1
|
Su Q, Gao Z, Zhang F, Wu Z, Ji Q, Zhu K, Gui L. Effect of miR-10a on the proliferation and differentiation of yak adipocyte precursors. J Appl Genet 2024:10.1007/s13353-024-00932-6. [PMID: 39715988 DOI: 10.1007/s13353-024-00932-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/14/2024] [Accepted: 12/13/2024] [Indexed: 12/25/2024]
Abstract
The fat content of yak meat is significantly correlated with the meat quality, and an appropriate fat content helps to improve the texture of the meat. The involvement of miR-10a in regulating the differentiation and proliferation of various cell types has been reported. Therefore, in this study, the effects of miR-10a on lipid droplet accumulation were investigated by transfection of yak adipocyte precursors with an miR-10a inhibitor, followed by Oil Red O, BODIPY, EdU staining, and cell cycle analysis of the transfected and control cells. The relative expression of lipogenic marker genes was determined by RT-qPCR to clarify the effect of miR-10a on the differentiation and proliferation of yak adipocyte precursors. Mature adipocytes were collected for transcriptome analysis to identify differentially expressed target genes and the association of these genes with adipogenic pathways was investigated by GO and KEGG enrichment analyses. In addition, the phylogeny and expression profiles of miR-10a were analyzed in various yak tissues. The results showed that miR-10a could inhibit the differentiation and promote the proliferation of yak adipocyte precursors. Analysis of the RNA-Seq results showed that miR-10a inhibitor and inhibitor NC had six differentially expressed genes: FABP4, AKR1B7, IGF2, ROCK1, IFNB1, and PLA2G3. These genes were found to be involved in the regulation of adipogenesis, with IGF2 and IFNB1 being upregulated in the PI3K-Akt signaling pathway, which is activated upon stimulation by IGF2 and IFNB1 and inhibits the differentiation and promotes the proliferation of yak adipocytes precursor, which in turn affected adipogenesis. Moreover, phylogenetic analysis indicated that miR-10a evolved relatively recently in yak and sheep, while tissue expression profiles showed that miR-10a was highly expressed in yak lung tissues.
Collapse
Affiliation(s)
- Quyangangmao Su
- College of Agriculture and Animal Husbandry, Qinghai University, Qinghai Province, Xining, 810016, People's Republic of China
| | - Zhanhong Gao
- College of Agriculture and Animal Husbandry, Qinghai University, Qinghai Province, Xining, 810016, People's Republic of China
| | - Fengshuo Zhang
- College of Agriculture and Animal Husbandry, Qinghai University, Qinghai Province, Xining, 810016, People's Republic of China
| | - Zhenling Wu
- College of Agriculture and Animal Husbandry, Qinghai University, Qinghai Province, Xining, 810016, People's Republic of China
| | - Qiurong Ji
- College of Agriculture and Animal Husbandry, Qinghai University, Qinghai Province, Xining, 810016, People's Republic of China
| | - Kaina Zhu
- College of Agriculture and Animal Husbandry, Qinghai University, Qinghai Province, Xining, 810016, People's Republic of China
| | - Linsheng Gui
- College of Agriculture and Animal Husbandry, Qinghai University, Qinghai Province, Xining, 810016, People's Republic of China.
| |
Collapse
|
2
|
Si M, Yu R, Lin H, Li F, Jung S, Thomas SS, Danesh FS, Wang Y, Peng H, Hu Z. ROCK1 activates mitochondrial fission leading to oxidative stress and muscle atrophy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.22.563469. [PMID: 37905139 PMCID: PMC10614981 DOI: 10.1101/2023.10.22.563469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Chronic kidney disease (CKD) is often associated with protein-energy wasting (PEW), which is characterized by a reduction in muscle mass and strength. Although mitochondrial dysfunction and oxidative stress have been implicated to play a role in the pathogenesis of muscle wasting, the underlying mechanisms remain unclear. In this study, we used transcriptomics, metabolomics analyses and mouse gene manipulating approaches to investigate the effects of mitochondrial plasticity and oxidative stress on muscle wasting in mouse CKD models. Our results showed that the expression of oxidative stress response genes was increased, and that of oxidative phosphorylation genes was decreased in the muscles of mice with CKD. This was accompanied by reduced oxygen consumption rates, decreased levels of mitochondrial electron transport chain proteins, and increased cellular oxidative damage. Excessive mitochondrial fission was also observed, and we found that the activation of ROCK1 was responsible for this process. Inducible expression of muscle-specific constitutively active ROCK1(mROCK1ca)exacerbated mitochondrial fragmentation and muscle wasting in CKD mice. Conversely, ROCK1 depletion (ROCK1-/-) alleviated these phenomena. Mechanistically, ROCK1 activation promoted the recruitment of Drp1 to mitochondria, thereby facilitating fragmentation. Notably, the pharmacological inhibition of ROCK1 mitigated muscle wasting by suppressing mitochondrial fission and oxidative stress. Our findings demonstrate that ROCK1 participates in CKD-induced muscle wasting by promoting mitochondrial fission and oxidative stress, and pharmacological suppression of ROCK1 could be a therapeutic strategy for combating muscle wasting in CKD conditions.
