Missouri 63110, United States
| | - Subhashis Banerjee
- Department of Pharmacology & Physiology, Saint Louis University School of Medicine, St. Louis, Missouri 63104, United States
| | - Ryan Welch
- Gene Expression Laboratory Salk Institute for Biological Studies, La Jolla, California 92037, United States
| | - Bahaa Elgendy
- Center for Clinical Pharmacology, Washington University School of Medicine and St. Louis College of Pharmacy, St. Louis, Missouri 63110, United States
| | - Lamees Hegazy
- Center for Clinical Pharmacology, Washington University School of Medicine and St. Louis College of Pharmacy, St. Louis, Missouri 63110, United States
| | - Tae Gyu Oh
- Gene Expression Laboratory Salk Institute for Biological Studies, La Jolla, California 92037, United States
| | - Melissa Kazantzis
- The Scripps Research Institute Jupiter, Jupiter, Florida 33458, United States
| | - Arindam Chatterjee
- Department of Pharmacology & Physiology, Saint Louis University School of Medicine, St. Louis, Missouri 63104, United States
| | - John Chrivia
- Department of Pharmacology & Physiology, Saint Louis University School of Medicine, St. Louis, Missouri 63104, United States
| | - Matthew E Hayes
- University of Florida Genetics Institute, Gainesville, Florida 32610, United States
| | - Weiyi Xu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Angelica Hamilton
- Department of Molecular & Cellular Endocrinology, City of Hope, Duarte, California 91010, United States
| | - Janice M Huss
- Department of Molecular & Cellular Endocrinology, City of Hope, Duarte, California 91010, United States
| | - Lilei Zhang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, United States
| | - John K Walker
- Department of Pharmacology & Physiology, Saint Louis University School of Medicine, St. Louis, Missouri 63104, United States
- Department of Chemistry, Saint Louis University, St. Louis, Missouri 63103, United States
| | - Michael Downes
- Gene Expression Laboratory Salk Institute for Biological Studies, La Jolla, California 92037, United States
| | - Ronald M Evans
- Gene Expression Laboratory Salk Institute for Biological Studies, La Jolla, California 92037, United States
- Howard Hughes Medical Institute, Salk Institute for Biological Studies, La Jolla, California 92037, United States
| | - Thomas P Burris
- Center for Clinical Pharmacology, Washington University School of Medicine and St. Louis College of Pharmacy, St. Louis, Missouri 63110, United States
- University of Florida Genetics Institute, Gainesville, Florida 32610, United States
| |
Collapse
|
7
|
Cerutti C, Shi JR, Vanacker JM. Multifaceted Transcriptional Network of Estrogen-Related Receptor Alpha in Health and Disease. Int J Mol Sci 2023; 24:ijms24054265. [PMID: 36901694 PMCID: PMC10002233 DOI: 10.3390/ijms24054265] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/15/2023] [Accepted: 02/18/2023] [Indexed: 02/23/2023] Open
Abstract
Estrogen-related receptors (ERRα, β and γ in mammals) are orphan members of the nuclear receptor superfamily acting as transcription factors. ERRs are expressed in several cell types and they display various functions in normal and pathological contexts. Amongst others, they are notably involved in bone homeostasis, energy metabolism and cancer progression. In contrast to other nuclear receptors, the activities of the ERRs are apparently not controlled by a natural ligand but they rely on other means such as the availability of transcriptional co-regulators. Here we focus on ERRα and review the variety of co-regulators that have been identified by various means for this receptor and their reported target genes. ERRα cooperates with distinct co-regulators to control the expression of distinct sets of target genes. This exemplifies the combinatorial specificity of transcriptional regulation that induces discrete cellular phenotypes depending on the selected coregulator. We finally propose an integrated view of the ERRα transcriptional network.
Collapse
|
8
|
Sopariwala DH, Rios AS, Pei G, Roy A, Tomaz da Silva M, Thi Thu Nguyen H, Saley A, Van Drunen R, Kralli A, Mahan K, Zhao Z, Kumar A, Narkar VA. Innately expressed estrogen-related receptors in the skeletal muscle are indispensable for exercise fitness. FASEB J 2023; 37:e22727. [PMID: 36583689 DOI: 10.1096/fj.202201518r] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 12/01/2022] [Accepted: 12/12/2022] [Indexed: 12/31/2022]
Abstract
Transcriptional determinants in the skeletal muscle that govern exercise capacity, while poorly defined, could provide molecular insights into how exercise improves fitness. Here, we have elucidated the role of nuclear receptors, estrogen-related receptor alpha and gamma (ERRα/γ) in regulating myofibrillar composition, contractility, and exercise capacity in skeletal muscle. We used muscle-specific single or double (DKO) ERRα/γ knockout mice to investigate the effect of ERRα/γ deletion on muscle and exercise parameters. Individual knockout of ERRα/γ did not have a significant impact on the skeletal muscle. On the other hand, DKO mice exhibit pale muscles compared to wild-type (WT) littermates. RNA-seq analysis revealed a predominant decrease in expression of genes linked to mitochondrial and oxidative metabolism in DKO versus WT muscles. DKO muscles exhibit marked repression of oxidative enzymatic capacity, as well as mitochondrial number and size compared to WT muscles. Mitochondrial function is also impaired in single myofibers isolated from DKO versus WT muscles. In addition, mutant muscles exhibit reduced angiogenic gene expression and decreased capillarity. Consequently, DKO mice have a significantly reduced exercise capacity, further reflected in poor fatigue resistance of DKO mice in in vivo contraction assays. These results show that ERRα and ERRγ together are a critical link between muscle aerobic capacity and exercise tolerance. The ERRα/γ mutant mice could be valuable for understanding the long-term impact of impaired mitochondria and vascular supply on the pathogenesis of muscle-linked disorders.
