1
|
Mann CG, MacArthur MR, Zhang J, Gong S, AbuSalim JE, Hunter CJ, Lu W, Agius T, Longchamp A, Allagnat F, Rabinowitz J, Mitchell JR, De Bock K, Mitchell SJ. Sulfur Amino Acid Restriction Enhances Exercise Capacity in Mice by Boosting Fat Oxidation in Muscle. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.27.601041. [PMID: 39005372 PMCID: PMC11244859 DOI: 10.1101/2024.06.27.601041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Dietary restriction of the sulfur-containing amino acids methionine and cysteine (SAAR) improves body composition, enhances insulin sensitivity, and extends lifespan; benefits seen also with endurance exercise. Yet, the impact of SAAR on skeletal muscle remains largely unexplored. Here we demonstrate that one week of SAAR in sedentary, young, male mice increases endurance exercise capacity. Indirect calorimetry showed that SAAR increased lipid oxidation at rest and delayed the onset of carbohydrate utilization during exercise. Transcriptomic analysis revealed increased expression of genes involved in fatty acid catabolism especially in glycolytic muscle following SAAR. These findings were functionally supported by increased fatty acid circulatory turnover flux and muscle β-oxidation. Reducing lipid uptake from circulation through endothelial cell (EC)-specific CD36 deletion attenuated the running phenotype. Mechanistically, VEGF-signaling inhibition prevented exercise increases following SAAR, without affecting angiogenesis, implicating noncanonical VEGF signaling and EC CD36-dependent fatty acid transport in regulating exercise capacity by influencing muscle substrate availability.
Collapse
Affiliation(s)
- Charlotte G Mann
- Department of Health Sciences and Technology, ETH Zurich, Zurich 8092, Switzerland
| | - Michael R MacArthur
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
- Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
- Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ 08544, USA
| | - Jing Zhang
- Department of Health Sciences and Technology, ETH Zurich, Zurich 8092, Switzerland
| | - Songlin Gong
- Department of Health Sciences and Technology, ETH Zurich, Zurich 8092, Switzerland
| | - Jenna E AbuSalim
- Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
- Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ 08544, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Craig J. Hunter
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
- Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
- Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ 08544, USA
| | - Wenyun Lu
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
- Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Thomas Agius
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), Lausanne 1005, Switzerland
| | - Alban Longchamp
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), Lausanne 1005, Switzerland
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Florent Allagnat
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), Lausanne 1005, Switzerland
| | - Joshua Rabinowitz
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
- Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
- Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ 08544, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - James R Mitchell
- Department of Health Sciences and Technology, ETH Zurich, Zurich 8092, Switzerland
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Katrien De Bock
- Department of Health Sciences and Technology, ETH Zurich, Zurich 8092, Switzerland
| | - Sarah J Mitchell
- Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
2
|
Zhong T, Gao N, Guan Y, Liu Z, Guan J. Co-Delivery of Bioengineered Exosomes and Oxygen for Treating Critical Limb Ischemia in Diabetic Mice. ACS NANO 2023; 17:25157-25174. [PMID: 38063490 PMCID: PMC10790628 DOI: 10.1021/acsnano.3c08088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2023]
Abstract
Diabetic patients with critical limb ischemia face a high rate of limb amputation. Regeneration of the vasculature and skeletal muscles can salvage diseased limbs. Therapy using stem cell-derived exosomes that contain multiple proangiogenic and promyogenic factors represents a promising strategy. Yet the therapeutic efficacy is not optimal because exosomes alone cannot efficiently rescue and recruit endothelial and skeletal muscle cells and restore their functions under hyperglycemic and ischemic conditions. To address these limitations, we fabricated ischemic-limb-targeting stem cell-derived exosomes and oxygen-releasing nanoparticles and codelivered them in order to recruit endothelial and skeletal muscle cells, improve cell survival under ischemia before vasculature is established, and restore cell morphogenic function under high glucose and ischemic conditions. The exosomes and oxygen-releasing nanoparticles, delivered by intravenous injection, specifically accumulated in the ischemic limbs. Following 4 weeks of delivery, the exosomes and released oxygen synergistically stimulated angiogenesis and muscle regeneration without inducing substantial inflammation and reactive oxygen species overproduction. Our work demonstrates that codelivery of exosomes and oxygen is a promising treatment solution for saving diabetic ischemic limbs.
Collapse
Affiliation(s)
- Ting Zhong
- Department of Mechanical Engineering & Materials Science, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Ning Gao
- Institute of Materials Science and Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Ya Guan
- Institute of Materials Science and Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Zhongting Liu
- Institute of Materials Science and Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Jianjun Guan
- Department of Mechanical Engineering & Materials Science, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
- Institute of Materials Science and Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
- Department of Biomedical Engineering, McKelvey School of Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| |
Collapse
|
3
|
Wang XX, Hua T, Wang HB. Estrogen receptor-related receptor γ uppresses hypoxia-induced angiogenesis by regulating VEGFA in endometrial cancer. Gynecol Endocrinol 2023; 39:2264411. [PMID: 37859604 DOI: 10.1080/09513590.2023.2264411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 09/21/2023] [Indexed: 10/21/2023] Open
Abstract
OBJECTIVE Estrogen receptor-related receptor γ (ERRγ), is implicated in cancer cell proliferation and metastasis. The function of ERRγ in tumor angiogenesis, however, is to be revealed. This study was designed to elaborate the regulatory effect of ERRγ on angiogenesis in endometrial cancer (EC). METHODS Immunohistochemistry (IHC) was adopted to determine the protein expression of ERRγ, VEGFA, CD31 and hypoxia-inducible factor-1 (HIF-1) in tumor tissues. HEC-1A cells stably expressing ERRγ were established bytransfection, and then an endothelial cell tube formation assay was performed. CCK-8 assay was employed for cell viability, and wound healing assay for cell migration ability. Besides, western blot, ELISA and qRT-PCR were used to examine the VEGFA expression. After hypoxia treatment of ERRγ overexpressing HEC-1A cells, the ERRγ expression and VEGFA expression were determined by western blot. Finally, EC xenografts in nude mice were constructed by subcutaneous injection of ERRγ stably expressing HEC-1A cells and control HEC-1A cells. RESULTS IHC results revealed a negative correlation between the expression of ERRγ and VEGFA in EC tissues. ERRγ overexpression significantly decreased the level of HIF-1 in tumor tissue of nude mice. ERRγ overexpression down-regulated inhibited angiogenesis capability and inhibited the proliferation and migration of HEC-1A cells. Furthermore, ERRγ expression was suppressed under the condition of hypoxia while restoration of ERRγ partially inhibited hypoxia-induced VEGFA expression in HEC-1A cells. CONCLUSIONS ERRγ is an angiogenesis suppressor and involved in hypoxia-induced VEGFA expression in EC. Hence, ERRγ might be a promising antiangiogenic target for human EC.
Collapse
Affiliation(s)
- Xiao-Xiao Wang
- Department of Gynecology, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Teng Hua
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan,China
| | - Hong-Bo Wang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan,China
| |
Collapse
|
4
|
Narkar VA. Exercise and Ischemia-Activated Pathways in Limb Muscle Angiogenesis and Vascular Regeneration. Methodist Debakey Cardiovasc J 2023; 19:58-68. [PMID: 38028974 PMCID: PMC10655757 DOI: 10.14797/mdcvj.1304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 10/26/2023] [Indexed: 12/01/2023] Open
Abstract
Exercise has a profound effect on cardiovascular disease, particularly through vascular remodeling and regeneration. Peripheral artery disease (PAD) is one such cardiovascular condition that benefits from regular exercise or rehabilitative physical therapy in terms of slowing the progression of disease and delaying amputations. Various rodent pre-clinical studies using models of PAD and exercise have shed light on molecular pathways of vascular regeneration. Here, I review key exercise-activated signaling pathways (nuclear receptors, kinases, and hypoxia inducible factors) in the skeletal muscle that drive paracrine regenerative angiogenesis. The rationale for highlighting the skeletal muscle is that it is the largest organ recruited during exercise. During exercise, skeletal muscle releases several myokines, including angiogenic factors and cytokines that drive tissue vascular regeneration via activation of endothelial cells, as well as by recruiting immune and endothelial progenitor cells. Some of these core exercise-activated pathways can be extrapolated to vascular regeneration in other organs. I also highlight future areas of exercise research (including metabolomics, single cell transcriptomics, and extracellular vesicle biology) to advance our understanding of how exercise induces vascular regeneration at the molecular level, and propose the idea of "exercise-mimicking" therapeutics for vascular recovery.
Collapse
Affiliation(s)
- Vihang A. Narkar
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, Texas, US
| |
Collapse
|
5
|
Sun Z, Yang L, Kiram A, Yang J, Yang Z, Xiao L, Yin Y, Liu J, Mao Y, Zhou D, Yu H, Zhou Z, Xu D, Jia Y, Ding C, Guo Q, Wang H, Li Y, Wang L, Fu T, Hu S, Gan Z. FNIP1 abrogation promotes functional revascularization of ischemic skeletal muscle by driving macrophage recruitment. Nat Commun 2023; 14:7136. [PMID: 37932296 PMCID: PMC10628247 DOI: 10.1038/s41467-023-42690-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 10/18/2023] [Indexed: 11/08/2023] Open
Abstract
Ischaemia of the heart and limbs attributable to compromised blood supply is a major cause of mortality and morbidity. The mechanisms of functional angiogenesis remain poorly understood, however. Here we show that FNIP1 plays a critical role in controlling skeletal muscle functional angiogenesis, a process pivotal for muscle revascularization during ischemia. Muscle FNIP1 expression is down-regulated by exercise. Genetic overexpression of FNIP1 in myofiber causes limited angiogenesis in mice, whereas its myofiber-specific ablation markedly promotes the formation of functional blood vessels. Interestingly, the increased muscle angiogenesis is independent of AMPK but due to enhanced macrophage recruitment in FNIP1-depleted muscles. Mechanistically, myofiber FNIP1 deficiency induces PGC-1α to activate chemokine gene transcription, thereby driving macrophage recruitment and muscle angiogenesis program. Furthermore, in a mouse hindlimb ischemia model of peripheral artery disease, the loss of myofiber FNIP1 significantly improved the recovery of blood flow. Thus, these results reveal a pivotal role of FNIP1 as a negative regulator of functional angiogenesis in muscle, offering insight into potential therapeutic strategies for ischemic diseases.
Collapse
Affiliation(s)
- Zongchao Sun
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Likun Yang
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Abdukahar Kiram
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Jing Yang
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Zhuangzhuang Yang
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China
| | - Liwei Xiao
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Yujing Yin
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Jing Liu
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Yan Mao
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Danxia Zhou
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Hao Yu
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Zheng Zhou
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Dengqiu Xu
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Yuhuan Jia
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Chenyun Ding
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Qiqi Guo
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China
| | - Hongwei Wang
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Yan Li
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Li Wang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Tingting Fu
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China.
| | - Shijun Hu
- Department of Cardiovascular Surgery of the First Affiliated Hospital & Institute for Cardiovascular Science, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Suzhou Medical College, Soochow University, Suzhou, China.
| | - Zhenji Gan
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing University, Nanjing, China.
- Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing University, Nanjing, China.
- Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, China.
| |
Collapse
|
6
|
Pass CG, Palzkill V, Tan J, Kim K, Thome T, Yang Q, Fazzone B, Robinson ST, O’Malley KA, Yue F, Scali ST, Berceli SA, Ryan TE. Single-Nuclei RNA-Sequencing of the Gastrocnemius Muscle in Peripheral Artery Disease. Circ Res 2023; 133:791-809. [PMID: 37823262 PMCID: PMC10599805 DOI: 10.1161/circresaha.123.323161] [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] [Received: 06/05/2023] [Revised: 09/22/2023] [Accepted: 09/26/2023] [Indexed: 10/13/2023]
Abstract
BACKGROUND Lower extremity peripheral artery disease (PAD) is a growing epidemic with limited effective treatment options. Here, we provide a single-nuclei atlas of PAD limb muscle to facilitate a better understanding of the composition of cells and transcriptional differences that comprise the diseased limb muscle. METHODS We obtained gastrocnemius muscle specimens from 20 patients with PAD and 12 non-PAD controls. Nuclei were isolated and single-nuclei RNA-sequencing was performed. The composition of nuclei was characterized by iterative clustering via principal component analysis, differential expression analysis, and the use of known marker genes. Bioinformatics analysis was performed to determine differences in gene expression between PAD and non-PAD nuclei, as well as subsequent analysis of intercellular signaling networks. Additional histological analyses of muscle specimens accompany the single-nuclei RNA-sequencing atlas. RESULTS Single-nuclei RNA-sequencing analysis indicated a fiber type shift with patients with PAD having fewer type I (slow/oxidative) and more type II (fast/glycolytic) myonuclei compared with non-PAD, which was confirmed using immunostaining of muscle specimens. Myonuclei from PAD displayed global upregulation of genes involved in stress response, autophagy, hypoxia, and atrophy. Subclustering of myonuclei also identified populations that were unique to PAD muscle characterized by metabolic dysregulation. PAD muscles also displayed unique transcriptional profiles and increased diversity of transcriptomes in muscle stem cells, regenerating myonuclei, and fibro-adipogenic progenitor cells. Analysis of intercellular communication networks revealed fibro-adipogenic progenitors as a major signaling hub in PAD muscle, as well as deficiencies in angiogenic and bone morphogenetic protein signaling which may contribute to poor limb function in PAD. CONCLUSIONS This reference single-nuclei RNA-sequencing atlas provides a comprehensive analysis of the cell composition, transcriptional signature, and intercellular communication pathways that are altered in the PAD condition.