Collapse
Affiliation(s)
- Meijun Si
- Nephrology Division, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Department of Nephrology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences; Guangzhou, China
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Rizhen Yu
- Nephrology Division, Zhejiang Provincial People’s Hospital, People’s Hospital of Hangzhou Medical College, Hangzhou, Hangzhou, Zhejiang, China
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Hongchun Lin
- Nephrology Division, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Feng Li
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Sungyun Jung
- Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - Sandhya S. Thomas
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Farhard S Danesh
- Nephrology Division, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yanlin Wang
- Division of Nephrology, Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Hui Peng
- Nephrology Division, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Zhaoyong Hu
- Nephrology Division, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
3
|
Zheng Y, He J, Yang D, Dai F, Yuan M, Liu S, Jia Y, Cheng Y. Irisin reduces the abnormal reproductive and metabolic phenotypes of PCOS by regulating the activity of brown adipose tissue in mice. Biol Reprod 2022; 107:1046-1058. [PMID: 35713297 PMCID: PMC9562123 DOI: 10.1093/biolre/ioac125] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 01/05/2022] [Accepted: 06/08/2022] [Indexed: 11/14/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrine and metabolic disease in women, with clinical manifestations of anovulation and hyperandrogenaemia. The treatment of PCOS mainly focuses on improving clinical symptoms, such as insulin sensitivity or menstrual disorder, through drug treatment. However, due to the pathogenesis diversity of PCOS, there is still a lack of effective treatment in clinics. Metabolic disorder is the key factor in the occurrence of PCOS. Brown adipose tissue (BAT) is a special adipose tissue in the human body that can participate in metabolic balance by improving heat production. BAT has been demonstrated to be an important substance involved in the metabolic disorder of PCOS. Although increasing evidence indicates that BAT transplantation can improve the symptoms of PCOS, it is difficult to achieve BAT transplantation at present due to technical limitations. Stimulation of BAT activation by exogenous substances may be an effective alternative therapy for PCOS. In this study, we investigated the effects of Irisin on dehydroepiandrosterone (DHEA)-induced PCOS in mice and evaluated the effect of Irisin on serum hormone levels and changes in body temperature, body weight and ovarian morphology. In our study, we found that Irisin can enhance the thermogenesis and insulin sensitivity of PCOS mice by activating the function of BAT. In addition, Irisin treatment can correct the menstrual cycle of PCOS mice, improve the serum steroid hormone disorder status, and reduce the formation of ovarian cystic follicles. In conclusion, our results showed that Irisin treatment significantly improved the metabolic disorder of PCOS and may provide a new and alternative therapy for the treatment of this pathology.
Collapse
Affiliation(s)
- Yajing Zheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Juan He
- Department of Obstetrics and Gynecology Ultrasound, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Dongyong Yang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Fangfang Dai
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Mengqin Yuan
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Shiyi Liu
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Yifan Jia
- Department of Pain, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Yanxiang Cheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| |
Collapse
|
4
|
Ho WT, Chang JS, Chen TC, Wang JK, Chang SW, Yang MH, Jou TS, Wang IJ. Inhibition of Rho-associated protein kinase activity enhances oxidative phosphorylation to support corneal endothelial cell migration. FASEB J 2022; 36:e22397. [PMID: 35661268 DOI: 10.1096/fj.202101442rr] [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: 09/14/2021] [Revised: 05/21/2022] [Accepted: 05/24/2022] [Indexed: 11/11/2022]
Abstract
Corneal endothelial cell (CEC) dysfunction causes corneal edema and severe visual impairment that require transplantation to restore vision. To address the unmet need of organ shortage, descemetorhexis without endothelial keratoplasty has been specifically employed to treat early stage Fuchs endothelial corneal dystrophy, which is pathophysiologically related to oxidative stress and exhibits centrally located corneal guttae. After stripping off central Descemet's membrane, rho-associated protein kinase (ROCK) inhibitor has been found to facilitate CEC migration, an energy-demanding task, thereby achieving wound closure. However, the correlation between ROCK inhibition and the change in bioenergetic status of CECs remained to be elucidated. Through transcriptomic profiling, we found that the inhibition of ROCK activity by the selective inhibitor, ripasudil or Y27632, promoted enrichment of oxidative phosphorylation (OXPHOS) gene set in bovine CECs (BCECs). Functional analysis revealed that ripasudil, a clinically approved anti-glaucoma agent, enhanced mitochondrial respiration, increased spare respiratory capacity, and induced overexpression of electron transport chain components through upregulation of AMP-activated protein kinase (AMPK) pathway. Accelerated BCEC migration and in vitro wound healing by ripasudil were diminished by OXPHOS and AMPK inhibition, but not by glycolysis inhibition. Correspondingly, lamellipodial protrusion and actin assembly that were augmented by ripasudil became reduced with additional OXPHOS or AMPK inhibition. These results indicate that ROCK inhibition induces metabolic reprogramming toward OXPHOS to support migration of CECs.
Collapse
Affiliation(s)
- Wei-Ting Ho
- Department of Ophthalmology, Far Eastern Memorial Hospital, New Taipei City, Taiwan.,School of Medicine, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Jung-Shen Chang
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan
| | - Tsan-Chi Chen
- Department of Ophthalmology, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Jia-Kang Wang
- Department of Ophthalmology, Far Eastern Memorial Hospital, New Taipei City, Taiwan.,Department of Electrical Engineering, Yuan Ze University, Taoyuan, Taiwan
| | - Shu-Wen Chang
- Department of Ophthalmology, Far Eastern Memorial Hospital, New Taipei City, Taiwan.,College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Muh-Hwa Yang
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Hsinchu, Taiwan.,Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Tzuu-Shuh Jou
- College of Medicine, National Taiwan University, Taipei, Taiwan.,Center of Precision Medicine, College of Medicine, National Taiwan University, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - I-Jong Wang
- Department of Ophthalmology, National Taiwan University Hospital, Taipei, Taiwan.,College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
5
|
Wei L, Shi J. Insight Into Rho Kinase Isoforms in Obesity and Energy Homeostasis. Front Endocrinol (Lausanne) 2022; 13:886534. [PMID: 35769086 PMCID: PMC9234286 DOI: 10.3389/fendo.2022.886534] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/06/2022] [Indexed: 11/13/2022] Open
Abstract
Obesity and associated complications increasingly jeopardize global health and contribute to the rapidly rising prevalence of type 2 diabetes mellitus and obesity-related diseases. Developing novel methods for the prevention and treatment of excess body adipose tissue expansion can make a significant contribution to public health. Rho kinase is a Rho-associated coiled-coil-containing protein kinase (Rho kinase or ROCK). The ROCK family including ROCK1 and ROCK2 has recently emerged as a potential therapeutic target for the treatment of metabolic disorders. Up-regulated ROCK activity has been involved in the pathogenesis of all aspects of metabolic syndrome including obesity, insulin resistance, dyslipidemia and hypertension. The RhoA/ROCK-mediated actin cytoskeleton dynamics have been implicated in both white and beige adipogenesis. Studies using ROCK pan-inhibitors in animal models of obesity, diabetes, and associated complications have demonstrated beneficial outcomes. Studies via genetically modified animal models further established isoform-specific roles of ROCK in the pathogenesis of metabolic disorders including obesity. However, most reported studies have been focused on ROCK1 activity during the past decade. Due to the progress in developing ROCK2-selective inhibitors in recent years, a growing body of evidence indicates more attention should be devoted towards understanding ROCK2 isoform function in metabolism. Hence, studying individual ROCK isoforms to reveal their specific roles and principal mechanisms in white and beige adipogenesis, insulin sensitivity, energy balancing regulation, and obesity development will facilitate significant breakthroughs for systemic treatment with isoform-selective inhibitors. In this review, we give an overview of ROCK functions in the pathogenesis of obesity and insulin resistance with a particular focus on the current understanding of ROCK isoform signaling in white and beige adipogenesis, obesity and thermogenesis in adipose tissue and other major metabolic organs involved in energy homeostasis regulation.