Collapse
Affiliation(s)
- Danesh H Sopariwala
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center, Houston, Texas, USA
| | - Andrea S Rios
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center, Houston, Texas, USA
| | - Guangsheng Pei
- Center for Precision Medicine, School of Biomedical Informatics, The University of Texas Health Science Center, Houston, Texas, USA
| | - Anirban Roy
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, Texas, USA
| | - Meiricris Tomaz da Silva
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, Texas, USA
| | - Hao Thi Thu Nguyen
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center, Houston, Texas, USA
| | - Addison Saley
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center, Houston, Texas, USA.,Department of Biosciences, Rice University, Houston, Texas, USA
| | - Rachel Van Drunen
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center, Houston, Texas, USA
| | - Anastasia Kralli
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Kristin Mahan
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center, Houston, Texas, USA.,Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center, Houston, Texas, USA
| | - Zhongming Zhao
- Center for Precision Medicine, School of Biomedical Informatics, The University of Texas Health Science Center, Houston, Texas, USA.,Human Genetics Center, School of Public Health, The University of Texas Health Science Center, Houston, Texas, USA
| | - Ashok Kumar
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston, Houston, Texas, USA
| | - Vihang A Narkar
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center, Houston, Texas, USA.,Graduate School of Biomedical Sciences at UTHealth, Houston, Texas, USA
| |
Collapse
|
9
|
Dysregulation of PGC-1α-Dependent Transcriptional Programs in Neurological and Developmental Disorders: Therapeutic Challenges and Opportunities. Cells 2021. [DOI: 10.3390/cells10020352
expr 820281011 + 880698691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Substantial evidence indicates that mitochondrial impairment contributes to neuronal dysfunction and vulnerability in disease states, leading investigators to propose that the enhancement of mitochondrial function should be considered a strategy for neuroprotection. However, multiple attempts to improve mitochondrial function have failed to impact disease progression, suggesting that the biology underlying the normal regulation of mitochondrial pathways in neurons, and its dysfunction in disease, is more complex than initially thought. Here, we present the proteins and associated pathways involved in the transcriptional regulation of nuclear-encoded genes for mitochondrial function, with a focus on the transcriptional coactivator peroxisome proliferator-activated receptor gamma coactivator-1alpha (PGC-1α). We highlight PGC-1α’s roles in neuronal and non-neuronal cell types and discuss evidence for the dysregulation of PGC-1α-dependent pathways in Huntington’s Disease, Parkinson’s Disease, and developmental disorders, emphasizing the relationship between disease-specific cellular vulnerability and cell-type-specific patterns of PGC-1α expression. Finally, we discuss the challenges inherent to therapeutic targeting of PGC-1α-related transcriptional programs, considering the roles for neuron-enriched transcriptional coactivators in co-regulating mitochondrial and synaptic genes. This information will provide novel insights into the unique aspects of transcriptional regulation of mitochondrial function in neurons and the opportunities for therapeutic targeting of transcriptional pathways for neuroprotection.
Collapse
|
10
|
Dysregulation of PGC-1α-Dependent Transcriptional Programs in Neurological and Developmental Disorders: Therapeutic Challenges and Opportunities. Cells 2021; 10:cells10020352. [PMID: 33572179 PMCID: PMC7915819 DOI: 10.3390/cells10020352] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/03/2021] [Accepted: 02/03/2021] [Indexed: 02/08/2023] Open
Abstract
Substantial evidence indicates that mitochondrial impairment contributes to neuronal dysfunction and vulnerability in disease states, leading investigators to propose that the enhancement of mitochondrial function should be considered a strategy for neuroprotection. However, multiple attempts to improve mitochondrial function have failed to impact disease progression, suggesting that the biology underlying the normal regulation of mitochondrial pathways in neurons, and its dysfunction in disease, is more complex than initially thought. Here, we present the proteins and associated pathways involved in the transcriptional regulation of nuclear-encoded genes for mitochondrial function, with a focus on the transcriptional coactivator peroxisome proliferator-activated receptor gamma coactivator-1alpha (PGC-1α). We highlight PGC-1α's roles in neuronal and non-neuronal cell types and discuss evidence for the dysregulation of PGC-1α-dependent pathways in Huntington's Disease, Parkinson's Disease, and developmental disorders, emphasizing the relationship between disease-specific cellular vulnerability and cell-type-specific patterns of PGC-1α expression. Finally, we discuss the challenges inherent to therapeutic targeting of PGC-1α-related transcriptional programs, considering the roles for neuron-enriched transcriptional coactivators in co-regulating mitochondrial and synaptic genes. This information will provide novel insights into the unique aspects of transcriptional regulation of mitochondrial function in neurons and the opportunities for therapeutic targeting of transcriptional pathways for neuroprotection.