Collapse
Affiliation(s)
- Caroline G. Pass
- Department of Applied Physiology and Kinesiology (C.G.P., V.P., J.T., K.K., T.T., Q.Y., T.E.R.), The University of Florida, Gainesville
| | - Victoria Palzkill
- Department of Applied Physiology and Kinesiology (C.G.P., V.P., J.T., K.K., T.T., Q.Y., T.E.R.), The University of Florida, Gainesville
| | - Jianna Tan
- Department of Applied Physiology and Kinesiology (C.G.P., V.P., J.T., K.K., T.T., Q.Y., T.E.R.), The University of Florida, Gainesville
| | - Kyoungrae Kim
- Department of Applied Physiology and Kinesiology (C.G.P., V.P., J.T., K.K., T.T., Q.Y., T.E.R.), The University of Florida, Gainesville
| | - Trace Thome
- Department of Applied Physiology and Kinesiology (C.G.P., V.P., J.T., K.K., T.T., Q.Y., T.E.R.), The University of Florida, Gainesville
| | - Qingping Yang
- Department of Applied Physiology and Kinesiology (C.G.P., V.P., J.T., K.K., T.T., Q.Y., T.E.R.), The University of Florida, Gainesville
| | - Brian Fazzone
- Department of Surgery, Division of Vascular Surgery and Endovascular Therapy (B.F., S.T.R., K.A.O., S.T.S., S.A.B.), The University of Florida, Gainesville
- Malcom Randall VA Medical Center, Gainesville, FL (B.F., S.T.R., K.A.O., S.T.S., S.A.B.)
| | - Scott T. Robinson
- Department of Surgery, Division of Vascular Surgery and Endovascular Therapy (B.F., S.T.R., K.A.O., S.T.S., S.A.B.), The University of Florida, Gainesville
- Malcom Randall VA Medical Center, Gainesville, FL (B.F., S.T.R., K.A.O., S.T.S., S.A.B.)
| | - Kerri A. O’Malley
- Department of Surgery, Division of Vascular Surgery and Endovascular Therapy (B.F., S.T.R., K.A.O., S.T.S., S.A.B.), The University of Florida, Gainesville
- Malcom Randall VA Medical Center, Gainesville, FL (B.F., S.T.R., K.A.O., S.T.S., S.A.B.)
| | - Feng Yue
- Department of Animal Sciences (F.Y.), The University of Florida, Gainesville
- Myology Institute (F.Y., T.E.R.), The University of Florida, Gainesville
| | - Salvatore T. Scali
- Department of Surgery, Division of Vascular Surgery and Endovascular Therapy (B.F., S.T.R., K.A.O., S.T.S., S.A.B.), The University of Florida, Gainesville
- Malcom Randall VA Medical Center, Gainesville, FL (B.F., S.T.R., K.A.O., S.T.S., S.A.B.)
| | - Scott A. Berceli
- Department of Surgery, Division of Vascular Surgery and Endovascular Therapy (B.F., S.T.R., K.A.O., S.T.S., S.A.B.), The University of Florida, Gainesville
- Malcom Randall VA Medical Center, Gainesville, FL (B.F., S.T.R., K.A.O., S.T.S., S.A.B.)
| | - Terence E. Ryan
- Department of Applied Physiology and Kinesiology (C.G.P., V.P., J.T., K.K., T.T., Q.Y., T.E.R.), The University of Florida, Gainesville
- Center for Exercise Science (T.E.R.), The University of Florida, Gainesville
- Myology Institute (F.Y., T.E.R.), The University of Florida, Gainesville
| |
Collapse
|
7
|
Kumar A, Narkar VA. Nuclear receptors as potential therapeutic targets in peripheral arterial disease and related myopathy. FEBS J 2023; 290:4596-4613. [PMID: 35942640 PMCID: PMC9908775 DOI: 10.1111/febs.16593] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/28/2022] [Accepted: 08/08/2022] [Indexed: 12/31/2022]
Abstract
Peripheral arterial disease (PAD) is a prevalent cardiovascular complication of limb vascular insufficiency, causing ischemic injury, mitochondrial metabolic damage and functional impairment in the skeletal muscle, and ultimately leading to immobility and mortality. While potential therapies have been mostly focussed on revascularization, none of the currently available pharmacological treatments are fully effective in PAD, often leading to amputations, particularly in chronic metabolic diseases. One major limitation of focussed angiogenesis and revascularization as a therapeutic strategy is a limited effect on metabolic restoration and muscle regeneration in the affected limb. Therefore, additional preclinical investigations are needed to discover novel treatment options for PAD preferably targeting multiple aspects of muscle recovery. In this review, we propose nuclear receptors expressed in the skeletal muscle as potential candidates for ischemic muscle repair in PAD. We review classic steroid and orphan receptors that have been reported to be involved in the regulation of paracrine muscle angiogenesis, oxidative metabolism, mitochondrial biogenesis and muscle regeneration, and discuss how these receptors could be critical for recovery from ischemic muscle damage. Furthermore, we identify existing gaps in our understanding of nuclear receptor signalling in the skeletal muscle and propose future areas of research that could be instrumental in exploring nuclear receptors as therapeutic candidates for treating PAD.
Collapse
Affiliation(s)
- Ashok Kumar
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204
| | - Vihang A. Narkar
- Brown Foundation Institute of Molecular Medicine, UTHealth McGovern Medical School, Houston, TX, 77030
- University of Texas MD Anderson and UTHealth Graduate School of Biomedical Sciences, Houston, TX, 77030
| |
Collapse
|
8
|
Dong H, Tsai SY. Mitochondrial Properties in Skeletal Muscle Fiber. Cells 2023; 12:2183. [PMID: 37681915 PMCID: PMC10486962 DOI: 10.3390/cells12172183] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/16/2023] [Accepted: 08/24/2023] [Indexed: 09/09/2023] Open
Abstract
Mitochondria are the primary source of energy production and are implicated in a wide range of biological processes in most eukaryotic cells. Skeletal muscle heavily relies on mitochondria for energy supplements. In addition to being a powerhouse, mitochondria evoke many functions in skeletal muscle, including regulating calcium and reactive oxygen species levels. A healthy mitochondria population is necessary for the preservation of skeletal muscle homeostasis, while mitochondria dysregulation is linked to numerous myopathies. In this review, we summarize the recent studies on mitochondria function and quality control in skeletal muscle, focusing mainly on in vivo studies of rodents and human subjects. With an emphasis on the interplay between mitochondrial functions concerning the muscle fiber type-specific phenotypes, we also discuss the effect of aging and exercise on the remodeling of skeletal muscle and mitochondria properties.
Collapse
Affiliation(s)
- Han Dong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore;
| | - Shih-Yin Tsai
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore;
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
| |
Collapse
|
9
|
Sopariwala DH, Rios AS, Saley A, Kumar A, Narkar VA. Estrogen-Related Receptor Gamma Gene Therapy Promotes Therapeutic Angiogenesis and Muscle Recovery in Preclinical Model of PAD. J Am Heart Assoc 2023; 12:e028880. [PMID: 37548153 PMCID: PMC10492941 DOI: 10.1161/jaha.122.028880] [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] [Received: 11/17/2022] [Accepted: 06/30/2023] [Indexed: 08/08/2023]
Abstract
Background Peripheral arterial disease and critical limb ischemia are cardiovascular complications associated with vascular insufficiency, oxidative metabolic dysfunction, and myopathy in the limbs. Estrogen-related receptor gamma (ERRγ) has emerged as a dual regulator of paracrine angiogenesis and oxidative metabolism through transgenic mouse studies. Here our objective was to investigate whether postischemic intramuscular targeting of ERRγ via gene therapy promotes ischemic recovery in a preclinical model of peripheral arterial disease/critical limb ischemia. Methods and Results Adeno-associated virus 9 (AAV9) Esrrg gene delivery vector was developed and first tested via intramuscular injection in murine skeletal muscle. AAV9-Esrrg robustly increased ERRγ protein expression, induced angiogenic and oxidative genes, and boosted capillary density and succinate dehydrogenase oxidative metabolic activity in skeletal muscles of C57Bl/6J mice. Next, hindlimb ischemia was induced via unilateral femoral vessel ligation in mice, followed by intramuscular AAV9-Esrrg (or AAV9-green fluorescent protein) gene delivery 24 hours after injury. ERRγ overexpression increased ischemic neoangiogenesis and markers of endothelial activation, and significantly improved ischemic revascularization measured using laser Doppler flowmetry. Moreover, ERRγ overexpression restored succinate dehydrogenase oxidative metabolic capacity in ischemic muscle, which correlated with increased mitochondrial respiratory complex protein expression. Most importantly, myofiber size to number quantification revealed that AAV9-Esrrg restores myofibrillar size and mitigates ischemia-induced myopathy. Conclusions These results demonstrate that intramuscular AAV9-Esrrg delivery rescues ischemic pathology after hindlimb ischemia, underscoring that Esrrg gene therapy or pharmacological activation could be a promising strategy for the management of peripheral arterial disease/critical limb ischemia.
Collapse
Affiliation(s)
- Danesh H. Sopariwala
- Brown Foundation Institute of Molecular MedicineMcGovern Medical School at The University of Texas Health Science Center (UTHealth)HoustonTXUSA
| | - Andrea S. Rios
- Brown Foundation Institute of Molecular MedicineMcGovern Medical School at The University of Texas Health Science Center (UTHealth)HoustonTXUSA
| | - Addison Saley
- Department of BiosciencesRice UniversityHoustonTXUSA
| | - Ashok Kumar
- Department of Pharmacological and Pharmaceutical SciencesUniversity of HoustonTXUSA
| | - Vihang A. Narkar
- Brown Foundation Institute of Molecular MedicineMcGovern Medical School at The University of Texas Health Science Center (UTHealth)HoustonTXUSA
- Graduate School of Biomedical Sciences at UTHealthHoustonTXUSA
| |
Collapse
|
10
|
Sopariwala DH, Hao NTT, Narkar VA. Estrogen-related Receptor Signaling in Skeletal Muscle Fitness. Int J Sports Med 2023; 44:609-617. [PMID: 36787804 PMCID: PMC11168301 DOI: 10.1055/a-2035-8192] [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] [Indexed: 02/16/2023]
Abstract
Skeletal muscle is a highly plastic tissue that can alter its metabolic and contractile features, as well as regenerative potential in response to exercise and other conditions. Multiple signaling factors including metabolites, kinases, receptors, and transcriptional factors have been studied in the regulation of skeletal muscle plasticity. Recently, estrogen-related receptors (ERRs) have emerged as a critical transcriptional hub in control of skeletal muscle homeostasis. ERRα and ERRγ - the two highly expressed ERR sub-types in the muscle respond to various extracellular cues such as exercise, hypoxia, fasting and dietary factors, in turn regulating gene expression in the skeletal muscle. On the other hand, conditions such as diabetes and muscular dystrophy suppress expression of ERRs in the skeletal muscle, likely contributing to disease progression. We highlight key functions of ERRs in the skeletal muscle including the regulation of fiber type, mitochondrial metabolism, vascularization, and regeneration. We also describe how ERRs are regulated in the skeletal muscle, and their interaction with important muscle regulators (e. g. AMPK and PGCs). Finally, we identify critical gaps in our understanding of ERR signaling in the skeletal muscle, and suggest future areas of investigation to advance ERRs as potential targets for function promoting therapeutics in muscle diseases.
Collapse
Affiliation(s)
- Danesh H. Sopariwala
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School at The University of Texas Health Science Center (UTHealth), Houston, TX, USA
| | - Nguyen Thi Thu Hao
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School at The University of Texas Health Science Center (UTHealth), Houston, TX, USA
| | - Vihang A. Narkar
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School at The University of Texas Health Science Center (UTHealth), Houston, TX, USA
| |
Collapse
|
11
|
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: 0] [Impact Index Per Article: 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
|
12
|
GPR174 knockdown enhances blood flow recovery in hindlimb ischemia mice model by upregulating AREG expression. Nat Commun 2022; 13:7519. [PMID: 36473866 PMCID: PMC9727025 DOI: 10.1038/s41467-022-35159-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
Regulatory T cells (Tregs) are critically involved in neovascularization, an important compensatory mechanism in peripheral artery disease. The contribution of G protein coupled receptor 174 (GPR174), which is a regulator of Treg function and development, in neovascularization remains elusive. Here, we show that genetic deletion of GPR174 in Tregs potentiated blood flow recovery in mice after hindlimb ischemia. GPR174 deficiency upregulates amphiregulin (AREG) expression in Tregs, thereby enhancing endothelial cell functions and reducing pro-inflammatory macrophage polarization and endothelial cell apoptosis. Mechanically, GPR174 regulates AREG expression by inhibiting the nuclear accumulation of early growth response protein 1 (EGR1) via Gαs/cAMP/PKA signal pathway activation. Collectively, these findings demonstrate that GPR174 negatively regulates angiogenesis and vascular remodeling in response to ischemic injury and that GPR174 may be a potential molecular target for therapeutic interventions of ischemic vascular diseases.
Collapse
|
13
|
Wang W, Shang W, Zou J, Liu K, Liu M, Qiu X, Zhang H, Wang K, Wang N. ZNF667 facilitates angiogenesis after myocardial ischemia through transcriptional regulation of VASH1 and Wnt signaling pathway. Int J Mol Med 2022; 50:129. [PMID: 36043524 PMCID: PMC9448299 DOI: 10.3892/ijmm.2022.5185] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 08/09/2022] [Indexed: 11/24/2022] Open
Abstract
Zinc finger protein 667 (ZNF667, also referred as Mipu1), a widely expressed KRAB/C2H2-type zinc finger transcription factor, can protect against hypoxic-ischemic myocardial injury. Pro-angiogenesis is regarded as a promising strategy for the treatment of acute myocardial infarction (AMI). However, whether ZNF667 is involved in the angiogenesis following AMI remains to be elucidated. The present study reported that the expression of ZNF667 in CD31-positive endothelial cells (ECs) was upregulated in the heart of AMI mice. Hypoxic challenge (1% oxygen) promoted the mRNA and protein expression of ZNF667 in the human umbilical vein endothelial cells (HUVECs) in a time-dependent manner. Moreover, ZNF667 promoted hypoxia-induced invasion and tube formation of HUVECs. Mechanically, ZNF667 could directly bind to the promoter of anti-angiogenic gene VASH1 and inhibit its expression. Consequently, VASH1 overexpression abolished hypoxic challenge or ZNF667 overexpression-induced invasion and tube formation of HUVECs. Further bioinformatic analyses suggested that overexpression of ZNF667 or knockdown of VASH1-induced differentially expressed genes in HUVECs were greatly enriched in the Wnt signaling pathway (DAAM1, LEF1, RAC2, FRAT1, NFATc2 and WNT5A). Together, these data suggested that ZNF667 facilitates myocardial ischemia-driven angiogenesis through transcriptional repression of VASH1 and regulation of Wnt signaling pathway.