Collapse
Affiliation(s)
- Lei Wei
- *Correspondence: Lei Wei, ; Jianjian Shi,
| | | |
Collapse
|
6
|
Rodríguez-Fdez S, Bustelo XR. Rho GTPases in Skeletal Muscle Development and Homeostasis. Cells 2021; 10:cells10112984. [PMID: 34831205 PMCID: PMC8616218 DOI: 10.3390/cells10112984] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 10/29/2021] [Accepted: 10/29/2021] [Indexed: 02/07/2023] Open
Abstract
Rho guanosine triphosphate hydrolases (GTPases) are molecular switches that cycle between an inactive guanosine diphosphate (GDP)-bound and an active guanosine triphosphate (GTP)-bound state during signal transduction. As such, they regulate a wide range of both cellular and physiological processes. In this review, we will summarize recent work on the role of Rho GTPase-regulated pathways in skeletal muscle development, regeneration, tissue mass homeostatic balance, and metabolism. In addition, we will present current evidence that links the dysregulation of these GTPases with diseases caused by skeletal muscle dysfunction. Overall, this information underscores the critical role of a number of members of the Rho GTPase subfamily in muscle development and the overall metabolic balance of mammalian species.
Collapse
Affiliation(s)
- Sonia Rodríguez-Fdez
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain;
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, 37007 Salamanca, Spain
- Wellcome-MRC Institute of Metabolic Science and MRC Metabolic Diseases Unit, University of Cambridge, Cambridge CB2 0QQ, UK
- Correspondence: or
| | - Xosé R. Bustelo
- Molecular Mechanisms of Cancer Program, Centro de Investigación del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain;
- Instituto de Biología Molecular y Celular del Cáncer, CSIC-University of Salamanca, 37007 Salamanca, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), CSIC-University of Salamanca, 37007 Salamanca, Spain
| |
Collapse
|
7
|
Chen X, Tan QQ, Tan XR, Li SJ, Zhang XX. Circ_0057558 promotes nonalcoholic fatty liver disease by regulating ROCK1/AMPK signaling through targeting miR-206. Cell Death Dis 2021; 12:809. [PMID: 34446693 PMCID: PMC8390503 DOI: 10.1038/s41419-021-04090-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 02/07/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is one of the most prevalent chronic liver disorders that is featured by the extensive deposition of fat in the hepatocytes. Current treatments are very limited due to its unclear pathogenesis. Here, we investigated the function of circ_0057558 and miR-206 in NAFLD. High-fat diet (HFD) feeding mouse was used as an in vivo NAFLD model and long-chain-free fatty acid (FFA)-treated liver cells were used as an in vitro NAFLD model. qRT-PCR was used to measure levels of miR-206, ROCK1 mRNA, and circ_0057558, while Western blotting was employed to determine protein levels of ROCK1, p-AMPK, AMPK, and lipogenesis-related proteins. Immunohistochemistry were performed to examine ROCK1 level. Oil-Red O staining was used to assess the lipid deposition in cells. ELISA was performed to examine secreted triglyceride (TG) level. Dual-luciferase assay was used to validate interactions of miR-206/ROCK1 and circ_0057558/miR-206. RNA immunoprecipitation was employed to confirm the binding of circ_0057558 with miR-206. Circ_0057558 was elevated while miR-206 was reduced in both in vivo and in vitro NAFLD models. miR-206 directly bound with ROCK1 3'-UTR and suppressed lipogenesis and TG secretion through targeting ROCK1/AMPK signaling. Circ_0057558 directly interacted with miR-206 to disinhibit ROCK1/AMPK signaling. Knockdown of circ_0057558 or overexpression of miR-206 inhibited lipogenesis, TG secretion and expression of lipogenesis-related proteins. ROCK1 knockdown reversed the effects of circ_0057558 overexpression. Injection of miR-206 mimics significantly ameliorated NAFLD progression in vivo. Circ_0057558 acts as a miR-206 sponge to de-repress the ROCK1/AMPK signaling and facilitates lipogenesis and TG secretion, which greatly contributes to NAFLD development and progression.
Collapse
Affiliation(s)
- Xi Chen
- Department of Pediatrics, The Second Xiangya Hospital, Central South Univeristy, Changsha, 410011, Hunan Province, China
| | - Qing-Qing Tan
- Department of Biology, The Dietrich School of Arts and Sciences, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Xin-Rui Tan
- Department of Pediatrics, The Second Xiangya Hospital, Central South Univeristy, Changsha, 410011, Hunan Province, China
| | - Shi-Jun Li
- Department of Pediatrics, The Second Xiangya Hospital, Central South Univeristy, Changsha, 410011, Hunan Province, China
| | - Xing-Xing Zhang
- Department of Pediatrics, The Second Xiangya Hospital, Central South Univeristy, Changsha, 410011, Hunan Province, China.
| |
Collapse
|
8
|
Maak S, Norheim F, Drevon CA, Erickson HP. Progress and Challenges in the Biology of FNDC5 and Irisin. Endocr Rev 2021; 42:436-456. [PMID: 33493316 PMCID: PMC8284618 DOI: 10.1210/endrev/bnab003] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Indexed: 01/10/2023]
Abstract
In 2002, a transmembrane protein-now known as FNDC5-was discovered and shown to be expressed in skeletal muscle, heart, and brain. It was virtually ignored for 10 years, until a study in 2012 proposed that, in response to exercise, the ectodomain of skeletal muscle FNDC5 was cleaved, traveled to white adipose tissue, and induced browning. The wasted energy of this browning raised the possibility that this myokine, named irisin, might mediate some beneficial effects of exercise. Since then, more than 1000 papers have been published exploring the roles of irisin. A major interest has been on adipose tissue and metabolism, following up the major proposal from 2012. Many studies correlating plasma irisin levels with physiological conditions have been questioned for using flawed assays for irisin concentration. However, experiments altering irisin levels by injecting recombinant irisin or by gene knockout are more promising. Recent discoveries have suggested potential roles of irisin in bone remodeling and in the brain, with effects potentially related to Alzheimer's disease. We discuss some discrepancies between research groups and the mechanisms that are yet to be determined. Some important questions raised in the initial discovery of irisin, such as the role of the mutant start codon of human FNDC5 and the mechanism of ectodomain cleavage, remain to be answered. Apart from these specific questions, a promising new tool has been developed-mice with a global or tissue-specific knockout of FNDC5. In this review, we critically examine the current knowledge and delineate potential solutions to resolve existing ambiguities.