Collapse
|
11
|
Kim S, Lee JY, Shin SG, Kim JK, Silwal P, Kim YJ, Shin NR, Kim PS, Won M, Lee SH, Kim SY, Sasai M, Yamamoto M, Kim JM, Bae JW, Jo EK. ESRRA (estrogen related receptor alpha) is a critical regulator of intestinal homeostasis through activation of autophagic flux via gut microbiota. Autophagy 2020; 17:2856-2875. [PMID: 33172329 DOI: 10.1080/15548627.2020.1847460] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The orphan nuclear receptor ESRRA (estrogen related receptor alpha) is critical in mitochondrial biogenesis and macroautophagy/autophagy function; however, the roles of ESRRA in intestinal function remain uncharacterized. Herein we identified that ESRRA acts as a key regulator of intestinal homeostasis by amelioration of colonic inflammation through activation of autophagic flux and control of host gut microbiota. Esrra-deficient mice presented with increased susceptibility to dextran sodium sulfate (DSS)-induced colitis with upregulation of intestinal inflammation. In addition, esrra-null mice had depressed AMP-activated protein kinase phosphorylation (AMPK), lower levels of TFEB (transcription factor EB), and accumulation of SQSTM1/p62 (sequestosome 1) with defective mitochondria in intestinal tissues. Esrra-deficient mice showed distinct gut microbiota composition and significantly higher microbial diversity than wild-type (WT) mice. Cohousing or fecal microbiota transplantation from WT mice to Esrra-deficient mice ameliorated DSS-induced colitis severity. Importantly, patients with ulcerative colitis (UC) had significantly decreased ESRRA expression in intestinal mucosal tissues that correlated with disease activity, suggesting clinical relevance of ESRRA in UC. Taken together, our results show that ESRRA contributes to intestinal homeostasis through autophagy activation and gut microbiota control to protect the host from detrimental inflammation and dysfunctional mitochondria.
Collapse
Affiliation(s)
- Sup Kim
- Department of Radiation Oncology, Chungnam National University Hospital, Daejeon, Korea
| | - June-Young Lee
- Department of Life and Nanopharmaceutical Sciences and Department of Biology, Kyung Hee University, Dongdaemun-gu, Seoul, Korea
| | - Seul Gi Shin
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon Korea.,Infection Control Convergence Research Center, Chungnam National University School of Medicine Daejeon, Korea
| | - Jin Kyung Kim
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon Korea.,Infection Control Convergence Research Center, Chungnam National University School of Medicine Daejeon, Korea
| | - Prashanta Silwal
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon Korea.,Infection Control Convergence Research Center, Chungnam National University School of Medicine Daejeon, Korea
| | - Young Jae Kim
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon Korea.,Infection Control Convergence Research Center, Chungnam National University School of Medicine Daejeon, Korea
| | - Na-Ri Shin
- Korean Collection for Type Cultures, Biological Resource Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup, Korea
| | - Pil Soo Kim
- Department of Life and Nanopharmaceutical Sciences and Department of Biology, Kyung Hee University, Dongdaemun-gu, Seoul, Korea
| | - Minho Won
- Biotechnology Process Engineering Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), Chungcheongbuk-do Korea
| | - Sang-Hee Lee
- Center for Research Equipment, Korea Basic Science Institute, Chungbuk, Korea
| | - Soo Yeon Kim
- Future Medicine Division, Korea Institute of Oriental Medicine, Daejeon Korea
| | - Miwa Sasai
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka, Japan.,Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka Japan
| | - Masahiro Yamamoto
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka, Japan.,Laboratory of Immunoparasitology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka Japan
| | - Jin-Man Kim
- Infection Control Convergence Research Center, Chungnam National University School of Medicine Daejeon, Korea.,Pathology and.,Department of Medical Science, Chungnam National University School of Medicine, Daejeon Korea
| | - Jin-Woo Bae
- Department of Life and Nanopharmaceutical Sciences and Department of Biology, Kyung Hee University, Dongdaemun-gu, Seoul, Korea
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon Korea.,Infection Control Convergence Research Center, Chungnam National University School of Medicine Daejeon, Korea
| |
Collapse
|
12
|
Li D, Cai Y, Teng D, Wu Z, Li W, Tang Y, Liu G. Insights into the interaction mechanisms of estrogen-related receptor alpha (ERRα) with ligands via molecular dynamics simulations. J Biomol Struct Dyn 2020; 38:3867-3878. [PMID: 31498028 DOI: 10.1080/07391102.2019.1666034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 09/03/2019] [Accepted: 09/03/2019] [Indexed: 10/26/2022]
Abstract
Estrogen-related receptor alpha (ERRα), a member of nuclear receptors (NRs), participates in energy metabolism. Recent experiments identified that several agonists to increase the activity of ERRα, which have a therapeutic effect in improving insulin sensitivity and lowering blood glucose levels. However, the detailed molecular mechanism about how the ligands affect the structure of ERRα remains elusive. To better understand the conformational change of ERRα complexed with agonists and inverse agonists, unbiased molecular dynamics (MD) simulations were performed on the ligand binding domain of ERRα (ERRα-LBD) bound with different ligands. According to the results, the ERRα-agonist interactions were more stable in the presence of the peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α). More importantly, we observed that the binding of inverse agonists would decrease the stability of helix 12 (H12) of ERRα. Moreover, we suggested that Phe232 and Phe414 should be key residues in the interaction pathway from ligands to H12, which provided a possible explanation about how ligands impact the structure of ERRα. These results would provide insights into the design of novel and efficient agonists of ERRα to treat metabolic diabetes.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Dongping Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Yingchun Cai
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Dan Teng
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Zengrui Wu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Weihua Li
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Yun Tang
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Guixia Liu
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| |
Collapse
|
13
|
Batista TM, Garcia-Martin R, Cai W, Konishi M, O'Neill BT, Sakaguchi M, Kim JH, Jung DY, Kim JK, Kahn CR. Multi-dimensional Transcriptional Remodeling by Physiological Insulin In Vivo. Cell Rep 2020; 26:3429-3443.e3. [PMID: 30893613 DOI: 10.1016/j.celrep.2019.02.081] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 01/11/2019] [Accepted: 02/21/2019] [Indexed: 12/12/2022] Open
Abstract
Regulation of gene expression is an important aspect of insulin action but in vivo is intertwined with changing levels of glucose and counter-regulatory hormones. Here we demonstrate that under euglycemic clamp conditions, physiological levels of insulin regulate interrelated networks of more than 1,000 transcripts in muscle and liver. These include expected pathways related to glucose and lipid utilization, mitochondrial function, and autophagy, as well as unexpected pathways, such as chromatin remodeling, mRNA splicing, and Notch signaling. These acutely regulated pathways extend beyond those dysregulated in mice with chronic insulin deficiency or insulin resistance and involve a broad network of transcription factors. More than 150 non-coding RNAs were regulated by insulin, many of which also responded to fasting and refeeding. Pathway analysis and RNAi knockdown revealed a role for lncRNA Gm15441 in regulating fatty acid oxidation in hepatocytes. Altogether, these changes in coding and non-coding RNAs provide an integrated transcriptional network underlying the complexity of insulin action.