Collapse
Affiliation(s)
- Wenmei Wang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410008, P.R. China
| | - Weite Shang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410008, P.R. China
| | - Jiang Zou
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410008, P.R. China
| | - Ke Liu
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410008, P.R. China
| | - Meidong Liu
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410008, P.R. China
| | - Xiaoqin Qiu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Huali Zhang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410008, P.R. China
| | - Kangkai Wang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410008, P.R. China
| | - Nian Wang
- Department of Pathophysiology, School of Basic Medical Science, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
14
|
Sopariwala DH, Rios AS, Park MK, Song MS, Kumar A, Narkar VA. Estrogen-related receptor alpha is an AMPK-regulated factor that promotes ischemic muscle revascularization and recovery in diet-induced obese mice. FASEB Bioadv 2022; 4:602-618. [PMID: 36089981 PMCID: PMC9447423 DOI: 10.1096/fba.2022-00015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 06/05/2022] [Accepted: 06/08/2022] [Indexed: 01/27/2023] Open
Abstract
Obesity and type II diabetes are leading causes of peripheral arterial disease (PAD), which is characterized by vascular insufficiency and ischemic damage in the limb skeletal muscle. Glycemic control is not sufficient to prevent progression of PAD, and molecular targets that can promote muscle neo-angiogenesis in obesity and diabetes remain poorly defined. Here, we have investigated whether nuclear receptor estrogen-related receptor alpha (ERRα) can promote ischemic revascularization in the skeletal muscles of diet-induced obese (DIO) mice. Using muscle-specific ERRα transgenic mice, we found that ERRα overexpression promotes revascularization, marked by increased capillary staining and muscle perfusion in DIO mice after hindlimb ischemic injury. Furthermore, ERRα facilitates repair and restoration of skeletal muscle myofiber size after limb ischemia in DIO mice. The ameliorative effects of ERRα overexpression did not involve the prevention of weight gain, hyperglycemia or glucose/insulin intolerance, suggesting a direct role for ERRα in promoting angiogenesis. Interestingly, levels of endogenous ERRα protein are suppressed in the skeletal muscles of DIO mice compared to lean controls, coinciding with the suppression of angiogenic gene expression, and reduced AMPK signaling in the DIO skeletal muscles. Upon further investigating the link between AMPK and ERRα, we found that AMPK activation increases the expression and recruitment of ERRα protein to specific angiogenic gene promoters in muscle cells. Further, the induction of angiogenic factors by AMPK activators in muscle cells is blocked by repressing ERRα. In summary, our results identify an AMPK/ERRα-dependent angiogenic gene program in the skeletal muscle, which is repressed by DIO, and demonstrate that forced ERRα activation can promote ischemic revascularization and muscle recovery in obesity.
Collapse
Affiliation(s)
- Danesh H. Sopariwala
- Center for Metabolic & Degenerative DiseasesInstitute of Molecular Medicine, UTHealth McGovern Medical SchoolHoustonTexasUSA
| | - Andrea S. Rios
- Center for Metabolic & Degenerative DiseasesInstitute of Molecular Medicine, UTHealth McGovern Medical SchoolHoustonTexasUSA
| | - Mi Kyung Park
- Department of Molecular and Cellular OncologyThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Min Sup Song
- Department of Molecular and Cellular OncologyThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Ashok Kumar
- Department of Pharmacological and Pharmaceutical SciencesCollege of Pharmacy, University of HoustonHoustonTexasUSA
| | - Vihang A. Narkar
- Center for Metabolic & Degenerative DiseasesInstitute of Molecular Medicine, UTHealth McGovern Medical SchoolHoustonTexasUSA
| |
Collapse
|
15
|
Jia L, Zheng P, Wang H, Kang L, Wu H, Fu X. VEGF alleviates lower limb ischemia in diabetic mice by altering muscle fiber types. Exp Ther Med 2022; 23:251. [PMID: 35261623 PMCID: PMC8855503 DOI: 10.3892/etm.2022.11176] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 01/08/2021] [Indexed: 11/05/2022] Open
Abstract
Lower limb ischemia caused by diabetic foot (DF) is one of the most serious complications of diabetes. The therapeutic role of VEGF in DF is well documented. However, the mechanism for action of VEGF is still not clear. The present study aimed to explore the effects of VEGF-mediated skeletal muscle fiber type switch in angiogenesis and the treatment of DF. C57BL/6 mice housed in cages equipped with a voluntary running wheel were used to access VEGF protein level and citrate synthase activity (by ELISA) as well as muscle fiber type changes (by immunofluorescence) in the gastrocnemius muscle. C57BL/6 mice were fed on a high-fat diet for 6 weeks and then injected with streptozocin to induce diabetic lower limb ischemia model. Control adenovirus (Ad-GFP) or Ad-VEGF-GFP were then injected into the left gastrocnemius of the ischemic diabetic mice. Blood flow perfusion was examined by laser Doppler imaging at 1, 3, 7 and 14 days after adenovirus transduction. On day 14, all mice were anesthetized and sacrificed. VEGF expression levels, citrate synthase activity and muscle fiber type changes in the gastrocnemius muscle were assayed by ELISA and immunofluorescence analysis of myosin heavy chain IIa (MHCIIa) expression, respectively. Transwell assays were performed to determine whether VEGF-treated C2C12 myotubes played a role on tubule formation and migration of HUVECs. It was found that VEGF levels and citrate synthase activity were upregulated after voluntary exercise, along with the increased frequency of oxidized muscle fibers. Notably, adenovirus-mediated VEGF overexpression in the muscle also increased the frequency of oxidized (MHCIIa-positive) muscle fibers, enhanced citrate synthase activity and ameliorated lower limb ischemia in diabetic mice. VEGF treatment enhanced the phosphorylation of PI3K, Akt and AMPK (assayed by western blotting), as well as glucose consumption and metabolism (assayed by western blotting and glucose uptake assay), in the C2C12 myotubes. Interestingly, VEGF-treated C2C12 myotubes promoted the migration and tubule formation of HUVEC cells. The present findings suggest that skeletal muscle fiber conversion might be a potential approach for VEGF-mediated angiogenesis and disease treatment, which may provide new options for the prevention and treatment of DF.
Collapse
Affiliation(s)
- Lijing Jia
- Laboratory of Wound Healing and Cell Biology, Institute of Basic Medical Science, Chinese People's Liberation Army General Hospital, Beijing 100048, P.R. China
| | - Peilin Zheng
- Department of Endocrinology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong 518020, P.R. China
| | - Hongbo Wang
- Department of Respiratory Medicine, Tianjin Children's Hospital, Tianjin 300134, P.R. China
| | - Lin Kang
- Department of Endocrinology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong 518020, P.R. China
| | - Han Wu
- Department of Endocrinology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong 518020, P.R. China
| | - Xiaobing Fu
- Laboratory of Wound Healing and Cell Biology, Institute of Basic Medical Science, Chinese People's Liberation Army General Hospital, Beijing 100048, P.R. China
| |
Collapse
|
16
|
Nishida Y, Nawaz A, Hecht K, Tobe K. Astaxanthin as a Novel Mitochondrial Regulator: A New Aspect of Carotenoids, beyond Antioxidants. Nutrients 2021; 14:nu14010107. [PMID: 35010981 PMCID: PMC8746862 DOI: 10.3390/nu14010107] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/21/2021] [Accepted: 12/23/2021] [Indexed: 12/12/2022] Open
Abstract
Astaxanthin is a member of the carotenoid family that is found abundantly in marine organisms, and has been gaining attention in recent years due to its varied biological/physiological activities. It has been reported that astaxanthin functions both as a pigment, and as an antioxidant with superior free radical quenching capacity. We recently reported that astaxanthin modulated mitochondrial functions by a novel mechanism independent of its antioxidant function. In this paper, we review astaxanthin’s well-known antioxidant activity, and expand on astaxanthin’s lesser-known molecular targets, and its role in mitochondrial energy metabolism.
Collapse
Affiliation(s)
- Yasuhiro Nishida
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
- Fuji Chemical Industries, Co., Ltd., 55 Yokohoonji, Kamiich-machi, Nakaniikawa-gun, Toyama 930-0405, Japan
- Correspondence: (Y.N.); (A.N.); (K.T.)
| | - Allah Nawaz
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
- Correspondence: (Y.N.); (A.N.); (K.T.)
| | - Karen Hecht
- AstaReal, Inc., 3 Terri Lane, Unit 12, Burlington, NJ 08016, USA;
| | - Kazuyuki Tobe
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
- Correspondence: (Y.N.); (A.N.); (K.T.)
| |
Collapse
|
17
|
Nawaz A, Nishida Y, Takikawa A, Fujisaka S, Kado T, Aminuddin A, Bilal M, Jeelani I, Aslam MR, Nishimura A, Kuwano T, Watanabe Y, Igarashi Y, Okabe K, Ahmed S, Manzoor A, Usui I, Yagi K, Nakagawa T, Tobe K. Astaxanthin, a Marine Carotenoid, Maintains the Tolerance and Integrity of Adipose Tissue and Contributes to Its Healthy Functions. Nutrients 2021; 13:4374. [PMID: 34959926 PMCID: PMC8703397 DOI: 10.3390/nu13124374] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/04/2021] [Accepted: 12/05/2021] [Indexed: 12/12/2022] Open
Abstract
Recently, obesity-induced insulin resistance, type 2 diabetes, and cardiovascular disease have become major social problems. We have previously shown that Astaxanthin (AX), which is a natural antioxidant, significantly ameliorates obesity-induced glucose intolerance and insulin resistance. It is well known that AX is a strong lipophilic antioxidant and has been shown to be beneficial for acute inflammation. However, the actual effects of AX on chronic inflammation in adipose tissue (AT) remain unclear. To observe the effects of AX on AT functions in obese mice, we fed six-week-old male C57BL/6J on high-fat-diet (HFD) supplemented with or without 0.02% of AX for 24 weeks. We determined the effect of AX at 10 and 24 weeks of HFD with or without AX on various parameters including insulin sensitivity, glucose tolerance, inflammation, and mitochondrial function in AT. We found that AX significantly reduced oxidative stress and macrophage infiltration into AT, as well as maintaining healthy AT function. Furthermore, AX prevented pathological AT remodeling probably caused by hypoxia in AT. Collectively, AX treatment exerted anti-inflammatory effects via its antioxidant activity in AT, maintained the vascular structure of AT and preserved the stem cells and progenitor's niche, and enhanced anti-inflammatory hypoxia induction factor-2α-dominant hypoxic response. Through these mechanisms of action, it prevented the pathological remodeling of AT and maintained its integrity.
Collapse
Affiliation(s)
- Allah Nawaz
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan; (I.J.); (K.O.); (T.N.)
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan; (Y.N.); (A.T.); (S.F.); (T.K.); (A.A.); (M.B.); (M.R.A.); (A.N.); (T.K.); (Y.W.); (Y.I.); (K.Y.)
| | - Yasuhiro Nishida
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan; (Y.N.); (A.T.); (S.F.); (T.K.); (A.A.); (M.B.); (M.R.A.); (A.N.); (T.K.); (Y.W.); (Y.I.); (K.Y.)
- Fuji Chemical Industries, Co., Ltd., 55 Yokohoonji, Kamiich-machi, Nakaniikawa-gun, Toyama 930-0405, Japan
| | - Akiko Takikawa
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan; (Y.N.); (A.T.); (S.F.); (T.K.); (A.A.); (M.B.); (M.R.A.); (A.N.); (T.K.); (Y.W.); (Y.I.); (K.Y.)
| | - Shiho Fujisaka
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan; (Y.N.); (A.T.); (S.F.); (T.K.); (A.A.); (M.B.); (M.R.A.); (A.N.); (T.K.); (Y.W.); (Y.I.); (K.Y.)
| | - Tomonobu Kado
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan; (Y.N.); (A.T.); (S.F.); (T.K.); (A.A.); (M.B.); (M.R.A.); (A.N.); (T.K.); (Y.W.); (Y.I.); (K.Y.)
| | - Aminuddin Aminuddin
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan; (Y.N.); (A.T.); (S.F.); (T.K.); (A.A.); (M.B.); (M.R.A.); (A.N.); (T.K.); (Y.W.); (Y.I.); (K.Y.)
- Department of Nutrition, Faculty of Medicine, University of Hasanuddin, Makassar 90245, Indonesia
| | - Muhammad Bilal
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan; (Y.N.); (A.T.); (S.F.); (T.K.); (A.A.); (M.B.); (M.R.A.); (A.N.); (T.K.); (Y.W.); (Y.I.); (K.Y.)
| | - Ishtiaq Jeelani
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan; (I.J.); (K.O.); (T.N.)
| | - Muhammad Rahil Aslam
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan; (Y.N.); (A.T.); (S.F.); (T.K.); (A.A.); (M.B.); (M.R.A.); (A.N.); (T.K.); (Y.W.); (Y.I.); (K.Y.)
| | - Ayumi Nishimura
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan; (Y.N.); (A.T.); (S.F.); (T.K.); (A.A.); (M.B.); (M.R.A.); (A.N.); (T.K.); (Y.W.); (Y.I.); (K.Y.)
| | - Takahide Kuwano
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan; (Y.N.); (A.T.); (S.F.); (T.K.); (A.A.); (M.B.); (M.R.A.); (A.N.); (T.K.); (Y.W.); (Y.I.); (K.Y.)
| | - Yoshiyuki Watanabe
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan; (Y.N.); (A.T.); (S.F.); (T.K.); (A.A.); (M.B.); (M.R.A.); (A.N.); (T.K.); (Y.W.); (Y.I.); (K.Y.)
| | - Yoshiko Igarashi
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan; (Y.N.); (A.T.); (S.F.); (T.K.); (A.A.); (M.B.); (M.R.A.); (A.N.); (T.K.); (Y.W.); (Y.I.); (K.Y.)
| | - Keisuke Okabe
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan; (I.J.); (K.O.); (T.N.)
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan; (Y.N.); (A.T.); (S.F.); (T.K.); (A.A.); (M.B.); (M.R.A.); (A.N.); (T.K.); (Y.W.); (Y.I.); (K.Y.)
- Center for Clinical Research, Faculty of Medicine, Toyama University Hospital, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan
| | - Saeed Ahmed
- Department of Medicine and Surgery, Rawalpindi Medical University, Rawalpindi 46000, Pakistan;
| | | | - Isao Usui
- Department of Endocrinology and Metabolism, Dokkyo Medical University, Mibu 321-0293, Japan;
| | - Kunimasa Yagi
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan; (Y.N.); (A.T.); (S.F.); (T.K.); (A.A.); (M.B.); (M.R.A.); (A.N.); (T.K.); (Y.W.); (Y.I.); (K.Y.)
| | - Takashi Nakagawa
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan; (I.J.); (K.O.); (T.N.)
| | - Kazuyuki Tobe
- First Department of Internal Medicine, Faculty of Medicine, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan; (Y.N.); (A.T.); (S.F.); (T.K.); (A.A.); (M.B.); (M.R.A.); (A.N.); (T.K.); (Y.W.); (Y.I.); (K.Y.)
| |
Collapse
|
18
|
Fan Z, Turiel G, Ardicoglu R, Ghobrial M, Masschelein E, Kocijan T, Zhang J, Tan G, Fitzgerald G, Gorski T, Alvarado-Diaz A, Gilardoni P, Adams CM, Ghesquière B, De Bock K. Exercise-induced angiogenesis is dependent on metabolically primed ATF3/4 + endothelial cells. Cell Metab 2021; 33:1793-1807.e9. [PMID: 34358431 PMCID: PMC8432967 DOI: 10.1016/j.cmet.2021.07.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 05/18/2021] [Accepted: 07/14/2021] [Indexed: 12/21/2022]
Abstract
Exercise is a powerful driver of physiological angiogenesis during adulthood, but the mechanisms of exercise-induced vascular expansion are poorly understood. We explored endothelial heterogeneity in skeletal muscle and identified two capillary muscle endothelial cell (mEC) populations that are characterized by differential expression of ATF3/4. Spatial mapping showed that ATF3/4+ mECs are enriched in red oxidative muscle areas while ATF3/4low ECs lie adjacent to white glycolytic fibers. In vitro and in vivo experiments revealed that red ATF3/4+ mECs are more angiogenic when compared with white ATF3/4low mECs. Mechanistically, ATF3/4 in mECs control genes involved in amino acid uptake and metabolism and metabolically prime red (ATF3/4+) mECs for angiogenesis. As a consequence, supplementation of non-essential amino acids and overexpression of ATF4 increased proliferation of white mECs. Finally, deleting Atf4 in ECs impaired exercise-induced angiogenesis. Our findings illustrate that spatial metabolic angiodiversity determines the angiogenic potential of muscle ECs.