Collapse
Affiliation(s)
- Steffen Maak
- Institute of Muscle Biology and Growth, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Frode Norheim
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Christian A Drevon
- Department of Nutrition, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | | |
Collapse
|
9
|
Ganel L, Chen L, Christ R, Vangipurapu J, Young E, Das I, Kanchi K, Larson D, Regier A, Abel H, Kang CJ, Scott A, Havulinna A, Chiang CWK, Service S, Freimer N, Palotie A, Ripatti S, Kuusisto J, Boehnke M, Laakso M, Locke A, Stitziel NO, Hall IM. Mitochondrial genome copy number measured by DNA sequencing in human blood is strongly associated with metabolic traits via cell-type composition differences. Hum Genomics 2021; 15:34. [PMID: 34099068 PMCID: PMC8185936 DOI: 10.1186/s40246-021-00335-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 05/26/2021] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Mitochondrial genome copy number (MT-CN) varies among humans and across tissues and is highly heritable, but its causes and consequences are not well understood. When measured by bulk DNA sequencing in blood, MT-CN may reflect a combination of the number of mitochondria per cell and cell-type composition. Here, we studied MT-CN variation in blood-derived DNA from 19184 Finnish individuals using a combination of genome (N = 4163) and exome sequencing (N = 19034) data as well as imputed genotypes (N = 17718). RESULTS We identified two loci significantly associated with MT-CN variation: a common variant at the MYB-HBS1L locus (P = 1.6 × 10-8), which has previously been associated with numerous hematological parameters; and a burden of rare variants in the TMBIM1 gene (P = 3.0 × 10-8), which has been reported to protect against non-alcoholic fatty liver disease. We also found that MT-CN is strongly associated with insulin levels (P = 2.0 × 10-21) and other metabolic syndrome (metS)-related traits. Using a Mendelian randomization framework, we show evidence that MT-CN measured in blood is causally related to insulin levels. We then applied an MT-CN polygenic risk score (PRS) derived from Finnish data to the UK Biobank, where the association between the PRS and metS traits was replicated. Adjusting for cell counts largely eliminated these signals, suggesting that MT-CN affects metS via cell-type composition. CONCLUSION These results suggest that measurements of MT-CN in blood-derived DNA partially reflect differences in cell-type composition and that these differences are causally linked to insulin and related traits.
Collapse
Affiliation(s)
- Liron Ganel
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Lei Chen
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Ryan Christ
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - Jagadish Vangipurapu
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
| | - Erica Young
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
- Department of Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, USA
| | - Indraniel Das
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - Krishna Kanchi
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - David Larson
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Allison Regier
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Haley Abel
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Chul Joo Kang
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
| | - Alexandra Scott
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Aki Havulinna
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- Finnish Institute for Health and Welfare (THL), Helsinki, Finland
| | - Charleston W K Chiang
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Quantitative and Computational Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, USA
| | - Susan Service
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, USA
| | - Nelson Freimer
- Center for Neurobehavioral Genetics, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California Los Angeles, Los Angeles, CA, USA
| | - Aarno Palotie
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- Analytical and Translational Genetics Unit (ATGU), Psychiatric & Neurodevelopmental Genetics Unit, Departments of Psychiatry and Neurology, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Johanna Kuusisto
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Michael Boehnke
- Department of Biostatistics and Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Markku Laakso
- Institute of Clinical Medicine, Internal Medicine, University of Eastern Finland, Kuopio, Finland
- Department of Medicine, Kuopio University Hospital, Kuopio, Finland
| | - Adam Locke
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Nathan O Stitziel
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA.
| | - Ira M Hall
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
10
|
Pang BPS, Chan WS, Chan CB. Mitochondria Homeostasis and Oxidant/Antioxidant Balance in Skeletal Muscle-Do Myokines Play a Role? Antioxidants (Basel) 2021; 10:antiox10020179. [PMID: 33513795 PMCID: PMC7911667 DOI: 10.3390/antiox10020179] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/19/2021] [Accepted: 01/21/2021] [Indexed: 12/19/2022] Open
Abstract
Mitochondria are the cellular powerhouses that generate adenosine triphosphate (ATP) to substantiate various biochemical activities. Instead of being a static intracellular structure, they are dynamic organelles that perform constant structural and functional remodeling in response to different metabolic stresses. In situations that require a high ATP supply, new mitochondria are assembled (mitochondrial biogenesis) or formed by fusing the existing mitochondria (mitochondrial fusion) to maximize the oxidative capacity. On the other hand, nutrient overload may produce detrimental metabolites such as reactive oxidative species (ROS) that wreck the organelle, leading to the split of damaged mitochondria (mitofission) for clearance (mitophagy). These vital processes are tightly regulated by a sophisticated quality control system involving energy sensing, intracellular membrane interaction, autophagy, and proteasomal degradation to optimize the number of healthy mitochondria. The effective mitochondrial surveillance is particularly important to skeletal muscle fitness because of its large tissue mass as well as its high metabolic activities for supporting the intensive myofiber contractility. Indeed, the failure of the mitochondrial quality control system in skeletal muscle is associated with diseases such as insulin resistance, aging, and muscle wasting. While the mitochondrial dynamics in cells are believed to be intrinsically controlled by the energy content and nutrient availability, other upstream regulators such as hormonal signals from distal organs or factors generated by the muscle itself may also play a critical role. It is now clear that skeletal muscle actively participates in systemic energy homeostasis via producing hundreds of myokines. Acting either as autocrine/paracrine or circulating hormones to crosstalk with other organs, these secretory myokines regulate a large number of physiological activities including insulin sensitivity, fuel utilization, cell differentiation, and appetite behavior. In this article, we will review the mechanism of myokines in mitochondrial quality control and ROS balance, and discuss their translational potential.