Collapse
Affiliation(s)
- Thiago M Batista
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Ruben Garcia-Martin
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Weikang Cai
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Masahiro Konishi
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA
| | - Brian T O'Neill
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA; Division of Endocrinology and Metabolism, Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Masaji Sakaguchi
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA; Department of Metabolic Medicine, Kumamoto University, 1-1-1 Honjo, Chuoku, Kumamoto 860-8556, Japan
| | - Jong Hun Kim
- Program in Molecular Medicine, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA; Department of Food Science and Biotechnology, Sungshin University, Seoul 01133, Republic of Korea
| | - Dae Young Jung
- Program in Molecular Medicine, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Jason K Kim
- Program in Molecular Medicine, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA; Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - C Ronald Kahn
- Section of Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
14
|
How Epigenetic Modifications Drive the Expression and Mediate the Action of PGC-1α in the Regulation of Metabolism. Int J Mol Sci 2019; 20:ijms20215449. [PMID: 31683747 PMCID: PMC6862278 DOI: 10.3390/ijms20215449] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/29/2019] [Accepted: 10/29/2019] [Indexed: 12/12/2022] Open
Abstract
Epigenetic changes are a hallmark of short- and long-term transcriptional regulation, and hence instrumental in the control of cellular identity and plasticity. Epigenetic mechanisms leading to changes in chromatin structure, accessibility for recruitment of transcriptional complexes, and interaction of enhancers and promoters all contribute to acute and chronic adaptations of cells, tissues and organs to internal and external perturbations. Similarly, the peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) is activated by stimuli that alter the cellular energetic demand, and subsequently controls complex transcriptional networks responsible for cellular plasticity. It thus is of no surprise that PGC-1α is under the control of epigenetic mechanisms, and constitutes a mediator of epigenetic changes in various tissues and contexts. In this review, we summarize the current knowledge of the link between epigenetics and PGC-1α in health and disease.
Collapse
|
15
|
Soledad RB, Charles S, Samarjit D. The secret messages between mitochondria and nucleus in muscle cell biology. Arch Biochem Biophys 2019; 666:52-62. [PMID: 30935885 PMCID: PMC6538274 DOI: 10.1016/j.abb.2019.03.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 03/01/2019] [Accepted: 03/28/2019] [Indexed: 02/06/2023]
Abstract
Over two thousand proteins are found in the mitochondrial compartment but the mitochondrial genome codes for only 13 proteins. The majority of mitochondrial proteins are products of nuclear genes and are synthesized in the cytosol, then translocated into the mitochondria. Most of the subunits of the five respiratory chain complexes in the inner mitochondrial membrane, which generate a proton gradient across the membrane and produce ATP, are encoded by nuclear genes. Therefore, it is quite clear that import of nuclear-encoded proteins into the mitochondria is essential for mitochondrial function. Nuclear to mitochondrial communication is well studied. However, there is another arm to this communication, mitochondria to nucleus retrograde signaling. This plays an important role in the maintenance of cellular homeostasis, and is less well studied. Several transcription factors, including Sp1, SIRT3 and GSP2, are activated by altered mitochondrial function. These activated transcription factors then translocate to the nucleus. Based on the mitochondrially generated molecular signal, nuclear genes are targeted, which alters transcription of nuclear genes that code for mitochondrial proteins. This review article will mainly focus on this interactive and bi-directional communication between mitochondria and nucleus, and how this communication plays a significant role in muscle cell biology.