Collapse
Affiliation(s)
- Zheng Fan
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zürich), Zürich 8603, Switzerland
| | - Guillermo Turiel
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zürich), Zürich 8603, Switzerland
| | - Raphaela Ardicoglu
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zürich), Zürich 8603, Switzerland; Laboratory of Molecular and Behavioral Neuroscience, Department of Health Sciences and Technology, ETH Zürich, Zürich 8057, Switzerland
| | - Moheb Ghobrial
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zürich), Zürich 8603, Switzerland; Group Brain Vasculature and Perivascular Niche, Division of Experimental and Translational Neuroscience, Krembil Brain Institute, Krembil Research Institute, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, ON M5T 2S8, Canada
| | - Evi Masschelein
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zürich), Zürich 8603, Switzerland
| | - Tea Kocijan
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zürich), Zürich 8603, Switzerland
| | - Jing Zhang
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zürich), Zürich 8603, Switzerland
| | - Ge Tan
- Functional Genomics Center Zürich, ETH/University of Zürich, Zürich 8093, Switzerland
| | - Gillian Fitzgerald
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zürich), Zürich 8603, Switzerland
| | - Tatiane Gorski
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zürich), Zürich 8603, Switzerland
| | - Abdiel Alvarado-Diaz
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zürich), Zürich 8603, Switzerland
| | - Paola Gilardoni
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zürich), Zürich 8603, Switzerland
| | - Christopher M Adams
- Division of Endocrinology, Metabolism and Nutrition, Mayo Clinic, Rochester, MN 55905, USA
| | - Bart Ghesquière
- Metabolomics Expertise Center, VIB Center for Cancer Biology, VIB, Leuven, Belgium; Metabolomics Expertise Center, Department of Oncology, Cancer Institute, KU Leuven, Leuven, Belgium
| | - Katrien De Bock
- Laboratory of Exercise and Health, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH Zürich), Zürich 8603, Switzerland.
| |
Collapse
|
19
|
Sopariwala DH, Likhite N, Pei G, Haroon F, Lin L, Yadav V, Zhao Z, Narkar VA. Estrogen-related receptor α is involved in angiogenesis and skeletal muscle revascularization in hindlimb ischemia. FASEB J 2021; 35:e21480. [PMID: 33788962 PMCID: PMC11135633 DOI: 10.1096/fj.202001794rr] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 02/10/2021] [Accepted: 02/12/2021] [Indexed: 12/19/2022]
Abstract
Skeletal muscle ischemia is a major consequence of peripheral arterial disease (PAD) or critical limb ischemia (CLI). Although therapeutic options for resolving muscle ischemia in PAD/CLI are limited, the issue is compounded by poor understanding of the mechanisms driving muscle vascularization. We found that nuclear receptor estrogen-related receptor alpha (ERRα) expression is induced in murine skeletal muscle by hindlimb ischemia (HLI), and in cultured myotubes by hypoxia, suggesting a potential role for ERRα in ischemic response. To test this, we generated skeletal muscle-specific ERRα transgenic (TG) mice. In these mice, ERRα drives myofiber type switch from glycolytic type IIB to oxidative type IIA/IIX myofibers, which are typically associated with more vascular supply in muscle. Indeed, RNA sequencing and functional enrichment analysis of TG muscle revealed that "paracrine angiogenesis" is the top-ranked transcriptional program activated by ERRα in the skeletal muscle. Immunohistochemistry and angiography showed that ERRα overexpression increases baseline capillarity, arterioles and non-leaky blood vessel formation in the skeletal muscles. Moreover, ERRα overexpression facilitates ischemic neo-angiogenesis and perfusion recovery in hindlimb musculature of mice subjected to HLI. Therefore, ERRα is a hypoxia inducible nuclear receptor that is involved in skeletal muscle angiogenesis and could be potentially targeted for treating PAD/CLI.
Collapse
Affiliation(s)
- Danesh H. Sopariwala
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, TX, USA
| | - Neah Likhite
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, TX, USA
| | - Gungsheng Pei
- Center for Precision Medicine, School of Biomedical Informatics, UTHealth, Houston, TX, USA
| | - Fnu Haroon
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, TX, USA
| | - Lisa Lin
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, TX, USA
- Biochemistry and Cell Biology, Rice University, Houston, TX, USA
| | - Vikas Yadav
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, TX, USA
- Current address: Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Zhongming Zhao
- Center for Precision Medicine, School of Biomedical Informatics, UTHealth, Houston, TX, USA
- Human Genetics Center, School of Public Health, UTHealth, Houston, TX, USA
| | - Vihang A. Narkar
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, TX, USA
- Graduate School of Biomedical Sciences, UTHealth, TX, USA
| |
Collapse
|
20
|
Barlow J, Sfyri PP, Mitchell R, Verpoorten S, Scully D, Andreou C, Papadopoulos P, Patel K, Matsakas A. Platelet releasate normalises the compromised muscle regeneration in a mouse model of hyperlipidaemia. Exp Physiol 2021; 106:700-713. [PMID: 33450106 DOI: 10.1113/ep088937] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 01/12/2021] [Indexed: 12/19/2022]
Abstract
NEW FINDINGS What is the central question of this study? What is the impact of obesity-independent hyperlipidaemia on skeletal muscle stem cell function of ApoE-deficient (ApoE-/- ) mice? What is the main finding and its importance? Compromised muscle stem cell function accounts for the impaired muscle regeneration in hyperlipidaemic ApoE-/- mice. Importantly, impaired muscle regeneration is normalised by administration of platelet releasate. ABSTRACT Muscle satellite cells are important stem cells for skeletal muscle regeneration and repair after injury. ApoE-deficient mice, an established mouse model of hyperlipidaemia and atherosclerosis, show evidence of oxidative stress-induced lesions and fat infiltration in skeletal muscle followed by impaired repair after injury. However, the mechanisms underpinning attenuated muscle regeneration remain to be fully defined. Key to addressing the latter is to understand the properties of muscle stem cells from ApoE-deficient mice and their myogenic potential. Muscle stem cells from ApoE-deficient mice were cultured both ex vivo (on single fibres) and in vitro (primary myoblasts) and their myogenic capacity was determined. Skeletal muscle regeneration was studied on days 5 and 10 after cardiotoxin injury. ApoE-deficient muscle stem cells showed delayed activation and differentiation on single muscle fibres ex vivo. Impaired proliferation and differentiation profiles were also evident on isolated primary muscle stem cells in culture. ApoE-deficient mice displayed impaired skeletal muscle regeneration after acute injury in vivo. Administration of platelet releasate in ApoE-deficient mice reversed the deficits of muscle regeneration after acute injury to wild-type levels. These findings indicate that muscle stem cell myogenic potential is perturbed in skeletal muscle of a mouse model of hyperlipidaemia. We propose that platelet releasate could be a therapeutic intervention for conditions with associated myopathy such as peripheral arterial disease.
Collapse
Affiliation(s)
- Joseph Barlow
- Molecular Physiology Laboratory, Hull York Medical School, Centre for Atherothrombosis & Metabolic Disease, University of Hull, Hull, UK
| | - Pagona Panagiota Sfyri
- Molecular Physiology Laboratory, Hull York Medical School, Centre for Atherothrombosis & Metabolic Disease, University of Hull, Hull, UK
| | - Rob Mitchell
- School of Biological Sciences, University of Reading, Reading, UK
| | - Sandrine Verpoorten
- Molecular Physiology Laboratory, Hull York Medical School, Centre for Atherothrombosis & Metabolic Disease, University of Hull, Hull, UK
| | - David Scully
- Molecular Physiology Laboratory, Hull York Medical School, Centre for Atherothrombosis & Metabolic Disease, University of Hull, Hull, UK
| | - Charalampos Andreou
- Molecular Physiology Laboratory, Hull York Medical School, Centre for Atherothrombosis & Metabolic Disease, University of Hull, Hull, UK
| | - Petros Papadopoulos
- Department of Hematology, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid, Spain
| | - Ketan Patel
- School of Biological Sciences, University of Reading, Reading, UK
| | - Antonios Matsakas
- Molecular Physiology Laboratory, Hull York Medical School, Centre for Atherothrombosis & Metabolic Disease, University of Hull, Hull, UK
| |
Collapse
|
21
|
Adeyemo A, Johnson C, Stiene A, LaSance K, Qi Z, Lemen L, Schultz JEJ. Limb functional recovery is impaired in fibroblast growth factor-2 (FGF2) deficient mice despite chronic ischaemia-induced vascular growth. Growth Factors 2020; 38:75-93. [PMID: 32496882 PMCID: PMC8601595 DOI: 10.1080/08977194.2020.1767612] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 05/05/2020] [Indexed: 01/07/2023]
Abstract
FGF2 is a potent stimulator of vascular growth; however, even with a deficiency of FGF2 (Fgf2-/-), developmental vessel growth or ischaemia-induced revascularization still transpires. It remains to be elucidated as to what function, if any, FGF2 has during ischaemic injury. Wildtype (WT) or Fgf2-/- mice were subjected to hindlimb ischaemia for up to 42 days. Limb function, vascular growth, inflammatory- and angiogenesis-related proteins, and inflammatory cell infiltration were assessed in sham and ischaemic limbs at various timepoints. Recovery of ischaemic limb function was delayed in Fgf2-/- mice. Yet, vascular growth response to ischaemia was similar between WT and Fgf2-/- hindlimbs. Several angiogenesis- and inflammatory-related proteins (MCP-1, CXCL16, MMPs and PAI-1) were increased in Fgf2-/- ischaemic muscle. Neutrophil or monocyte recruitment/infiltration was elevated in Fgf2-/- ischaemic muscle. In summary, our study indicates that loss of FGF2 induces a pro-inflammatory microenvironment in skeletal muscle which exacerbates ischaemic injury and delays functional limb use.
Collapse
Affiliation(s)
- Adeola Adeyemo
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267
| | - Christopher Johnson
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267
| | - Andrew Stiene
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267
| | - Kathleen LaSance
- Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267
- Preclinical Imaging Core, University of Cincinnati College of Medicine, Cincinnati, OH 45267
| | - Zhihua Qi
- Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267
- Preclinical Imaging Core, University of Cincinnati College of Medicine, Cincinnati, OH 45267
| | - Lisa Lemen
- Department of Radiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267
- Preclinical Imaging Core, University of Cincinnati College of Medicine, Cincinnati, OH 45267
| | - Jo El J. Schultz
- Department of Pharmacology and Systems Physiology, University of Cincinnati College of Medicine, Cincinnati, OH 45267
| |
Collapse
|
22
|
Scully D, Sfyri P, Wilkinson HN, Acebes-Huerta A, Verpoorten S, Muñoz-Turrillas MC, Parnell A, Patel K, Hardman MJ, Gutiérrez L, Matsakas A. Optimising platelet secretomes to deliver robust tissue-specific regeneration. J Tissue Eng Regen Med 2019; 14:82-98. [PMID: 31603629 DOI: 10.1002/term.2965] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 09/04/2019] [Accepted: 09/09/2019] [Indexed: 12/19/2022]
Abstract
Promoting cell proliferation is the cornerstone of most tissue regeneration therapies. As platelet-based applications promote cell division and can be customised for tissue-specific efficacy, this makes them strong candidates for developing novel regenerative therapies. Therefore, the aim of this study was to determine if platelet releasate could be optimised to promote cellular proliferation and differentiation of specific tissues. Growth factors in platelet releasate were profiled for physiological and supraphysiological platelet concentrations. We analysed the effect of physiological and supraphysiological releasate on C2C12 skeletal myoblasts, H9C2 rat cardiomyocytes, human dermal fibroblasts (HDF), HaCaT keratinocytes, and chondrocytes. Cellular proliferation and differentiation were assessed through proliferation assays, mRNA, and protein expression. We show that supraphysiological releasate is not simply a concentrated version of physiological releasate. Physiological releasate promoted C2C12, HDF, and chondrocyte proliferation with no effect on H9C2 or HaCaT cells. Supraphysiological releasate induced stronger proliferation in C2C12 and HDF cells compared with physiological releasate. Importantly, supraphysiological releasate induced proliferation of H9C2 cells. The proliferative effects of skeletal and cardiac muscle cells were in part driven by vascular endothelial growth factor alpha. Furthermore, supraphysiological releasate induced differentiation of H9C2 and C2C12, HDF, and keratinocytes. This study provides insights into the ability of releasate to promote muscle, heart, skin, and cartilage cell proliferation and differentiation and highlights the importance of optimising releasate composition for tissue-specific regeneration.