Collapse
|
11
|
Muñoz VR, Gaspar RC, Kuga GK, Pavan ICB, Simabuco FM, da Silva ASR, de Moura LP, Cintra DE, Ropelle ER, Pauli JR. The Effects of Aging on Rho-Kinase and Insulin Signaling in Skeletal Muscle and White Adipose Tissue of Rats. J Gerontol A Biol Sci Med Sci 2020; 75:432-436. [PMID: 30596894 DOI: 10.1093/gerona/gly293] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Indexed: 12/21/2022] Open
Abstract
The insulin receptor substrate 1 regulates insulin-mediated glucose uptake and is a target of Rho-kinase (Rock); however, the relationship between age-related insulin resistance and Rock signaling specifically in skeletal muscle and adipose tissue is unknown. We evaluated the content and activity of Rock in C2C12 myotubes, and in skeletal muscle and white adipose tissue (WAT) from two rodent models that differ in their patterns of body fat accumulation during aging (Wistar and Fischer 344 rats). Body fat gain in the Wistar rats was greater than in Fischer rats and only Wistar rats had impairment of whole-body insulin sensitivity. Rock activity and insulin signaling were impaired in skeletal muscle in both rat models, but only middle-aged Wistar rats had higher Rock activity in WAT. These data are consistent with a positive role of Rock in regulating insulin signaling in both skeletal muscle and its negative role in the adipose tissue, suggesting that Rock activity in adipose tissue is important in age-related insulin resistance.
Collapse
Affiliation(s)
- Vitor Rosetto Muñoz
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Rafael Calais Gaspar
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Gabriel Keine Kuga
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Isadora Carolina Betim Pavan
- Laboratory of Functional Properties in Foods, School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Fernando Moreira Simabuco
- Laboratory of Functional Properties in Foods, School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Adelino Sanchez Ramos da Silva
- Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, USP, Ribeirão Preto, São Paulo, Brazil.,School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Leandro Pereira de Moura
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil.,OCRC - Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.,Postgraduate Program in Motor Science - São Paulo State University (UNESP).,CEPECE - Center of Research in Sport Sciences. School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Dennys Esper Cintra
- OCRC - Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.,Laboratory of Nutritional Genomics, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Eduardo Rochete Ropelle
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil.,OCRC - Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.,CEPECE - Center of Research in Sport Sciences. School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - José Rodrigo Pauli
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil.,OCRC - Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.,CEPECE - Center of Research in Sport Sciences. School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| |
Collapse
|
12
|
Muñoz VR, Gaspar RC, Esteca MV, Baptista IL, Vieira RFL, da Silva ASR, de Moura LP, Cintra DE, Ropelle ER, Pauli JR. Physical exercise increases ROCK activity in the skeletal muscle of middle-aged rats. Mech Ageing Dev 2020; 186:111213. [PMID: 32032622 DOI: 10.1016/j.mad.2020.111213] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 01/17/2020] [Accepted: 01/30/2020] [Indexed: 12/23/2022]
Abstract
The physical exercise is a potential strategy to control age-related metabolic disorders, such as insulin resistance, impaired glucose homeostasis, and type 2 diabetes. Rho-kinase (ROCK) increases skeletal muscle glucose uptake through Insulin Receptor Substrate 1 (IRS1) phosphorylation. Here, we investigated the role of physical exercise in ROCK pathway in the skeletal muscle of Fischer middle-aged rats. Firstly, we observed the ROCK distribution in different skeletal muscle fiber types. ROCK signaling pathway (ROCK1 and ROCK2) and activity (pMYPT1) were higher in the soleus, which was associated with increased insulin signaling pathway (pIR, pIRS1, pPDK, pGSK3β). Middle-aged rats submitted to physical exercise, showed the upregulation of ROCK2 content and normalized RhoA (ROCK activator enzyme) levels in soleus muscle compared with middle-aged sedentary rats. These molecular changes in middle-aged exercised rats were accompanied by higher insulin signaling (pIRS1, pGSK3β, pAS160, GLUT4) in the soleus muscle. Reinforcing these findings, when pharmacological inhibition of ROCK activity was performed (using Y-27632), the insulin signaling pathway and glucose metabolism-related genes (Tpi, Pgk1, Pgam2, Eno3) were decreased in the soleus muscle of exercised rats. In summary, ROCK signaling seems to contribute with whole-body glucose homeostasis (∼50 %) through its higher upregulation in the soleus muscle in middle-aged exercised rats.
Collapse
Affiliation(s)
- Vitor Rosetto Muñoz
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Rafael Calais Gaspar
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Marcos Vinicius Esteca
- Laboratory of Cellular and Tissue Biology, Faculty of Applied Sciences, University of Campinas (UNICAMP), Limeira, Brazil
| | - Igor Luchini Baptista
- Laboratory of Cellular and Tissue Biology, Faculty of Applied Sciences, University of Campinas (UNICAMP), Limeira, Brazil
| | - Renan Fudoli Lins Vieira
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Adelino Sanchez Ramos da Silva
- Postgraduate Program in Rehabilitation and Functional Performance, Ribeirão Preto Medical School, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil; School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil
| | - Leandro Pereira de Moura
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil; OCRC - Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Dennys Esper Cintra
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto, São Paulo, Brazil; Laboratory of Nutritional Genomics, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Eduardo Rochete Ropelle
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil; OCRC - Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - José Rodrigo Pauli
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil; OCRC - Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil.
| |
Collapse
|
13
|
Hong SH, Choi KM. Sarcopenic Obesity, Insulin Resistance, and Their Implications in Cardiovascular and Metabolic Consequences. Int J Mol Sci 2020; 21:ijms21020494. [PMID: 31941015 PMCID: PMC7013734 DOI: 10.3390/ijms21020494] [Citation(s) in RCA: 182] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 01/06/2020] [Accepted: 01/10/2020] [Indexed: 12/14/2022] Open
Abstract
The prevalence of sarcopenic obesity is increasing worldwide, particularly amongst aging populations. Insulin resistance is the core mechanism of sarcopenic obesity and is also associated with variable cardiometabolic diseases such as cardiovascular disease, type 2 diabetes mellitus, and non-alcoholic fatty liver disease. Fat accumulation in muscle tissue promotes a proinflammatory cascade and oxidative stress, leading to mitochondrial dysfunction, impaired insulin signaling, and muscle atrophy. To compound the problem, decreased muscle mass aggravates insulin resistance. In addition, the crosstalk between myokines and adipokines leads to negative feedback, which in turn aggravates sarcopenic obesity and insulin resistance. In this review, we focus on the molecular mechanisms linking sarcopenic obesity and insulin resistance with various biological pathways. We also discuss the impact and mechanism of sarcopenic obesity and insulin resistance on cardiometabolic disease.