Collapse
Affiliation(s)
| | - Steenbergen Charles
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| | - Das Samarjit
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States.
| |
Collapse
|
16
|
Vargas G, Bouchet M, Bouazza L, Reboul P, Boyault C, Gervais M, Kan C, Benetollo C, Brevet M, Croset M, Mazel M, Cayrefourcq L, Geraci S, Vacher S, Pantano F, Filipits M, Driouch K, Bieche I, Gnant M, Jacot W, Aubin JE, Duterque-Coquillaud M, Alix-Panabières C, Clézardin P, Bonnelye E. ERRα promotes breast cancer cell dissemination to bone by increasing RANK expression in primary breast tumors. Oncogene 2019; 38:950-964. [PMID: 30478447 DOI: 10.1038/s41388-018-0579-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 10/20/2018] [Indexed: 02/07/2023]
Abstract
Bone is the most common metastatic site for breast cancer. Estrogen-related-receptor alpha (ERRα) has been implicated in cancer cell invasiveness. Here, we established that ERRα promotes spontaneous metastatic dissemination of breast cancer cells from primary mammary tumors to the skeleton. We carried out cohort studies, pharmacological inhibition, gain-of-function analyses in vivo and cellular and molecular studies in vitro to identify new biomarkers in breast cancer metastases. Meta-analysis of human primary breast tumors revealed that high ERRα expression levels were associated with bone but not lung metastases. ERRα expression was also detected in circulating tumor cells from metastatic breast cancer patients. ERRα overexpression in murine 4T1 breast cancer cells promoted spontaneous bone micro-metastases formation when tumor cells were inoculated orthotopically, whereas lung metastases occurred irrespective of ERRα expression level. In vivo, Rank was identified as a target for ERRα. That was confirmed in vitro in Rankl stimulated tumor cell invasion, in mTOR/pS6K phosphorylation, by transactivation assay, ChIP and bioinformatics analyses. Moreover, pharmacological inhibition of ERRα reduced primary tumor growth, bone micro-metastases formation and Rank expression in vitro and in vivo. Transcriptomic studies and meta-analysis confirmed a positive association between metastases and ERRα/RANK in breast cancer patients and also revealed a positive correlation between ERRα and BRCA1mut carriers. Taken together, our results reveal a novel ERRα/RANK axis by which ERRα in primary breast cancer promotes early dissemination of cancer cells to bone. These findings suggest that ERRα may be a useful therapeutic target to prevent bone metastases.
Collapse
Affiliation(s)
- G Vargas
- INSERM-UMR1033, Lyon, France
- University of Lyon1, Lyon, France
| | - M Bouchet
- INSERM-UMR1033, Lyon, France
- University of Lyon1, Lyon, France
- IGFL, Lyon, France
| | - L Bouazza
- INSERM-UMR1033, Lyon, France
- University of Lyon1, Lyon, France
| | - P Reboul
- UMR7365-CNRS-Université de Lorraine, Nancy, France
| | - C Boyault
- Institute for Advanced Biosciences, Grenoble, France
| | - M Gervais
- INSERM-UMR1033, Lyon, France
- University of Lyon1, Lyon, France
| | - C Kan
- INSERM-UMR1033, Lyon, France
- University of Lyon1, Lyon, France
- Center for Cancer Research, University of Sydney, Sydney, Australia
| | - C Benetollo
- University of Lyon1, Lyon, France
- INSERM-U1028-CNRS-UMR5292, Lyon, France
| | - M Brevet
- INSERM-UMR1033, Lyon, France
- Centre de Biologie et de Pathologie Est, Bron, France
| | - M Croset
- INSERM-UMR1033, Lyon, France
- University of Lyon1, Lyon, France
| | - M Mazel
- EA2415-Institut Universitaire de Recherche Clinique, Montpellier, France
| | - L Cayrefourcq
- EA2415-Institut Universitaire de Recherche Clinique, Montpellier, France
| | - S Geraci
- INSERM-UMR1033, Lyon, France
- University of Lyon1, Lyon, France
| | - S Vacher
- Department of Genetics, Institut-Curie, Paris, France
| | - F Pantano
- University-Campus-Bio-Medico, Rome, 00128, Italy
| | - M Filipits
- Department of Surgery and Comprehensive Cancer Center, Medical-University of Vienna, Vienna, Austria
| | - K Driouch
- Department of Genetics, Institut-Curie, Paris, France
| | - I Bieche
- Department of Genetics, Institut-Curie, Paris, France
| | - M Gnant
- Department of Surgery and Comprehensive Cancer Center, Medical-University of Vienna, Vienna, Austria
| | - W Jacot
- Montpellier Cancer Institute, Montpellier, France
| | - J E Aubin
- University of Toronto, Toronto, Canada
| | | | - C Alix-Panabières
- EA2415-Institut Universitaire de Recherche Clinique, Montpellier, France
| | - P Clézardin
- INSERM-UMR1033, Lyon, France
- University of Lyon1, Lyon, France
| | - E Bonnelye
- INSERM-UMR1033, Lyon, France.