Collapse
Affiliation(s)
- David Scully
- Molecular Physiology Laboratory, Centre for Atherothrombosis & Metabolic Disease, Hull York Medical School, University of Hull, Hull, UK
| | - Peggy Sfyri
- Molecular Physiology Laboratory, Centre for Atherothrombosis & Metabolic Disease, Hull York Medical School, University of Hull, Hull, UK
| | - Holly N Wilkinson
- Molecular Physiology Laboratory, Centre for Atherothrombosis & Metabolic Disease, Hull York Medical School, University of Hull, Hull, UK
| | - Andrea Acebes-Huerta
- Platelet Research Lab, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Sandrine Verpoorten
- Molecular Physiology Laboratory, Centre for Atherothrombosis & Metabolic Disease, Hull York Medical School, University of Hull, Hull, UK
| | - María Carmen Muñoz-Turrillas
- Platelet Research Lab, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain.,Centro Comunitario de Sangre y Tejidos, de Asturias, Oviedo, Spain
| | - Andrew Parnell
- School of Biological Sciences, University of Reading, Reading, UK
| | - Ketan Patel
- School of Biological Sciences, University of Reading, Reading, UK
| | - Matthew J Hardman
- Molecular Physiology Laboratory, Centre for Atherothrombosis & Metabolic Disease, Hull York Medical School, University of Hull, Hull, UK
| | - Laura Gutiérrez
- Platelet Research Lab, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain.,Department of Medicine, University of Oviedo, Oviedo, Spain
| | - Antonios Matsakas
- Molecular Physiology Laboratory, Centre for Atherothrombosis & Metabolic Disease, Hull York Medical School, University of Hull, Hull, UK
| |
Collapse
|
23
|
Ramachandran K, Senagolage MD, Sommars MA, Futtner CR, Omura Y, Allred AL, Barish GD. Dynamic enhancers control skeletal muscle identity and reprogramming. PLoS Biol 2019; 17:e3000467. [PMID: 31589602 PMCID: PMC6799888 DOI: 10.1371/journal.pbio.3000467] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 10/17/2019] [Accepted: 09/11/2019] [Indexed: 12/27/2022] Open
Abstract
Skeletal muscles consist of fibers of differing metabolic activities and contractility, which become remodeled in response to chronic exercise, but the epigenomic basis for muscle identity and adaptation remains poorly understood. Here, we used chromatin immunoprecipitation sequencing of dimethylated histone 3 lysine 4 and acetylated histone 3 lysine 27 as well as transposase-accessible chromatin profiling to dissect cis-regulatory networks across muscle groups. We demonstrate that in vivo enhancers specify muscles in accordance with myofiber composition, show little resemblance to cultured myotube enhancers, and identify glycolytic and oxidative muscle-specific regulators. Moreover, we find that voluntary wheel running and muscle-specific peroxisome proliferator-activated receptor gamma coactivator-1 alpha (Pgc1a) transgenic (mTg) overexpression, which stimulate endurance performance in mice, result in markedly different changes to the epigenome. Exercise predominantly leads to enhancer hypoacetylation, whereas mTg causes hyperacetylation at different sites. Integrative analysis of regulatory regions and gene expression revealed that exercise and mTg are each associated with myocyte enhancer factor (MEF) 2 and estrogen-related receptor (ERR) signaling and transcription of genes promoting oxidative metabolism. However, exercise was additionally associated with regulation by retinoid X receptor (RXR), jun proto-oncogene (JUN), sine oculis homeobox factor (SIX), and other factors. Overall, our work defines the unique enhancer repertoires of skeletal muscles in vivo and reveals that divergent exercise-induced or PGC1α-driven epigenomic programs direct partially convergent transcriptional networks.
Collapse
Affiliation(s)
- Krithika Ramachandran
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Madhavi D. Senagolage
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Meredith A. Sommars
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Christopher R. Futtner
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Yasuhiro Omura
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Amanda L. Allred
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
| | - Grant D. Barish
- Department of Medicine, Division of Endocrinology, Metabolism, and Molecular Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, United States of America
- Jesse Brown VA Medical Center, Chicago, Illinois, United States of America
| |
Collapse
|
24
|
Likhite N, Yadav V, Milliman EJ, Sopariwala DH, Lorca S, Narayana NP, Sheth M, Reineke EL, Giguère V, Narkar V. Loss of Estrogen-Related Receptor Alpha Facilitates Angiogenesis in Endothelial Cells. Mol Cell Biol 2019; 39:e00411-18. [PMID: 30602497 PMCID: PMC6379583 DOI: 10.1128/mcb.00411-18] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 09/04/2018] [Accepted: 12/11/2018] [Indexed: 01/08/2023] Open
Abstract
Estrogen-related receptors (ERRs) have emerged as major metabolic regulators in various tissues. However, their expression and function in the vasculature remains unknown. Here, we report the transcriptional program and cellular function of ERRα in endothelial cells (ECs), a cell type with a multifaceted role in vasculature. Of the three ERR subtypes, ECs exclusively express ERRα. Gene expression profiling of ECs lacking ERRα revealed that ERRα predominantly acts as a transcriptional repressor, targeting genes linked with angiogenesis, cell migration, and cell adhesion. ERRα-deficient ECs exhibit decreased proliferation but increased migration and tube formation. ERRα depletion increased basal as well as vascular endothelial growth factor A (VEGFA)- and ANG1/2-stimulated angiogenic sprouting in endothelial spheroids. Moreover, retinal angiogenesis is enhanced in ERRα knockout mice compared to that in wild-type mice. Surprisingly, ERRα is dispensable for the regulation of its classic targets, such as metabolism, mitochondrial biogenesis, and cellular respiration in the ECs. ERRα is enriched at the promoters of angiogenic, migratory, and cell adhesion genes. Further, VEGFA increased ERRα recruitment to angiogenesis-associated genes and simultaneously decreased their expression. Despite increasing its gene occupancy, proangiogenic stimuli decrease ERRα expression in ECs. Our work shows that endothelial ERRα plays a repressive role in angiogenesis and potentially fine-tunes growth factor-mediated angiogenesis.
Collapse
Affiliation(s)
- Neah Likhite
- Metabolic and Degenerative Diseases, Institute of Molecular Medicine, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Vikas Yadav
- Metabolic and Degenerative Diseases, Institute of Molecular Medicine, The University of Texas McGovern Medical School, Houston, Texas, USA
| | | | - Danesh H Sopariwala
- Metabolic and Degenerative Diseases, Institute of Molecular Medicine, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Sabina Lorca
- Metabolic and Degenerative Diseases, Institute of Molecular Medicine, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Nithya P Narayana
- Metabolic and Degenerative Diseases, Institute of Molecular Medicine, The University of Texas McGovern Medical School, Houston, Texas, USA
| | - Megha Sheth
- Metabolic and Degenerative Diseases, Institute of Molecular Medicine, The University of Texas McGovern Medical School, Houston, Texas, USA
- Department of Biochemistry and Cell Biology, Rice University, Houston, Texas, USA
| | - Erin L Reineke
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, Texas, USA
| | - Vincent Giguère
- Department of Biochemistry, Medicine and Oncology, Faculty of Medicine, Goodman Cancer Research Centre, McGill University, Montreal, Canada
| | - Vihang Narkar
- Metabolic and Degenerative Diseases, Institute of Molecular Medicine, The University of Texas McGovern Medical School, Houston, Texas, USA
- Integrative Biology and Pharmacology, The University of Texas McGovern Medical School, Houston, Texas, USA
- Graduate School of Biomedical Sciences at The University of Texas Health Science Center, Houston, Texas, USA
| |
Collapse
|
25
|
Scully D, Sfyri P, Verpoorten S, Papadopoulos P, Muñoz‐Turrillas MC, Mitchell R, Aburima A, Patel K, Gutiérrez L, Naseem KM, Matsakas A. Platelet releasate promotes skeletal myogenesis by increasing muscle stem cell commitment to differentiation and accelerates muscle regeneration following acute injury. Acta Physiol (Oxf) 2019; 225:e13207. [PMID: 30339324 DOI: 10.1111/apha.13207] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 09/27/2018] [Accepted: 10/14/2018] [Indexed: 12/11/2022]
Abstract
AIM The use of platelets as biomaterials has gained intense research interest. However, the mechanisms regarding platelet-mediated skeletal myogenesis remain to be established. The aim of this study was to determine the role of platelet releasate in skeletal myogenesis and muscle stem cell fate in vitro and ex vivo respectively. METHODS We analysed the effect of platelet releasate on proliferation and differentiation of C2C12 myoblasts by means of cell proliferation assays, immunohistochemistry, gene expression and cell bioenergetics. We expanded in vitro findings on single muscle fibres by determining the effect of platelet releasate on murine skeletal muscle stem cells using protein expression profiles for key myogenic regulatory factors. RESULTS TRAP6 and collagen used for releasate preparation had a more pronounced effect on myoblast proliferation vs thrombin and sonicated platelets (P < 0.05). In addition, platelet concentration positively correlated with myoblast proliferation. Platelet releasate increased myoblast and muscle stem cell proliferation in a dose-dependent manner, which was mitigated by VEGFR and PDGFR inhibition. Inhibition of VEGFR and PDGFR ablated MyoD expression on proliferating muscle stem cells, compromising their commitment to differentiation in muscle fibres (P < 0.001). Platelet releasate was detrimental to myoblast fusion and affected differentiation of myoblasts in a temporal manner. Most importantly, we show that platelet releasate promotes skeletal myogenesis through the PDGF/VEGF-Cyclin D1-MyoD-Scrib-Myogenin axis and accelerates skeletal muscle regeneration after acute injury. CONCLUSION This study provides novel mechanistic insights on the role of platelet releasate in skeletal myogenesis and set the physiological basis for exploiting platelets as biomaterials in regenerative medicine.
Collapse
Affiliation(s)
- David Scully
- Molecular Physiology Laboratory, Centre for Atherothrombotic and Metabolic Disease, Hull York Medical School University of Hull Hull UK
| | - Peggy Sfyri
- Molecular Physiology Laboratory, Centre for Atherothrombotic and Metabolic Disease, Hull York Medical School University of Hull Hull UK
| | - Sandrine Verpoorten
- Molecular Physiology Laboratory, Centre for Atherothrombotic and Metabolic Disease, Hull York Medical School University of Hull Hull UK
| | - Petros Papadopoulos
- Department of Hematology, Instituto de Investigación Sanitaria San Carlos (IdISSC) Hospital Clínico San Carlos Madrid Spain
| | - María Carmen Muñoz‐Turrillas
- Centro Comunitario de Sangre y Tejidos de Asturias and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA) Oviedo Spain
| | - Robert Mitchell
- School of Biological Sciences University of Reading Reading UK
| | - Ahmed Aburima
- Molecular Physiology Laboratory, Centre for Atherothrombotic and Metabolic Disease, Hull York Medical School University of Hull Hull UK
| | - Ketan Patel
- School of Biological Sciences University of Reading Reading UK
| | - Laura Gutiérrez
- Department of Medicine Universidad de Oviedo and Instituto de Investigación Sanitaria del Principado de Asturias (ISPA) Oviedo Spain
| | - Khalid M. Naseem
- Leeds Institute of Cardiovascular and Metabolic Medicine University of Leeds Leeds UK
| | - Antonios Matsakas
- Molecular Physiology Laboratory, Centre for Atherothrombotic and Metabolic Disease, Hull York Medical School University of Hull Hull UK
| |
Collapse
|
26
|
Chen L, Yang Y, Zhang L, Li C, Coffie JW, Geng X, Qiu L, You X, Fang Z, Song M, Gao X, Wang H. Aucubin promotes angiogenesis via estrogen receptor beta in a mouse model of hindlimb ischemia. J Steroid Biochem Mol Biol 2017; 172:149-159. [PMID: 28711487 DOI: 10.1016/j.jsbmb.2017.07.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 07/10/2017] [Accepted: 07/11/2017] [Indexed: 11/19/2022]
Abstract
Aucubin (AU) is an iridoid glycoside that has been shown to display estrogenic properties and has various pharmacological effects. Herein, we described the angiogenic properties of AU. In the study, hindlimb ischemia was induced by ligation of femoral artery on the right leg of ovariectomized mice. AU treatment significantly accelerated perfusion recovery and reduced tissue injury in mice muscle. Quantification of CD31-positive vessels in hindlimb muscles provided evidences that AU promoted angiogenesis in peripheral ischemia. In addition, results from quantitative PCR and western blot suggested AU induced angiogenesis via vascular endothelial cell growth factor (VEGF)/Akt/endothelial nitric oxide synthase (eNOS) signaling pathway. More interestingly, AU's angiogenic effects could be completely abolished in estrogen receptor beta (ERβ) knockout mice. In conclusion, the underlying mechanisms were elucidated that AU produced pro-angiogenic effects through ERβ-mediated VEGF signaling pathways. These results expand knowledge about the beneficial effects of AU in angiogenesis and blood flow recovery. It might provide insight into the ERβ regulating neovascularisation in hindlimb ischemia and identify AU as a potent new compound used for the treatment of peripheral vascular disease.
Collapse
Affiliation(s)
- Lu Chen
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China; School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yue Yang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formula, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Lusha Zhang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formula, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Chunxiao Li
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formula, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin, China
| | - Joel Wake Coffie
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formula, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiao Geng
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formula, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin, China
| | - Lizhen Qiu
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formula, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin, China
| | - Xingyu You
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formula, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin, China
| | - Zhirui Fang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formula, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Min Song
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formula, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiumei Gao
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formula, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China; Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin, China
| | - Hong Wang
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin, China; Tianjin Key Laboratory of Traditional Chinese Medicine Pharmacology, Tianjin, China; School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| |
Collapse
|
27
|
Sopariwala DH, Yadav V, Badin PM, Likhite N, Sheth M, Lorca S, Vila IK, Kim ER, Tong Q, Song MS, Rodney GG, Narkar VA. Long-term PGC1β overexpression leads to apoptosis, autophagy and muscle wasting. Sci Rep 2017; 7:10237. [PMID: 28860475 PMCID: PMC5578977 DOI: 10.1038/s41598-017-10238-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 08/01/2017] [Indexed: 12/22/2022] Open
Abstract
Skeletal muscle wasting is prevalent in many chronic diseases, necessitating inquiries into molecular regulation of muscle mass. Nuclear receptor co-activator peroxisome proliferator-activated receptor co-activator 1 alpha (PGC1α) and its splice variant PGC1α4 increase skeletal muscle mass. However, the effect of the other PGC1 sub-type, PGC1β, on muscle size is unclear. In transgenic mice selectively over-expressing PGC1β in the skeletal muscle, we have found that PGC1β progressively decreases skeletal muscle mass predominantly associated with loss of type 2b fast-twitch myofibers. Paradoxically, PGC1β represses the ubiquitin-proteolysis degradation pathway genes resulting in ubiquitinated protein accumulation in muscle. However, PGC1β overexpression triggers up-regulation of apoptosis and autophagy genes, resulting in robust activation of these cell degenerative processes, and a concomitant increase in muscle protein oxidation. Concurrently, PGC1β up-regulates apoptosis and/or autophagy transcriptional factors such as E2f1, Atf3, Stat1, and Stat3, which may be facilitating myopathy. Therefore, PGC1β activation negatively affects muscle mass over time, particularly fast-twitch muscles, which should be taken into consideration along with its known aerobic effects in the skeletal muscle.
Collapse
Affiliation(s)
- Danesh H Sopariwala
- Metabolic and Degenerative Diseases, Institute of Molecular Medicine, The University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Vikas Yadav
- Metabolic and Degenerative Diseases, Institute of Molecular Medicine, The University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Pierre-Marie Badin
- Metabolic and Degenerative Diseases, Institute of Molecular Medicine, The University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Neah Likhite
- Metabolic and Degenerative Diseases, Institute of Molecular Medicine, The University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Megha Sheth
- Metabolic and Degenerative Diseases, Institute of Molecular Medicine, The University of Texas McGovern Medical School, Houston, TX, 77030, USA
- Department of Biochemistry and Cell Biology, Rice University, Houston, TX, 77005, USA
| | - Sabina Lorca
- Metabolic and Degenerative Diseases, Institute of Molecular Medicine, The University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Isabelle K Vila
- Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Eun Ran Kim
- Metabolic and Degenerative Diseases, Institute of Molecular Medicine, The University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Qingchun Tong
- Metabolic and Degenerative Diseases, Institute of Molecular Medicine, The University of Texas McGovern Medical School, Houston, TX, 77030, USA
| | - Min Sup Song
- Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Graduate School of Biomedical Sciences at The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - George G Rodney
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Vihang A Narkar
- Metabolic and Degenerative Diseases, Institute of Molecular Medicine, The University of Texas McGovern Medical School, Houston, TX, 77030, USA.