Collapse
|
14
|
Landry T, Shookster D, Huang H. Tissue-Specific Approaches Reveal Diverse Metabolic Functions of Rho-Kinase 1. Front Endocrinol (Lausanne) 2020; 11:622581. [PMID: 33633690 PMCID: PMC7901932 DOI: 10.3389/fendo.2020.622581] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 12/22/2020] [Indexed: 01/20/2023] Open
Abstract
Rho-kinase 1 (ROCK1) has been implicated in diverse metabolic functions throughout the body, with promising evidence identifying ROCK1 as a therapeutic target in diabetes and obesity. Considering these metabolic roles, several pharmacological inhibitors have been developed to elucidate the mechanisms underlying ROCK1 function. Y27632 and fasudil are two common ROCK1 inhibitors; however, they have varying non-specific selectivity to inhibit other AGC kinase subfamily members and whole-body pharmacological approaches lack tissue-specific insight. As a result, interpretation of studies with these inhibitors is difficult, and alternative approaches are needed to elucidate ROCK1's tissue specific metabolic functions. Fortunately, recent technological advances utilizing molecular carriers or genetic manipulation have facilitated discovery of ROCK1's tissue-specific mechanisms of action. In this article, we review the tissue-specific roles of ROCK1 in the regulation of energy balance and substrate utilization. We highlight prominent metabolic roles in liver, adipose, and skeletal muscle, in which ROCK1 regulates energy expenditure, glucose uptake, and lipid metabolism via inhibition of AMPK2α and paradoxical modulation of insulin signaling. Compared to ROCK1's roles in peripheral tissues, we also describe contradictory functions of ROCK1 in the hypothalamus to increase energy expenditure and decrease food intake via leptin signaling. Furthermore, dysregulated ROCK1 activity in either of these tissues results in metabolic disease phenotypes. Overall, tissue-specific approaches have made great strides in deciphering the many critical metabolic functions of ROCK1 and, ultimately, may facilitate the development of novel treatments for metabolic disorders.
Collapse
Affiliation(s)
- Taylor Landry
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, United States
- Department of Kinesiology, East Carolina University, Greenville, NC, United States
- Human Performance Laboratory, College of Human Performance and Health, East Carolina University, Greenville, NC, United States
| | - Daniel Shookster
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, United States
- Department of Kinesiology, East Carolina University, Greenville, NC, United States
- Human Performance Laboratory, College of Human Performance and Health, East Carolina University, Greenville, NC, United States
| | - Hu Huang
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, United States
- Department of Kinesiology, East Carolina University, Greenville, NC, United States
- Human Performance Laboratory, College of Human Performance and Health, East Carolina University, Greenville, NC, United States
- Department of Physiology, East Carolina University, Greenville, NC, United States
- *Correspondence: Hu Huang,
| |
Collapse
|
15
|
Wei L, Surma M, Yang Y, Tersey S, Shi J. ROCK2 inhibition enhances the thermogenic program in white and brown fat tissue in mice. FASEB J 2019; 34:474-493. [PMID: 31914704 DOI: 10.1096/fj.201901174rr] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 10/09/2019] [Accepted: 10/22/2019] [Indexed: 12/20/2022]
Abstract
The RhoA/ROCK-mediated actin cytoskeleton dynamics have been implicated in adipogenesis. The two ROCK isoforms, ROCK1 and ROCK2, are highly homologous. The contribution of ROCK2 to adipogenesis in vivo has not been elucidated. The present study aimed at the in vivo and in vitro roles of ROCK2 in the regulation of adipogenesis and the development of obesity. We performed molecular, histological, and metabolic analyses in ROCK2+/- and ROCK2+/KD mouse models, the latter harboring an allele with a kinase-dead (KD) mutation. Both ROCK2+/- and ROCK2+/KD mouse models showed a lean body mass phenotype during aging, associated with increased amounts of beige cells in subcutaneous white adipose tissue (sWAT) and increased thermogenic gene expression in all fat depots. ROCK2+/- mice on a high-fat diet showed increased energy expenditure accompanying by reduced obesity, and improved insulin sensitivity. In vitro differentiated ROCK2+/- stromal-vascular (SV) cells revealed increased beige adipogenesis associated with increased thermogenic gene expressions. Treatment with a selective ROCK2 inhibitor, KD025, to inhibit ROCK2 activity in differentiated SV cells reproduced the pro-beige phenotype of ROCK2+/- SV cells. In conclusion, ROCK2 activity-mediated actin cytoskeleton dynamics contribute to the inhibition of beige adipogenesis in WAT, and also promotes age-related and diet-induced fat mass gain and insulin resistance.
Collapse
Affiliation(s)
- Lei Wei
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA.,Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Michelle Surma
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yang Yang
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sarah Tersey
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA.,Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jianjian Shi
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
16
|
Wang L, Ma S, Ding Q, Wang X, Chen Y. CRISPR/Cas9-mediated MSTN gene editing induced mitochondrial alterations in C2C12 myoblast cells. ELECTRON J BIOTECHN 2019. [DOI: 10.1016/j.ejbt.2019.03.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
17
|
Tan Y, Ouyang H, Xiao X, Zhong J, Dong M. Irisin ameliorates septic cardiomyopathy via inhibiting DRP1-related mitochondrial fission and normalizing the JNK-LATS2 signaling pathway. Cell Stress Chaperones 2019; 24:595-608. [PMID: 30993599 PMCID: PMC6527615 DOI: 10.1007/s12192-019-00992-2] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 03/28/2019] [Accepted: 03/29/2019] [Indexed: 12/17/2022] Open
Abstract
Irisin plays a protective effect in acute and chronic myocardial damage, but its role in septic cardiomyopathy is unclear. The aim of our study was to explore the in vivo and in vitro effects of irisin using an LPS-induced septic cardiomyopathy model. Our results demonstrated that irisin treatment attenuated LPS-mediated cardiomyocyte death and myocardial dysfunction. At the molecular level, LPS application was associated with mitochondrial oxidative injury, cardiomyocyte ATP depletion and caspase-related apoptosis activation. In contrast, the irisin treatment sustained mitochondrial function by inhibiting DRP1-related mitochondrial fission and the reactivation of mitochondrial fission impaired the protective action of irisin on inflammation-attacked mitochondria and cardiomyocytes. Additionally, we found that irisin modulated DRP1-related mitochondrial fission through the JNK-LATS2 signaling pathway. JNK activation and/or LATS2 overexpression abolished the beneficial effects of irisin on LPS-mediated mitochondrial stress and cardiomyocyte death. Altogether, our results illustrate that LPS-mediated activation of DRP1-related mitochondrial fission through the JNK-LATS2 pathway participates in the pathogenesis of septic cardiomyopathy. Irisin could be used in the future as an effective therapy for sepsis-induced myocardial depression because it corrects DRP1-related mitochondrial fission and normalizes the JNK-LATS2 signaling pathway.