- University of Lyon1, Lyon, France.
| |
Collapse
|
17
|
Intensity-dependent gene expression after aerobic exercise in endurance-trained skeletal muscle. Biol Sport 2018; 35:277-289. [PMID: 30449946 PMCID: PMC6224845 DOI: 10.5114/biolsport.2018.77828] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 09/14/2017] [Accepted: 03/16/2018] [Indexed: 01/06/2023] Open
Abstract
We investigated acute exercise-induced gene expression in skeletal muscle adapted to aerobic training. Vastus lateralis muscle samples were taken in ten endurance-trained males prior to, and just after, 4 h, and 8 h after acute cycling sessions with different intensities, 70% and 50% V˙O2max. High-throughput RNA sequencing was applied in samples from two subjects to evaluate differentially expressed genes after intensive exercise (70% V˙O2max), and then the changes in expression for selected genes were validated by quantitative PCR (qPCR). To define exercise-induced genes, we compared gene expression after acute exercise with different intensities, 70% and 50% V˙O2max, by qPCR. The transcriptome is dynamically changed during the first hours of recovery after intensive exercise (70% V˙O2max). A computational approach revealed that the changes might be related to up- and down-regulation of the activity of transcription activators and repressors, respectively. The exercise increased expression of many genes encoding protein kinases, while genes encoding transcriptional regulators were both up- and down-regulated. Evaluation of the gene expression after exercise with different intensities revealed that some genes changed expression in an intensity-dependent manner, but others did not: the majority of genes encoding protein kinases, oxidative phosphorylation and activator protein (AP)-1-related genes significantly correlated with markers of exercise stress (power, blood lactate during exercise and post-exercise blood cortisol), while transcriptional repressors and circadian-related genes did not. Some of the changes in gene expression after exercise seemingly may be modulated by circadian rhythm.
Collapse
|
18
|
Singh BK, Sinha RA, Tripathi M, Mendoza A, Ohba K, Sy JAC, Xie SY, Zhou J, Ho JP, Chang CY, Wu Y, Giguère V, Bay BH, Vanacker JM, Ghosh S, Gauthier K, Hollenberg AN, McDonnell DP, Yen PM. Thyroid hormone receptor and ERRα coordinately regulate mitochondrial fission, mitophagy, biogenesis, and function. Sci Signal 2018; 11:eaam5855. [PMID: 29945885 DOI: 10.1126/scisignal.aam5855] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2023]
Abstract
Thyroid hormone receptor β1 (THRB1) and estrogen-related receptor α (ESRRA; also known as ERRα) both play important roles in mitochondrial activity. To understand their potential interactions, we performed transcriptome and ChIP-seq analyses and found that many genes that were co-regulated by both THRB1 and ESRRA were involved in mitochondrial metabolic pathways. These included oxidative phosphorylation (OXPHOS), the tricarboxylic acid (TCA) cycle, and β-oxidation of fatty acids. TH increased ESRRA expression and activity in a THRB1-dependent manner through the induction of the transcriptional coactivator PPARGC1A (also known as PGC1α). Moreover, TH induced mitochondrial biogenesis, fission, and mitophagy in an ESRRA-dependent manner. TH also induced the expression of the autophagy-regulating kinase ULK1 through ESRRA, which then promoted DRP1-mediated mitochondrial fission. In addition, ULK1 activated the docking receptor protein FUNDC1 and its interaction with the autophagosomal protein MAP1LC3B-II to induce mitophagy. siRNA knockdown of ESRRA, ULK1, DRP1, or FUNDC1 inhibited TH-induced autophagic clearance of mitochondria through mitophagy and decreased OXPHOS. These findings show that many of the mitochondrial actions of TH are mediated through stimulation of ESRRA expression and activity, and co-regulation of mitochondrial turnover through the PPARGC1A-ESRRA-ULK1 pathway is mediated by their regulation of mitochondrial fission and mitophagy. Hormonal or pharmacologic induction of ESRRA expression or activity could improve mitochondrial quality in metabolic disorders.
Collapse
Affiliation(s)
- Brijesh K Singh
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore.
| | - Rohit A Sinha
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
- Department of Endocrinology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Raebareli Road, Lucknow 226014, Uttar Pradesh, India
| | - Madhulika Tripathi
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Arturo Mendoza
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Center for Life Sciences, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Kenji Ohba
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
- Department of Internal Medicine, Enshu Hospital, Hamamatsu, Shizuoka 430-0929, Japan
| | - Jann A C Sy
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Sherwin Y Xie
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Jin Zhou
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Jia Pei Ho
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Ching-Yi Chang
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, C238A Levine Science Research Center, Durham, NC 27710, USA
| | - Yajun Wu
- Department of Anatomy, Yong Loo Lin School of Medicine, NUS, Singapore
| | - Vincent Giguère
- Goodman Cancer Research Centre, McGill University, 1160 Pine Avenue West, Montreal, Québec H3A 1A3, Canada
| | - Boon-Huat Bay
- Department of Anatomy, Yong Loo Lin School of Medicine, NUS, Singapore
| | - Jean-Marc Vanacker
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS, Ecole Normale Supérieure de Lyon, 46 Allée d'Italie, 69364 Lyon Cedex 07, France
| | - Sujoy Ghosh
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore
| | - Karine Gauthier
- Institut de Génomique Fonctionnelle de Lyon, Université de Lyon, Université Lyon 1, CNRS, Ecole Normale Supérieure de Lyon, 46 Allée d'Italie, 69364 Lyon Cedex 07, France
| | - Anthony N Hollenberg
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Center for Life Sciences, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Donald P McDonnell
- Department of Internal Medicine, Enshu Hospital, Hamamatsu, Shizuoka 430-0929, Japan
| | - Paul M Yen
- Laboratory of Hormonal Regulation, Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore (NUS) Medical School, Singapore 169857, Singapore.