- Graduate School of Biomedical Sciences at The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
- Integrative Biology and Pharmacology, The University of Texas McGovern Medical School, Houston, TX, 77030, USA.
| |
Collapse
|
28
|
Misra J, Kim DK, Choi HS. ERRγ: a Junior Orphan with a Senior Role in Metabolism. Trends Endocrinol Metab 2017; 28:261-272. [PMID: 28209382 DOI: 10.1016/j.tem.2016.12.005] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 12/18/2016] [Accepted: 12/22/2016] [Indexed: 01/01/2023]
Abstract
Estrogen-related receptor (ERR)γ is an orphan nuclear hormone receptor that belongs to the ERR subfamily of transcription factors. No endogenous ligand has been identified to date. ERRγ possesses ligand-independent transcriptional activity that is regulated by co-regulator interactions, and post-translational modifications (PTMs). Recent data from animal models have established ERRγ as a crucial mediator of multiple endocrine and metabolic signals. ERRγ plays important roles in pathological conditions such as insulin resistance, alcoholic liver injury, and cardiac hypertrophy, and controls energy metabolism in the heart, skeletal muscle, and pancreatic β cells. These findings corroborate the importance of ERRγ in metabolic homeostasis, and suggest that ERRγ is a good target for the treatment of metabolic diseases.
Collapse
Affiliation(s)
- Jagannath Misra
- National Creative Research Initiatives Center for Nuclear Receptor Signals and Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Don-Kyu Kim
- Department of Molecular Biotechnology, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Hueng-Sik Choi
- National Creative Research Initiatives Center for Nuclear Receptor Signals and Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju 61186, Republic of Korea.
| |
Collapse
|
29
|
Diao YP, Cui FK, Yan S, Chen ZG, Lian LS, Guo LL, Li YJ. Nerve Growth Factor Promotes Angiogenesis and Skeletal Muscle Fiber Remodeling in a Murine Model of Hindlimb Ischemia. Chin Med J (Engl) 2017; 129:313-9. [PMID: 26831234 PMCID: PMC4799576 DOI: 10.4103/0366-6999.174496] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Background: Therapeutic angiogenesis has been shown to promote blood vessel growth and improve tissue perfusion. Nerve growth factor (NGF) has been reported to play an important role in both physiological and pathological angiogenesis. This study aimed to investigate the effects of NGF on angiogenesis and skeletal muscle fiber remodeling in a murine model of hindlimb ischemia and study the relationship between NGF and vascular endothelial growth factor (VEGF) in angiogenesis. Methods: Twenty-four mice were randomly allocated to normal control group (n = 6), blank control group (n = 6), VEGF gene transfection group (n = 6), and NGF gene transfection group (n = 6). The model of left hindlimb ischemia model was established by ligating the femoral artery. VEGF165 plasmid (125 μg) and NGF plasmid (125 μg) was injected into the ischemic gastrocnemius of mice from VEGF group and NGF group, respectively. Left hindlimb function and ischemic damage were assessed with terminal points at 21th day postischemia induction. The gastrocnemius of four groups was tested by hematoxylin-eosin staining, proliferating cell nuclear antigen and CD34 immunohistochemistry staining, and myosin ATPase staining. NGF and VEGF protein expression was detected by enzyme-linked immunosorbent assay. Results: On the 21th day after surgery, the functional assessment score and skeletal muscle atrophy degree of VEGF group and NGF group were significantly lower than those of normal control group and blank control group. The endothelial cell proliferation index and the capillary density of VEGF group and NGF group were significantly increased compared with normal control group and blank control group (P < 0.05). The NGF and VEGF protein expression of NGF group showed a significant rise when compared with blank control group (P < 0.05). Similarly, the VEGF protein expression of VEGF group was significantly higher than that of blank control group (P < 0.05), but there was no significant difference of the NGF protein expression between VEGF group and blank control group (P > 0.05). The type I skeletal muscle fiber proportion in gastrocnemius of NGF group and VEGF group was significantly higher than that of blank control group (P < 0.05). Conclusions: NGF transfection can promote NGF and VEGF protein expression which not only can induce angiogenesis but also induce type I muscle fiber expression in ischemic limbs.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yong-Jun Li
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Science, Beijing 100730, China
| |
Collapse
|
30
|
Badin PM, Sopariwala DH, Lorca S, Narkar VA. Muscle Arnt/Hif1β Is Dispensable in Myofiber Type Determination, Vascularization and Insulin Sensitivity. PLoS One 2016; 11:e0168457. [PMID: 28005939 PMCID: PMC5178999 DOI: 10.1371/journal.pone.0168457] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 12/01/2016] [Indexed: 02/07/2023] Open
Abstract
Aryl Hydrocarbon Receptor Nuclear Translocator/ hypoxia-inducible factor 1 beta (ARNT/ HIF1β), a member of bHLH-PAS family of transcriptional factors, plays a critical role in metabolic homeostasis, insulin resistance and glucose intolerance. The contributions of ARNT in pancreas, liver and adipose tissue to energy balance through gene regulation have been described. Surprisingly, the impact of ARNT signaling in the skeletal muscles, one of the major organs involved in glucose disposal, has not been investigated, especially in type II diabetes. Here we report that ARNT is expressed in the skeletal muscles, particularly in the energy-efficient oxidative slow-twitch myofibers, which are characterized by increased oxidative capacity, mitochondrial content, vascular supply and insulin sensitivity. However, muscle-specific deletion of ARNT did not change myofiber type distribution, oxidative capacity, mitochondrial content, capillarity, or the expression of genes associated with these features. Consequently, the lack of ARNT in the skeletal muscle did not affect weight gain, lean/fat mass, insulin sensitivity and glucose tolerance in lean mice, nor did it impact insulin resistance and glucose intolerance in high fat diet-induced obesity. Therefore, skeletal muscle ARNT is dispensable for controlling muscle fiber type and metabolic regulation, as well as diet-induced weight control, insulin sensitivity and glucose tolerance.
Collapse
Affiliation(s)
- Pierre-Marie Badin
- Metabolic and Degenerative Diseases, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, TX, United States of America
| | - Danesh H. Sopariwala
- Metabolic and Degenerative Diseases, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, TX, United States of America
| | - Sabina Lorca
- Metabolic and Degenerative Diseases, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, TX, United States of America
| | - Vihang A. Narkar
- Metabolic and Degenerative Diseases, Brown Foundation Institute of Molecular Medicine, McGovern Medical School, UTHealth, Houston, TX, United States of America
- Integrative Biology and Pharmacology, McGovern Medical School, UTHealth, Houston, TX, United States of America
- Graduate School of Biomedical Sciences, McGovern Medical School, UTHealth, Houston, TX, United States of America
- * E-mail:
| |
Collapse
|
31
|
Omairi S, Matsakas A, Degens H, Kretz O, Hansson KA, Solbrå AV, Bruusgaard JC, Joch B, Sartori R, Giallourou N, Mitchell R, Collins-Hooper H, Foster K, Pasternack A, Ritvos O, Sandri M, Narkar V, Swann JR, Huber TB, Patel K. Enhanced exercise and regenerative capacity in a mouse model that violates size constraints of oxidative muscle fibres. eLife 2016; 5. [PMID: 27494364 PMCID: PMC4975572 DOI: 10.7554/elife.16940] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 07/19/2016] [Indexed: 12/28/2022] Open
Abstract
A central tenet of skeletal muscle biology is the existence of an inverse relationship between the oxidative fibre capacity and its size. However, robustness of this relationship is unknown. We show that superimposition of Estrogen-related receptor gamma (Errγ) on the myostatin (Mtn) mouse null background (Mtn-/-/ErrγTg/+) results in hypertrophic muscle with a high oxidative capacity thus violating the inverse relationship between fibre size and oxidative capacity. We also examined the canonical view that oxidative muscle phenotype positively correlate with Satellite cell number, the resident stem cells of skeletal muscle. Surprisingly, hypertrophic fibres from Mtn-/-/ErrγTg/+ mouse showed satellite cell deficit which unexpectedly did not affect muscle regeneration. These observations 1) challenge the concept of a constraint between fibre size and oxidative capacity and 2) indicate the important role of the microcirculation in the regenerative capacity of a muscle even when satellite cell numbers are reduced. DOI:http://dx.doi.org/10.7554/eLife.16940.001
Collapse
Affiliation(s)
- Saleh Omairi
- School of Biological Sciences, University of Reading, Reading, United Kingdom
| | | | - Hans Degens
- School of Healthcare Science, Manchester Metropolitan University, Manchester, United Kingdom.,Lithuanian Sports University, Kaunas, Lithuania
| | - Oliver Kretz
- Renal Division, University Medical Center Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Kenth-Arne Hansson
- Centre for Integrative Neuroplasticity, Department of Biosciences, University of Oslo, Oslo, Norway
| | - Andreas Våvang Solbrå
- Centre for Integrative Neuroplasticity, Department of Biosciences, University of Oslo, Oslo, Norway.,Department of Physics, University of Oslo, Oslo, Norway
| | - Jo C Bruusgaard
- Centre for Integrative Neuroplasticity, Department of Biosciences, University of Oslo, Oslo, Norway.,Department of Health Sciences, Kristiania University College, Oslo, Norway
| | - Barbara Joch
- Department of Neuroanatomy, University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Roberta Sartori
- Venetian Institute of Molecular Medicine, University of Padua, Padua, Italy
| | - Natasa Giallourou
- Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | - Robert Mitchell
- School of Biological Sciences, University of Reading, Reading, United Kingdom
| | | | - Keith Foster
- School of Biological Sciences, University of Reading, Reading, United Kingdom
| | - Arja Pasternack
- Department of Bacteriology and Immunology, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Olli Ritvos
- Department of Bacteriology and Immunology, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Marco Sandri
- Venetian Institute of Molecular Medicine, University of Padua, Padua, Italy
| | - Vihang Narkar
- Institute of Molecular Medicine, University of Health Science Center, Houston, Texas
| | - Jonathan R Swann
- Department of Food and Nutritional Sciences, University of Reading, Reading, United Kingdom
| | - Tobias B Huber
- Renal Division, University Medical Center Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany.,BIOSS Center for Biological Signalling Studies, Albert-Ludwigs-University Freiburg, Houston, Texas.,FRIAS, Freiburg Institute for Advanced Studies and Center for Biological System Analysis ZBSA, Freiburg, Germany
| | - Ketan Patel
- School of Biological Sciences, University of Reading, Reading, United Kingdom.,FRIAS, Freiburg Institute for Advanced Studies and Center for Biological System Analysis ZBSA, Freiburg, Germany
| |
Collapse
|
32
|
Exercise-like effects by Estrogen-related receptor-gamma in muscle do not prevent insulin resistance in db/db mice. Sci Rep 2016; 6:26442. [PMID: 27220353 PMCID: PMC4879541 DOI: 10.1038/srep26442] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 04/29/2016] [Indexed: 12/11/2022] Open
Abstract
Dissecting exercise-mimicking pathways that can replicate the benefits of exercise in obesity and diabetes may lead to promising treatments for metabolic disorders. Muscle estrogen-related receptor gamma (ERRγ) is induced by exercise, and when over-expressed in the skeletal muscle mimics exercise by stimulating glycolytic-to-oxidative myofiber switch, mitochondrial biogenesis and angiogenesis in lean mice. The objective of this study was to test whether muscle ERRγ in obese mice mitigates weight gain and insulin resistance. To do so, ERRγ was selectively over-expressed in the skeletal muscle of obese and diabetic db/db mice. Muscle ERRγ over-expression successfully triggered glycolytic-to-oxidative myofiber switch, increased functional mitochondrial content and boosted vascular supply in the db/db mice. Despite aerobic remodeling, ERRγ surprisingly failed to improve whole-body energy expenditure, block muscle accumulation of triglycerides, toxic diacylglycerols (DAG) and ceramides or suppress muscle PKCε sarcolemmal translocation in db/db mice. Consequently, muscle ERRγ did not mitigate impaired muscle insulin signaling or insulin resistance in these mice. In conclusion, obesity and diabetes in db/db mice are not amenable to selective ERRγ-directed programming of classic exercise-like effects in the skeletal muscle. Other biochemical pathways or integrated whole-body effects of exercise may be critical for resisting diabetes and obesity.
Collapse
|
33
|
Electrical stimulation influences chronic intermittent hypoxia-hypercapnia induction of muscle fibre transformation by regulating the microRNA/Sox6 pathway. Sci Rep 2016; 6:26415. [PMID: 27199002 PMCID: PMC4873781 DOI: 10.1038/srep26415] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 05/03/2016] [Indexed: 01/07/2023] Open
Abstract
Chronic obstructive pulmonary disease can cause muscle fibre transformation due to chronic intermittent hypoxia-hypercapnia (CIHH). Studies have shown that high expression of Sox6 in muscle could suppress type-I fibres through downregulating the PPARβ (peroxisome proliferator-activated receptor β)/ERRγ (oestrogen-related receptor γ)/microRNA pathway. However, whether this pathway is involved in CIHH-induced muscle fibre transformation is unknown. Electrical stimulation (ES) is an effective approach to ameliorate muscle dysfunction. Here, we explored the effects of ES on CIHH-induced muscle fibre transformation and the microRNA/Sox6 pathway. After CIHH exposure, both the soleus (SOL) and gastrocnemius (GC) muscles showed decreased type-I fibres. The PPARβ/ERRγ/mir-499&208b (PEM, for GC) and PPARβ/mir-499&208b (PM, for SOL) signalling cascades were suppressed, followed by elevated Sox6 expression. Low frequency electrical stimulation (LFES) activated the PEM/PM pathway and enhanced type-I fibre numbers through suppressing Sox6 in SOL and GC. High frequency electrical stimulation (HFES) promoted type-I fibre expression through activating the PEM pathway in GC. Although PPARβ expression and type-I fibres were suppressed in SOL after HFES, no significant change was found in mir-499&208b/Sox6 expression. These results suggest that the microRNA/Sox6 pathway is disturbed after CIHH. Both low and high frequency electrical stimulations induce muscle fibre transformation partly through regulating the microRNA/Sox6 pathway.