Collapse
Affiliation(s)
- Ying Tan
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China
| | - Haichun Ouyang
- Department of Cardiology, Shunde Hospital, Southern Medical University, Foshan, 528300 Guangdong China
| | - Xiaochan Xiao
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China
| | - Jiankai Zhong
- Department of Cardiology, Shunde Hospital, Southern Medical University, Foshan, 528300 Guangdong China
| | - Maolong Dong
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China
| |
Collapse
|
18
|
Liu R, Shi L, Peng N, Zhang Q, Li H. Higher Baseline Serum Irisin Decreases Risk for Body Mass Index Increment in Chinese Populations: A 3.2-Year Cohort Study. Diabetes Ther 2019; 10:713-723. [PMID: 30815828 PMCID: PMC6437307 DOI: 10.1007/s13300-019-0588-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Indexed: 01/23/2023] Open
Abstract
INTRODUCTION Irisin, a newly discovered myokine, exerts beneficial effects on energy metabolism. However, published results from studies examining the relationship between irisin concentration and obesity have been conflicting. The aim of our study was to investigate the association between serum irisin level and obese individuals with different body mass index (BMI) values and to explore the question of whether serum irisin can predict the risk of increases in the BMI. METHODS This study based on the data collected in the Risk Evaluation of cAncers in Chinese diabeTic Individuals: a lONgitudinal (REACTION). The cross-sectional cohort study was carried out from May 2011 to August 2011, and a longitudinal cohort study was conducted from July 2014 to October 2014 to complete the first 3.2-year follow-up. We enrolled 93 low-weight subjects (BMI < 18.5 kg/m2), 94 normal-weight subjects (BMI 18.5-23.9 kg/m2), 98 overweight subjects (BMI 24.0-27.9 kg/m2) and 93 obese subjects (BMI ≥ 28 kg/m2). Subjects in the normal-weight, overweight and obese groups were selected to match low-weight subjects by age and sex. Serum samples were obtained from all subjects to determine the irisin level. RESULTS Subjects with a higher serum irisin level tended to have significantly lower changes in BMI and body fat percentage and higher baseline high-density lipoprotein cholesterol level (p < 0.05). No significant correlation was observed between serum irisin level and the baseline obesity index. Serum irisin level was positively correlated to an active lifestyle (i.e. physical activity; β = 1.138, p = 0.032) and negatively correlated to fasting plasma glucose level (β = - 0.996, p = 0.023), changes in BMI (β = - 0.533, p = 0.002), waist circumference (β = - 0.102, p = 0.018), body fat percentage (β = - 0.457, p = 0.001) and Chinese visceral adiposity index (β = - 0.280, p = 0.028). After adjustment for cofactors, higher baseline serum irisin was an independent factor for a decreased BMI increment (baseline serum irisin: odds ratio 0.747, 95% confidence interval 0.652-0.949, p = 0.002). CONCLUSIONS Higher serum irisin at baseline independently predicted a lower BMI increment in Chinese populations.
Collapse
Affiliation(s)
- Ruoyi Liu
- Department of Endocrinology and Metabolism, Affiliated Hospital of Guizhou Medical University, No. 28, Guiyi Road, Guiyang, China
| | - Lixin Shi
- Department of Endocrinology and Metabolism, Affiliated Hospital of Guizhou Medical University, No. 28, Guiyi Road, Guiyang, China.
| | - Nianchun Peng
- Department of Endocrinology and Metabolism, Affiliated Hospital of Guizhou Medical University, No. 28, Guiyi Road, Guiyang, China
| | - Qiao Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Guizhou Medical University, No. 28, Guiyi Road, Guiyang, China
| | - Hong Li
- Department of Endocrinology and Metabolism, Affiliated Hospital of Guizhou Medical University, No. 28, Guiyi Road, Guiyang, China
| |
Collapse
|
19
|
Saadeldin MK, Elshaer SS, Emara IA, Maged M, Abdel-Aziz AK. Serum sclerostin and irisin as predictive markers for atherosclerosis in Egyptian type II diabetic female patients: A case control study. PLoS One 2018; 13:e0206761. [PMID: 30403705 PMCID: PMC6221312 DOI: 10.1371/journal.pone.0206761] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 10/18/2018] [Indexed: 12/22/2022] Open
Abstract
Diabetes mellitus represents a major independent risk factor for developing fatal cardiovascular diseases (CVDs) presumably through accelerating atherosclerosis; the underlying cause of most CVDs. Notably, this relative risk is reported to be higher in women than men. Endeavors directed towards identifying novel reliable predictive biomarkers are immensely thereby urged to improve the long-term outcome in these diabetic female patients. Sclerostin (SOST) is a Wnt signaling antagonist whereas irisin is a muscle-derived factor released after exercising which enhances browning of white adipose tissue. Emerging lines of evidence hint at potential crosstalk between them and CVDs. The present study aimed to assess the serum levels of SOST and irisin in Egyptian type 2 diabetic (T2DM) female patients with and without atherosclerosis and explore the possible relationship between both markers and other studied parameters among the studied cohorts. In this case-control study, 69 female subjects were enrolled; 39 type 2 diabetes patients with atherosclerosis (T2DM+ATHR), 22 type 2 diabetes patients without atherosclerosis (T2DM-ATHR) and 8 healthy controls. Their serum levels of SOST and irisin were assessed using ELISA. Significant increase in SOST levels were found in T2DM+ATHR compared to T2DM-ATHR and control (259.9 ±17.98 vs. 165.8±13.12 and 142.0±13.31 pg/mL respectively, P<0.001). Conversely, irisin levels were significantly lower in T2DM+ATHR (P<0.001) and T2DM-ATHR (P<0.01) compared to the control group (32.91±2.545 and 58.55±13.19 vs. 473.6±112.7 pg/mL). Interestingly, significant correlations between the levels of SOST and both irisin and fasting blood glucose were noticed in T2DM+ATHR group (r = 0.3754 and 0.3381 respectively, P<0.05). In conclusion, to the best of our knowledge, this study is the first to demonstrate the correlation between SOST and irisin levels in atherosclerotic T2DM female patients implying their potential implication in diabetic cardiovascular pathophysiology and supporting their use as reliable diagnostic/prognostic biomarkers for monitoring and preventing CVDs progression of T2DM female patients.