| |
Collapse
|
19
|
Popov DV. Adaptation of Skeletal Muscles to Contractile Activity of Varying Duration and Intensity: The Role of PGC-1α. BIOCHEMISTRY (MOSCOW) 2018; 83:613-628. [DOI: 10.1134/s0006297918060019] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
20
|
Wang M, Yang G, Jiang X, Lu D, Mei H, Chen B. Peroxisome Proliferator-Activated Receptor-γ Coactivator-1α (PGC-1α) Regulates the Expression of B-Cell Lymphoma/Leukemia-2 (Bcl-2) and Promotes the Survival of Mesenchymal Stem Cells (MSCs) via PGC-1α/ERRα Interaction in the Absence of Serum, Hypoxia, and High Glucose Conditions. Med Sci Monit 2017; 23:3451-3460. [PMID: 28711948 PMCID: PMC5525574 DOI: 10.12659/msm.902183] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 12/08/2016] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND To study the effect of estrogen-related receptor α (ERRα) and peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) on mesenchymal stem cells (MSCs) apoptosis, and further investigated its detailed molecular mechanisms in the absence of serum, hypoxia, and high glucose conditions. MATERIAL AND METHODS In our study, we first evaluated the expression rates of CD14, CD34, CD45, CD44, CD29, and Sca-1 surface markers on MSCs by flow cytometry. Then, the ability of osteogenic and fatty differentiation of MSCs was determined by osteogenic differentiation and adipogenesis reagent kit. Next, Annexin V-APC/7-AAD apoptosis kit was used for detecting the apoptosis rate of MSCs. RT-PCR and Western blotting were used for detection of mRNA expression and proteins expression, respectively. RESULTS Our data showed that the MSCs used in our study were capable of self-renewal and differentiating into many cell lineages, such as osteogenic differentiation and adipogenesis. Our results further showed that over-expression of PGC-1α could protect MSCs from apoptosis induced by rotenone. We also found that PGC-1α over-expression could enhance the expression of anti-apoptotic gene Bcl-2, and inhibit the expression of pro-apoptotic gene Bax in MSCs. In addition, our data demonstrated that PGC-1α could induce upregulation of Bcl-2 and further promote the survival of MSCs by interacting with ERRα. CONCLUSIONS In the absence of serum, hypoxia and high glucose conditions, PGC-1α can regulate the expression of Bcl-2 and promote the survival of MSCs via PGC-1α/ERRα interaction.
Collapse
Affiliation(s)
- Min Wang
- Department of Endocrinology, Southwest Hospital, Third Military Medical University, Chongqing, P.R. China
| | - Guangxin Yang
- Department of Endocrinology, Southwest Hospital, Third Military Medical University, Chongqing, P.R. China
| | - Xiaoyan Jiang
- Department of Endocrinology, Southwest Hospital, Third Military Medical University, Chongqing, P.R. China
| | - Debin Lu
- Department of Endocrinology, Southwest Hospital, Third Military Medical University, Chongqing, P.R. China
| | - Hao Mei
- Center of Outcomes Research and Evaluation, Yale-New Haven Hospital, New Haven, CT, U.S.A
| | - Bing Chen
- Department of Endocrinology, Southwest Hospital, Third Military Medical University, Chongqing, P.R. China
| |
Collapse
|
21
|
Kupr B, Schnyder S, Handschin C. Role of Nuclear Receptors in Exercise-Induced Muscle Adaptations. Cold Spring Harb Perspect Med 2017; 7:a029835. [PMID: 28242783 PMCID: PMC5453380 DOI: 10.1101/cshperspect.a029835] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Skeletal muscle is not only one of the largest, but also one of the most dynamic organs. For example, plasticity elicited by endurance or resistance exercise entails complex transcriptional programs that are still poorly understood. Various signaling pathways are engaged in the contracting muscle fiber and collectively culminate in the modulation of the activity of numerous transcription factors (TFs) and coregulators. Because exercise confers many benefits for the prevention and treatment of a wide variety of pathologies, pharmacological activation of signaling pathways and TFs is an attractive avenue to elicit therapeutic effects. Members of the nuclear receptor (NR) superfamily are of particular interest owing to the presence of well-defined DNA- and ligand-binding domains. In this review, we summarize the current understanding of the involvement of NRs in muscle biology and exercise adaptation.
Collapse
Affiliation(s)
- Barbara Kupr
- Biozentrum, University of Basel, Basel 4056, Switzerland
| | | | | |
Collapse
|
22
|
Paracrine cross-talk between skeletal muscle and macrophages in exercise by PGC-1α-controlled BNP. Sci Rep 2017; 7:40789. [PMID: 28091624 PMCID: PMC5238507 DOI: 10.1038/srep40789] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 12/09/2016] [Indexed: 12/11/2022] Open
Abstract
Activation of resident and infiltrating immune cells is a central event in training adaptation and other contexts of skeletal muscle repair and regeneration. A precise orchestration of inflammatory events in muscle fibers and immune cells is required after recurrent contraction-relaxation cycles. However, the mechanistic aspects of this important regulation remain largely unknown. We now demonstrate that besides a dominant role in controlling cellular metabolism, the peroxisome proliferator-activated receptor γ co-activator 1α (PGC-1α) also has a profound effect on cytokine expression in muscle tissue. Muscle PGC-1α expression results in activation of tissue-resident macrophages, at least in part mediated by PGC-1α-dependent B-type natriuretic peptide (BNP) production and secretion. Positive effects of exercise in metabolic diseases and other pathologies associated with chronic inflammation could accordingly involve the PGC-1α-BNP axis and thereby provide novel targets for therapeutic approaches.