Collapse
|
34
|
Cho Y, Hazen BC, Gandra PG, Ward SR, Schenk S, Russell AP, Kralli A. Perm1 enhances mitochondrial biogenesis, oxidative capacity, and fatigue resistance in adult skeletal muscle. FASEB J 2015; 30:674-87. [PMID: 26481306 DOI: 10.1096/fj.15-276360] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 09/28/2015] [Indexed: 12/14/2022]
Abstract
Skeletal muscle mitochondrial content and oxidative capacity are important determinants of muscle function and whole-body health. Mitochondrial content and function are enhanced by endurance exercise and impaired in states or diseases where muscle function is compromised, such as myopathies, muscular dystrophies, neuromuscular diseases, and age-related muscle atrophy. Hence, elucidating the mechanisms that control muscle mitochondrial content and oxidative function can provide new insights into states and diseases that affect muscle health. In past studies, we identified Perm1 (PPARGC1- and ESRR-induced regulator, muscle 1) as a gene induced by endurance exercise in skeletal muscle, and regulating mitochondrial oxidative function in cultured myotubes. The capacity of Perm1 to regulate muscle mitochondrial content and function in vivo is not yet known. In this study, we use adeno-associated viral (AAV) vectors to increase Perm1 expression in skeletal muscles of 4-wk-old mice. Compared to control vector, AAV1-Perm1 leads to significant increases in mitochondrial content and oxidative capacity (by 40-80%). Moreover, AAV1-Perm1-transduced muscles show increased capillary density and resistance to fatigue (by 33 and 31%, respectively), without prominent changes in fiber-type composition. These findings suggest that Perm1 selectively regulates mitochondrial biogenesis and oxidative function, and implicate Perm1 in muscle adaptations that also occur in response to endurance exercise.
Collapse
Affiliation(s)
- Yoshitake Cho
- *Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, USA; Department of Orthopedic Surgery, School of Medicine, University of California, San Diego, La Jolla, California, USA; and Centre for Physical Activity and Nutrition Research, School of Exercise and Nutrition Sciences, Deakin University, Burwood, Australia
| | - Bethany C Hazen
- *Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, USA; Department of Orthopedic Surgery, School of Medicine, University of California, San Diego, La Jolla, California, USA; and Centre for Physical Activity and Nutrition Research, School of Exercise and Nutrition Sciences, Deakin University, Burwood, Australia
| | - Paulo G Gandra
- *Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, USA; Department of Orthopedic Surgery, School of Medicine, University of California, San Diego, La Jolla, California, USA; and Centre for Physical Activity and Nutrition Research, School of Exercise and Nutrition Sciences, Deakin University, Burwood, Australia
| | - Samuel R Ward
- *Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, USA; Department of Orthopedic Surgery, School of Medicine, University of California, San Diego, La Jolla, California, USA; and Centre for Physical Activity and Nutrition Research, School of Exercise and Nutrition Sciences, Deakin University, Burwood, Australia
| | - Simon Schenk
- *Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, USA; Department of Orthopedic Surgery, School of Medicine, University of California, San Diego, La Jolla, California, USA; and Centre for Physical Activity and Nutrition Research, School of Exercise and Nutrition Sciences, Deakin University, Burwood, Australia
| | - Aaron P Russell
- *Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, USA; Department of Orthopedic Surgery, School of Medicine, University of California, San Diego, La Jolla, California, USA; and Centre for Physical Activity and Nutrition Research, School of Exercise and Nutrition Sciences, Deakin University, Burwood, Australia
| | - Anastasia Kralli
- *Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California, USA; Department of Orthopedic Surgery, School of Medicine, University of California, San Diego, La Jolla, California, USA; and Centre for Physical Activity and Nutrition Research, School of Exercise and Nutrition Sciences, Deakin University, Burwood, Australia
| |
Collapse
|
35
|
Kim JH, Choi YK, Do JY, Choi YK, Ha CM, Lee SJ, Jeon JH, Lee WK, Choi HS, Park KG, Lee IK. Estrogen-Related Receptor γ Plays a Key Role in Vascular Calcification Through the Upregulation of BMP2 Expression. Arterioscler Thromb Vasc Biol 2015; 35:2384-90. [PMID: 26404484 DOI: 10.1161/atvbaha.115.306102] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 09/08/2015] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Vascular calcification which refers to ectopic mineralization in vascular cells is associated with several conditions, such as chronic kidney disease, atherosclerosis, and diabetes mellitus. Estrogen-related receptor (ERR)γ is a member of the orphan nuclear receptor superfamily, which plays diverse roles in regulating homeostatic and metabolic processes. However, the role of ERRγ in vascular calcification has not been investigated to date. The aim of the present study was to examine the role of ERRγ in vascular calcification. APPROACH AND RESULTS Vascular calcification was induced by treating rat aortic vascular smooth muscle cells with calcification medium. ERRγ expression in vascular smooth muscle cells was induced during calcification medium-induced vascular calcification. Adenovirus-mediated overexpression of ERRγ in vascular smooth muscle cells resulted in the upregulation of the expression of osteogenic genes, including runt-related transcription factor 2, osteopontin, and Msx2, and the downregulation of α-smooth muscle actin. Adenovirus-mediated overexpression of ERRγ induced bone morphogenetic protein 2 (BMP2) expression, leading to increased phosphorylation of the intracellular BMP2 effector proteins SMAD1/5/8. Collectively, these data suggested that ERRγ promotes dedifferentiation of vascular smooth muscle cells to an osteogenic phenotype during the vascular calcification process. Inhibition of endogenous ERRγ expression or activity using a specific siRNA or the selective inverse agonist GSK5182 attenuated vascular calcification and osteogenic gene expression in vitro and in vivo. CONCLUSIONS The present results indicate that ERRγ plays a key role in vascular calcification by upregulating the BMP2 signaling pathway, suggesting that inhibition of ERRγ is a potential therapeutic strategy for the prevention of vascular calcification.
Collapse
Affiliation(s)
- Ji-Hyun Kim
- From the Department of Internal Medicine (J.-H.K., Y.-K.C., Y.-K.C., S.J.L., J.-H.J., K.-G.P., I.-K.L.), Research Institute of Aging and Metabolism (J.-H.K., S.J.L., K.-G.P., I.-K.L.), and BK21 Plus KNU Biomedical Convergence Program (C.-M.H., I.-K.L.), Kyungpook National University School of Medicine, Daegu, South Korea; Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease (Y.-K.C., J.-Y.D., Y.-K.C., J.-H.J., K.-G.P., I.-K.L.), Department of Biomedical Science, Graduate School of Medicine (C.-M.H., K.-G.P., I.-K.L.), Department of Ophthalmology, School of Medicine (J.-Y.D.), and Biomedical Research Institute, School of Medicine (W.-K.L.), Kyungpook National University, Daegu, South Korea; and National Creative Research Initiatives Center for Nuclear Receptor Signals, Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, South Korea (H.-S.C.)
| | - Yeon-Kyung Choi
- From the Department of Internal Medicine (J.-H.K., Y.-K.C., Y.-K.C., S.J.L., J.-H.J., K.-G.P., I.-K.L.), Research Institute of Aging and Metabolism (J.-H.K., S.J.L., K.-G.P., I.-K.L.), and BK21 Plus KNU Biomedical Convergence Program (C.-M.H., I.-K.L.), Kyungpook National University School of Medicine, Daegu, South Korea; Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease (Y.-K.C., J.-Y.D., Y.-K.C., J.-H.J., K.-G.P., I.-K.L.), Department of Biomedical Science, Graduate School of Medicine (C.-M.H., K.-G.P., I.-K.L.), Department of Ophthalmology, School of Medicine (J.-Y.D.), and Biomedical Research Institute, School of Medicine (W.-K.L.), Kyungpook National University, Daegu, South Korea; and National Creative Research Initiatives Center for Nuclear Receptor Signals, Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, South Korea (H.-S.C.)
| | - Ji-Yeon Do
- From the Department of Internal Medicine (J.-H.K., Y.-K.C., Y.-K.C., S.J.L., J.-H.J., K.-G.P., I.-K.L.), Research Institute of Aging and Metabolism (J.-H.K., S.J.L., K.-G.P., I.-K.L.), and BK21 Plus KNU Biomedical Convergence Program (C.-M.H., I.-K.L.), Kyungpook National University School of Medicine, Daegu, South Korea; Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease (Y.-K.C., J.-Y.D., Y.-K.C., J.-H.J., K.-G.P., I.-K.L.), Department of Biomedical Science, Graduate School of Medicine (C.-M.H., K.-G.P., I.-K.L.), Department of Ophthalmology, School of Medicine (J.-Y.D.), and Biomedical Research Institute, School of Medicine (W.-K.L.), Kyungpook National University, Daegu, South Korea; and National Creative Research Initiatives Center for Nuclear Receptor Signals, Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, South Korea (H.-S.C.)
| | - Young-Keun Choi
- From the Department of Internal Medicine (J.-H.K., Y.-K.C., Y.-K.C., S.J.L., J.-H.J., K.-G.P., I.-K.L.), Research Institute of Aging and Metabolism (J.-H.K., S.J.L., K.-G.P., I.-K.L.), and BK21 Plus KNU Biomedical Convergence Program (C.-M.H., I.-K.L.), Kyungpook National University School of Medicine, Daegu, South Korea; Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease (Y.-K.C., J.-Y.D., Y.-K.C., J.-H.J., K.-G.P., I.-K.L.), Department of Biomedical Science, Graduate School of Medicine (C.-M.H., K.-G.P., I.-K.L.), Department of Ophthalmology, School of Medicine (J.-Y.D.), and Biomedical Research Institute, School of Medicine (W.-K.L.), Kyungpook National University, Daegu, South Korea; and National Creative Research Initiatives Center for Nuclear Receptor Signals, Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, South Korea (H.-S.C.)
| | - Chae-Myeong Ha
- From the Department of Internal Medicine (J.-H.K., Y.-K.C., Y.-K.C., S.J.L., J.-H.J., K.-G.P., I.-K.L.), Research Institute of Aging and Metabolism (J.-H.K., S.J.L., K.-G.P., I.-K.L.), and BK21 Plus KNU Biomedical Convergence Program (C.-M.H., I.-K.L.), Kyungpook National University School of Medicine, Daegu, South Korea; Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease (Y.-K.C., J.-Y.D., Y.-K.C., J.-H.J., K.-G.P., I.-K.L.), Department of Biomedical Science, Graduate School of Medicine (C.-M.H., K.-G.P., I.-K.L.), Department of Ophthalmology, School of Medicine (J.-Y.D.), and Biomedical Research Institute, School of Medicine (W.-K.L.), Kyungpook National University, Daegu, South Korea; and National Creative Research Initiatives Center for Nuclear Receptor Signals, Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, South Korea (H.-S.C.)
| | - Sun Joo Lee
- From the Department of Internal Medicine (J.-H.K., Y.-K.C., Y.-K.C., S.J.L., J.-H.J., K.-G.P., I.-K.L.), Research Institute of Aging and Metabolism (J.-H.K., S.J.L., K.-G.P., I.-K.L.), and BK21 Plus KNU Biomedical Convergence Program (C.-M.H., I.-K.L.), Kyungpook National University School of Medicine, Daegu, South Korea; Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease (Y.-K.C., J.-Y.D., Y.-K.C., J.-H.J., K.-G.P., I.-K.L.), Department of Biomedical Science, Graduate School of Medicine (C.-M.H., K.-G.P., I.-K.L.), Department of Ophthalmology, School of Medicine (J.-Y.D.), and Biomedical Research Institute, School of Medicine (W.-K.L.), Kyungpook National University, Daegu, South Korea; and National Creative Research Initiatives Center for Nuclear Receptor Signals, Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, South Korea (H.-S.C.)
| | - Jae-Han Jeon
- From the Department of Internal Medicine (J.-H.K., Y.-K.C., Y.-K.C., S.J.L., J.-H.J., K.-G.P., I.-K.L.), Research Institute of Aging and Metabolism (J.-H.K., S.J.L., K.-G.P., I.-K.L.), and BK21 Plus KNU Biomedical Convergence Program (C.-M.H., I.-K.L.), Kyungpook National University School of Medicine, Daegu, South Korea; Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease (Y.-K.C., J.-Y.D., Y.-K.C., J.-H.J., K.-G.P., I.-K.L.), Department of Biomedical Science, Graduate School of Medicine (C.-M.H., K.-G.P., I.-K.L.), Department of Ophthalmology, School of Medicine (J.-Y.D.), and Biomedical Research Institute, School of Medicine (W.-K.L.), Kyungpook National University, Daegu, South Korea; and National Creative Research Initiatives Center for Nuclear Receptor Signals, Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, South Korea (H.-S.C.)
| | - Won-Kee Lee
- From the Department of Internal Medicine (J.-H.K., Y.-K.C., Y.-K.C., S.J.L., J.-H.J., K.-G.P., I.-K.L.), Research Institute of Aging and Metabolism (J.-H.K., S.J.L., K.-G.P., I.-K.L.), and BK21 Plus KNU Biomedical Convergence Program (C.-M.H., I.-K.L.), Kyungpook National University School of Medicine, Daegu, South Korea; Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease (Y.-K.C., J.-Y.D., Y.-K.C., J.-H.J., K.-G.P., I.-K.L.), Department of Biomedical Science, Graduate School of Medicine (C.-M.H., K.-G.P., I.-K.L.), Department of Ophthalmology, School of Medicine (J.-Y.D.), and Biomedical Research Institute, School of Medicine (W.-K.L.), Kyungpook National University, Daegu, South Korea; and National Creative Research Initiatives Center for Nuclear Receptor Signals, Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, South Korea (H.-S.C.)
| | - Hueng-Sik Choi
- From the Department of Internal Medicine (J.-H.K., Y.-K.C., Y.-K.C., S.J.L., J.-H.J., K.-G.P., I.-K.L.), Research Institute of Aging and Metabolism (J.-H.K., S.J.L., K.-G.P., I.-K.L.), and BK21 Plus KNU Biomedical Convergence Program (C.-M.H., I.-K.L.), Kyungpook National University School of Medicine, Daegu, South Korea; Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease (Y.-K.C., J.-Y.D., Y.-K.C., J.-H.J., K.-G.P., I.-K.L.), Department of Biomedical Science, Graduate School of Medicine (C.-M.H., K.-G.P., I.-K.L.), Department of Ophthalmology, School of Medicine (J.-Y.D.), and Biomedical Research Institute, School of Medicine (W.-K.L.), Kyungpook National University, Daegu, South Korea; and National Creative Research Initiatives Center for Nuclear Receptor Signals, Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, South Korea (H.-S.C.)
| | - Keun-Gyu Park
- From the Department of Internal Medicine (J.-H.K., Y.-K.C., Y.-K.C., S.J.L., J.-H.J., K.-G.P., I.-K.L.), Research Institute of Aging and Metabolism (J.-H.K., S.J.L., K.-G.P., I.-K.L.), and BK21 Plus KNU Biomedical Convergence Program (C.-M.H., I.-K.L.), Kyungpook National University School of Medicine, Daegu, South Korea; Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease (Y.-K.C., J.-Y.D., Y.-K.C., J.-H.J., K.-G.P., I.-K.L.), Department of Biomedical Science, Graduate School of Medicine (C.-M.H., K.-G.P., I.-K.L.), Department of Ophthalmology, School of Medicine (J.-Y.D.), and Biomedical Research Institute, School of Medicine (W.-K.L.), Kyungpook National University, Daegu, South Korea; and National Creative Research Initiatives Center for Nuclear Receptor Signals, Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, South Korea (H.-S.C.).