Collapse
|
23
|
Schnyder S, Kupr B, Handschin C. Coregulator-mediated control of skeletal muscle plasticity - A mini-review. Biochimie 2017; 136:49-54. [PMID: 28057584 DOI: 10.1016/j.biochi.2016.12.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 12/13/2016] [Accepted: 12/19/2016] [Indexed: 10/20/2022]
Abstract
Skeletal muscle plasticity is a complex process entailing massive transcriptional programs. These changes are mediated by the action of nuclear receptors and other transcription factors. In addition, coregulator proteins have emerged as important players in this process by linking transcription factors to the RNA polymerase II complex and inducing changes in the chromatic structure. An accumulating body of work highlights the pleiotropic functions of coregulator proteins in the control of tissue-specific and whole body metabolism. In skeletal muscle, several coregulators have been identified as potent modulators of metabolic and myofibrillar plasticity. In this mini-review, we will discuss the control, function and physiological significance of these coregulators in skeletal muscle biology.
Collapse
Affiliation(s)
- Svenia Schnyder
- Biozentrum, University of Basel, Klingelbergstrasse 50/70, CH-4056 Basel, Switzerland
| | - Barbara Kupr
- Biozentrum, University of Basel, Klingelbergstrasse 50/70, CH-4056 Basel, Switzerland
| | - Christoph Handschin
- Biozentrum, University of Basel, Klingelbergstrasse 50/70, CH-4056 Basel, Switzerland.
| |
Collapse
|
24
|
Koga T, Yao PL, Goudarzi M, Murray IA, Balandaram G, Gonzalez FJ, Perdew GH, Fornace AJ, Peters JM. Regulation of Cytochrome P450 2B10 (CYP2B10) Expression in Liver by Peroxisome Proliferator-activated Receptor-β/δ Modulation of SP1 Promoter Occupancy. J Biol Chem 2016; 291:25255-25263. [PMID: 27765815 DOI: 10.1074/jbc.m116.755447] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 10/13/2016] [Indexed: 01/12/2023] Open
Abstract
Alcoholic liver disease is a pathological condition caused by overconsumption of alcohol. Because of the high morbidity and mortality associated with this disease, there remains a need to elucidate the molecular mechanisms underlying its etiology and to develop new treatments. Because peroxisome proliferator-activated receptor-β/δ (PPARβ/δ) modulates ethanol-induced hepatic effects, the present study examined alterations in gene expression that may contribute to this disease. Chronic ethanol treatment causes increased hepatic CYP2B10 expression inPparβ/δ+/+ mice but not in Pparβ/δ-/- mice. Nuclear and cytosolic localization of the constitutive androstane receptor (CAR), a transcription factor known to regulate Cyp2b10 expression, was not different between genotypes. PPARγ co-activator 1α, a co-activator of both CAR and PPARβ/δ, was up-regulated in Pparβ/δ+/+ liver following ethanol exposure, but not in Pparβ/δ-/- liver. Functional mapping of the Cyp2b10 promoter and ChIP assays revealed that PPARβ/δ-dependent modulation of SP1 promoter occupancy up-regulated Cyp2b10 expression in response to ethanol. These results suggest that PPARβ/δ regulates Cyp2b10 expression indirectly by modulating SP1 and PPARγ co-activator 1α expression and/or activity independent of CAR activity. Ligand activation of PPARβ/δ attenuates ethanol-induced Cyp2b10 expression in Pparβ/δ+/+ liver but not in Pparβ/δ-/- liver. Strikingly, Cyp2b10 suppression by ligand activation of PPARβ/δ following ethanol treatment occurred in hepatocytes and was mediated by paracrine signaling from Kupffer cells. Combined, results from the present study demonstrate a novel regulatory role of PPARβ/δ in modulating CYP2B10 that may contribute to the etiology of alcoholic liver disease.
Collapse
Affiliation(s)
- Takayuki Koga
- From the Department of Veterinary and Biomedical Sciences and the Center of Molecular Toxicology and Carcinogenesis, Pennsylvania State University, University Park, Pennsylvania 16802
| | - Pei-Li Yao
- From the Department of Veterinary and Biomedical Sciences and the Center of Molecular Toxicology and Carcinogenesis, Pennsylvania State University, University Park, Pennsylvania 16802
| | - Maryam Goudarzi
- the Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, D. C., 20057, and
| | - Iain A Murray
- From the Department of Veterinary and Biomedical Sciences and the Center of Molecular Toxicology and Carcinogenesis, Pennsylvania State University, University Park, Pennsylvania 16802
| | - Gayathri Balandaram
- From the Department of Veterinary and Biomedical Sciences and the Center of Molecular Toxicology and Carcinogenesis, Pennsylvania State University, University Park, Pennsylvania 16802
| | - Frank J Gonzalez
- the Laboratory of Metabolism, National Cancer Institute, Bethesda, Maryland 20892
| | - Gary H Perdew
- From the Department of Veterinary and Biomedical Sciences and the Center of Molecular Toxicology and Carcinogenesis, Pennsylvania State University, University Park, Pennsylvania 16802
| | - Albert J Fornace
- the Department of Biochemistry and Molecular and Cellular Biology, Georgetown University, Washington, D. C., 20057, and
| | - Jeffrey M Peters
- From the Department of Veterinary and Biomedical Sciences and the Center of Molecular Toxicology and Carcinogenesis, Pennsylvania State University, University Park, Pennsylvania 16802,
| |
Collapse
|