| | - In-Kyu Lee
- From the Department of Internal Medicine (J.-H.K., Y.-K.C., Y.-K.C., S.J.L., J.-H.J., K.-G.P., I.-K.L.), Research Institute of Aging and Metabolism (J.-H.K., S.J.L., K.-G.P., I.-K.L.), and BK21 Plus KNU Biomedical Convergence Program (C.-M.H., I.-K.L.), Kyungpook National University School of Medicine, Daegu, South Korea; Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease (Y.-K.C., J.-Y.D., Y.-K.C., J.-H.J., K.-G.P., I.-K.L.), Department of Biomedical Science, Graduate School of Medicine (C.-M.H., K.-G.P., I.-K.L.), Department of Ophthalmology, School of Medicine (J.-Y.D.), and Biomedical Research Institute, School of Medicine (W.-K.L.), Kyungpook National University, Daegu, South Korea; and National Creative Research Initiatives Center for Nuclear Receptor Signals, Hormone Research Center, School of Biological Sciences and Technology, Chonnam National University, Gwangju, South Korea (H.-S.C.).
| |
Collapse
|
36
|
Constitutive activities of estrogen-related receptors: Transcriptional regulation of metabolism by the ERR pathways in health and disease. Biochim Biophys Acta Mol Basis Dis 2015; 1852:1912-27. [PMID: 26115970 DOI: 10.1016/j.bbadis.2015.06.016] [Citation(s) in RCA: 134] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 06/15/2015] [Accepted: 06/17/2015] [Indexed: 12/17/2022]
Abstract
The estrogen-related receptors (ERRs) comprise a small group of orphan nuclear receptor transcription factors. The ERRα and ERRγ isoforms play a central role in the regulation of metabolic genes and cellular energy metabolism. Although less is known about ERRβ, recent studies have revealed the importance of this isoform in the maintenance of embryonic stem cell pluripotency. Thus, ERRs are essential to many biological processes. The development of several ERR knockout and overexpression models and the application of advanced functional genomics have allowed rapid advancement of our understanding of the physiology regulated by ERR pathways. Moreover, it has enabled us to begin to delineate the distinct programs regulated by ERRα and ERRγ that have overlapping effects on metabolism and growth. The current review primarily focuses on the physiologic roles of ERR isoforms related to their metabolic regulation; therefore, the ERRα and ERRγ are discussed in the greatest detail. We emphasize findings from gain- and loss-of-function models developed to characterize ERR control of skeletal muscle, heart and musculoskeletal physiology. These models have revealed that coordinating metabolic capacity with energy demand is essential for seemingly disparate processes such as muscle differentiation and hypertrophy, innate immune function, thermogenesis, and bone remodeling. Furthermore, the models have revealed that ERRα- and ERRγ-deficiency in mice accelerates progression of pathologic processes and implicates ERRs as etiologic factors in disease. We highlight the human diseases in which ERRs and their downstream metabolic pathways are perturbed, including heart failure and diabetes. While no natural ligand has been identified for any of the ERR isoforms, the potential for using synthetic small molecules to modulate their activity has been demonstrated. Based on our current understanding of their transcriptional mechanisms and physiologic relevance, the ERRs have emerged as potential therapeutic targets for treatment of osteoporosis, muscle atrophy, insulin resistance and heart failure in humans.
Collapse
|
37
|
Retinal hypoxia induces vascular endothelial growth factor through induction of estrogen-related receptor γ. Biochem Biophys Res Commun 2015; 460:457-63. [DOI: 10.1016/j.bbrc.2015.03.055] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Accepted: 03/10/2015] [Indexed: 11/18/2022]
|
38
|
Yadav V, Matsakas A, Lorca S, Narkar V. PGC1β Activates an Antiangiogenic Program to Repress Neoangiogenesis in Muscle Ischemia. Cell Rep 2014; 8:783-97. [DOI: 10.1016/j.celrep.2014.06.040] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Revised: 05/07/2014] [Accepted: 06/20/2014] [Indexed: 01/06/2023] Open
|
39
|
Luo Y, Kumar P, Chen CC, Latham J, Wang L, Tudela C, Alexander JM, Shelton JM, McKown L, Mendelson CR. Estrogen-related receptor γ serves a role in blood pressure homeostasis during pregnancy. Mol Endocrinol 2014; 28:965-75. [PMID: 24725083 DOI: 10.1210/me.2014-1003] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Persistent hypoxia caused by shallow trophoblast invasion and poor placental perfusion may underlie the pathophysiology of preeclampsia, a leading cause of maternal and neonatal morbidity and mortality. Previously, we found that estrogen-related receptor γ (ERRγ) serves a critical and O2-dependent role in differentiation of human trophoblasts in culture and expression of tissue kallikrein and voltage-gated K(+) channels. In this study, we surprisingly observed that ERRγ expression was significantly increased in placentas from preeclamptic women compared with that in gestation-matched normotensive women. To further investigate a functional role for ERRγ during pregnancy, we analyzed ERRγ-deficient mice. Maternal systolic blood pressure was significantly reduced in pregnant ERRγ(+/-) females bred to ERRγ(+/-) males compared with that in wild-type (WT) mice and was markedly up-regulated by treatment of WT pregnant mice with the ERRγ agonist DY131. Placentas of ERRγ(+/-) mice manifested increased vascular endothelial growth factor A expression compared with that in WT mice. Notably, circulating levels of the antiangiogenic factor, soluble fms-like tyrosine kinase-1, were significantly reduced in ERRγ(+/-) pregnant mice as was serum aldosterone. These effects were associated with a decrease in maternal adrenal Cyp11b1 (steroid 11β-hydroxylase) and Cyp11b2 (aldosterone synthase) expression. In contrast, adrenal Cyp11b1 and Cyp11b2 mRNA were increased in pregnant WT mice treated with DY131. Moreover, chromatin immunoprecipitation and luciferase reporter assays identified Cyp11b2 as a transcriptional target of ERRγ. Collectively, these findings reveal a potential role of ERRγ in maternal blood pressure homeostasis during pregnancy and suggest that aberrant ERRγ expression may contribute to the pathogenesis of preeclampsia.
Collapse
Affiliation(s)
- Yanmin Luo
- Department of Biochemistry (Y.L., P.K., C.-C.C., J.L., L.W., C.R.M.), North Texas March of Dimes Birth Defects Center, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9038; Department of Obstetrics and Gynecology (Y.L.), The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China; and Department of Obstetrics and Gynecology (C.T., J.M.A., C.R.M.) and Department of Internal Medicine (J.M.S., L.M.), University of Texas Southwestern Medical Center, Dallas, Texas 75390-9038
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
|
41
|
Fan W, Atkins AR, Yu RT, Downes M, Evans RM. Road to exercise mimetics: targeting nuclear receptors in skeletal muscle. J Mol Endocrinol 2013; 51:T87-T100. [PMID: 24280961 PMCID: PMC3936671 DOI: 10.1530/jme-13-0258] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Skeletal muscle is the largest organ in the human body and is the major site for energy expenditure. It exhibits remarkable plasticity in response to physiological stimuli such as exercise. Physical exercise remodels skeletal muscle and enhances its capability to burn calories, which has been shown to be beneficial for many clinical conditions including the metabolic syndrome and cancer. Nuclear receptors (NRs) comprise a class of transcription factors found only in metazoans that regulate major biological processes such as reproduction, development, and metabolism. Recent studies have demonstrated crucial roles for NRs and their co-regulators in the regulation of skeletal muscle energy metabolism and exercise-induced muscle remodeling. While nothing can fully replace exercise, development of exercise mimetics that enhance or even substitute for the beneficial effects of physical exercise would be of great benefit. The unique property of NRs that allows modulation by endogenous or synthetic ligands makes them bona fide therapeutic targets. In this review, we present an overview of the current understanding of the role of NRs and their co-regulators in skeletal muscle oxidative metabolism and summarize recent progress in the development of exercise mimetics that target NRs and their co-regulators.
Collapse
Affiliation(s)
- Weiwei Fan
- Gene Expression Laboratory Howard Hughes Medical Institute, Salk Institute for Biological Studies, La Jolla, California 92037, USA
| | | | | | | | | |
Collapse
|
42
|
Sun Z, Huang P, Tong G, Lin J, Jin A, Rong P, Zhu L, Nie L, Niu G, Cao F, Chen X. VEGF-loaded graphene oxide as theranostics for multi-modality imaging-monitored targeting therapeutic angiogenesis of ischemic muscle. NANOSCALE 2013; 5:6857-66. [PMID: 23770832 PMCID: PMC4607062 DOI: 10.1039/c3nr01573d] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Herein we report the design and synthesis of multifunctional VEGF-loaded IR800-conjugated graphene oxide (GO-IR800-VEGF) for multi-modality imaging-monitored therapeutic angiogenesis of ischemic muscle. The as-prepared GO-IR800-VEGF positively targets VEGF receptors, maintains an elevated level of VEGF in ischemic tissues for a prolonged time, and finally leads to remarkable therapeutic angiogenesis of ischemic muscle. Although more efforts are required to further understand the in vivo behaviors and the long-term toxicology of GO, our work demonstrates the success of using GO for efficient VEGF delivery in vivo by intravenous administration and suggests the great promise of using graphene oxide in theranostic applications for treating ischemic disease.
Collapse
Affiliation(s)
- Zhongchan Sun
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China 710032
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD 20892-2281, USA
| | - Peng Huang
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD 20892-2281, USA
- Research Institute of Micro/Nano Science and Technology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Guang Tong
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Jing Lin
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD 20892-2281, USA
- Research Institute of Micro/Nano Science and Technology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Albert Jin
- Laboratory of Cellular Imaging and Macromolecular Biophysics, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD 20892-2281, USA
| | - Pengfei Rong
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD 20892-2281, USA
| | - Lei Zhu
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD 20892-2281, USA
- Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361005 China
| | - Liming Nie
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD 20892-2281, USA
| | - Gang Niu
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD 20892-2281, USA
| | - Feng Cao
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China 710032
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD 20892-2281, USA
| |
Collapse
|
43
|
Tiidus PM, Lowe DA, Brown M. Estrogen replacement and skeletal muscle: mechanisms and population health. J Appl Physiol (1985) 2013; 115:569-78. [PMID: 23869062 DOI: 10.1152/japplphysiol.00629.2013] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
There is a growing body of information supporting the beneficial effects of estrogen and estrogen-based hormone therapy (HT) on maintenance and enhancement of muscle mass, strength, and connective tissue. These effects are also evident in enhanced recovery from muscle atrophy or damage and have significant implications particularly for the muscular health of postmenopausal women. Evidence suggests that HT will also help maintain or increase muscle mass, improve postatrophy muscle recovery, and enhance muscle strength in aged females. This is important because this population, in particular, is at risk for a rapid onset of frailty. The potential benefits of estrogen and HT relative to skeletal muscle function and composition combined with other health-related enhancements associated with reduced risk of cardiovascular events, overall mortality, and metabolic dysfunction, as well as enhanced cognition and bone health cumulate in a strong argument for more widespread and prolonged consideration of HT if started proximal to menopausal onset in most women. Earlier reports of increased health risks with HT use in postmenopausal women has led to a decline in HT use. However, recent reevaluation regarding the health effects of HT indicates a general lack of risks and a number of significant health benefits of HT use when initiated at the onset of menopause. Although further research is still needed to fully delineate its mechanisms of action, the general use of HT by postmenopausal women, to enhance muscle mass and strength, as well as overall health, with initiation soon after the onset of menopause should be considered.
Collapse
Affiliation(s)
- Peter M Tiidus
- Department of Kinesology and Physical Education, Wilfrid Laurier University, Waterloo Ontario, Canada
| | | | | |
Collapse
|
44
|
Matsakas A, Yadav V, Lorca S, Narkar V. Muscle ERRγ mitigates Duchenne muscular dystrophy via metabolic and angiogenic reprogramming. FASEB J 2013; 27:4004-16. [PMID: 23781095 DOI: 10.1096/fj.13-228296] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Treatment of Duchenne muscular dystrophy (DMD) by replacing mutant dystrophin or restoring dystrophin-associated glycoprotein complex (DAG) has been clinically challenging. Instead, identifying and targeting muscle pathways deregulated in DMD will provide new therapeutic avenues. We report that the expression of nuclear receptor estrogen-related receptor-γ (ERRγ), and its metabolic and angiogenic targets are down-regulated (50-85%) in skeletal muscles of mdx mice (DMD model) vs. wild-type mice. Corelatively, oxidative myofibers, muscle vasculature, and exercise tolerance (33%) are decreased in mdx vs. wild-type mice. Overexpressing ERRγ selectively in the dystrophic muscles of the mdx mice restored metabolic and angiogenic gene expression compared with control mdx mice. Further, ERRγ enhanced muscle oxidative myofibers, vasculature, and blood flow (by 33-66%) and improved exercise tolerance (by 75%) in the dystrophic mice. Restoring muscle ERRγ pathway ameliorated muscle damage and also prevented DMD hallmarks of postexercise muscle damage, hypoxia, and fatigue in mdx mice. Notably, ERRγ did not restore sarcolemmal DAG complex, which is thus dispensable for antidystrophic effects of ERRγ. In summary, ERRγ-dependent metabolic and angiogenic gene program is defective in DMD, and we demonstrate that its restoration is a potential strategy for treating muscular dystrophy.
Collapse
Affiliation(s)
- Antonios Matsakas
- 1Center for Metabolic and Degenerative Diseases, Brown Foundation Institute of Molecular Medicine, University of Texas Medical School at Houston, IMM/SRB 430F, 1825 Pressler St., Houston, TX 77030, USA.
| | | | | | | |
Collapse
|
45
|
Circulation Research
Thematic Synopsis: Cardiovascular Development. Circ Res 2013. [DOI: 10.1161/circresaha.113.301305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
46
|
Affiliation(s)
- Alain-Pierre Gadeau
- From the Université de Bordeaux, Cardiovascular Adaptation to Ischemia, Pessac, France (A.-P.G.); INSERM, Cardiovascular Adaptation to Ischemia, Pessac, France (A.-P.G.); and INSERM, U1048-I2MC, Faculté de Médecine, Université de Toulouse et Centre Hospitalier Universitaire de Toulouse, Toulouse, France (J.-F.A.)
| | - Jean-François Arnal
- From the Université de Bordeaux, Cardiovascular Adaptation to Ischemia, Pessac, France (A.-P.G.); INSERM, Cardiovascular Adaptation to Ischemia, Pessac, France (A.-P.G.); and INSERM, U1048-I2MC, Faculté de Médecine, Université de Toulouse et Centre Hospitalier Universitaire de Toulouse, Toulouse, France (J.-F.A.)
| |
Collapse
|