Collapse
Affiliation(s)
- Mona Kamal Saadeldin
- Department of Biochemistry, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
- Department of Biochemistry, National Institute of Diabetes and Endocrinology (NIDE), Cairo, Egypt
- Faculty of Biotechnology, October University for Modern Sciences and Arts, 6 October City, Cairo, Egypt
- * E-mail: (MKS); (SSE)
| | - Shereen Saeid Elshaer
- Department of Biochemistry, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
- * E-mail: (MKS); (SSE)
| | - Ibrahim Ali Emara
- Department of Biochemistry, National Institute of Diabetes and Endocrinology (NIDE), Cairo, Egypt
| | - Mohamad Maged
- Faculty of Biotechnology, October University for Modern Sciences and Arts, 6 October City, Cairo, Egypt
| | - Amal Kamal Abdel-Aziz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|
20
|
Chen K, Xu Z, Liu Y, Wang Z, Li Y, Xu X, Chen C, Xia T, Liao Q, Yao Y, Zeng C, He D, Yang Y, Tan T, Yi J, Zhou J, Zhu H, Ma J, Zeng C. Irisin protects mitochondria function during pulmonary ischemia/reperfusion injury. Sci Transl Med 2018; 9:9/418/eaao6298. [PMID: 29187642 DOI: 10.1126/scitranslmed.aao6298] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Revised: 08/10/2017] [Accepted: 10/20/2017] [Indexed: 12/13/2022]
Abstract
Limb remote ischemic preconditioning (RIPC) is an effective means of protection against ischemia/reperfusion (IR)-induced injury to multiple organs. Many studies are focused on identifying endocrine mechanisms that underlie the cross-talk between muscle and RIPC-mediated organ protection. We report that RIPC releases irisin, a myokine derived from the extracellular portion of fibronectin domain-containing 5 protein (FNDC5) in skeletal muscle, to protect against injury to the lung. Human patients with neonatal respiratory distress syndrome show reduced concentrations of irisin in the serum and increased irisin concentrations in the bronchoalveolar lavage fluid, suggesting transfer of irisin from circulation to the lung under physiologic stress. In mice, application of brief periods of ischemia preconditioning stimulates release of irisin into circulation and transfer of irisin to the lung subjected to IR injury. Irisin, via lipid raft-mediated endocytosis, enters alveolar cells and targets mitochondria. Interaction between irisin and mitochondrial uncoupling protein 2 (UCP2) allows for prevention of IR-induced oxidative stress and preservation of mitochondrial function. Animal model studies show that intravenous administration of exogenous irisin protects against IR-induced injury to the lung via improvement of mitochondrial function, whereas in UCP2-deficient mice or in the presence of a UCP2 inhibitor, the protective effect of irisin is compromised. These results demonstrate that irisin is a myokine that facilitates RIPC-mediated lung protection. Targeting the action of irisin in mitochondria presents a potential therapeutic intervention for pulmonary IR injury.
Collapse
Affiliation(s)
- Ken Chen
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing 400042, P.R. China.,Chongqing Institute of Cardiology, Chongqing 400042, P.R. China.,Department of Cardiology, Chengdu Military General Hospital, Chengdu, Sichuan 610083, P.R. China
| | - Zaicheng Xu
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing 400042, P.R. China.,Chongqing Institute of Cardiology, Chongqing 400042, P.R. China
| | - Yukai Liu
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing 400042, P.R. China.,Chongqing Institute of Cardiology, Chongqing 400042, P.R. China
| | - Zhen Wang
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing 400042, P.R. China.,Chongqing Institute of Cardiology, Chongqing 400042, P.R. China
| | - Yu Li
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing 400042, P.R. China.,Chongqing Institute of Cardiology, Chongqing 400042, P.R. China
| | - Xuefei Xu
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing 400042, P.R. China.,Chongqing Institute of Cardiology, Chongqing 400042, P.R. China
| | - Caiyu Chen
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing 400042, P.R. China.,Chongqing Institute of Cardiology, Chongqing 400042, P.R. China
| | - Tianyang Xia
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing 400042, P.R. China.,Chongqing Institute of Cardiology, Chongqing 400042, P.R. China
| | - Qiao Liao
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing 400042, P.R. China.,Chongqing Institute of Cardiology, Chongqing 400042, P.R. China
| | - Yonggang Yao
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing 400042, P.R. China.,Chongqing Institute of Cardiology, Chongqing 400042, P.R. China
| | - Cindy Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing 400042, P.R. China.,Chongqing Institute of Cardiology, Chongqing 400042, P.R. China
| | - Duofen He
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing 400042, P.R. China.,Chongqing Institute of Cardiology, Chongqing 400042, P.R. China
| | - Yongjian Yang
- Department of Cardiology, Chengdu Military General Hospital, Chengdu, Sichuan 610083, P.R. China
| | - Tao Tan
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Jianxun Yi
- Department of Physiology, Kansas City University, Kansas City, MO 64106, USA
| | - Jingsong Zhou
- Department of Physiology, Kansas City University, Kansas City, MO 64106, USA
| | - Hua Zhu
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Jianjie Ma
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA.
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, The Third Military Medical University, Chongqing 400042, P.R. China. .,Chongqing Institute of Cardiology, Chongqing 400042, P.R. China
| |
Collapse
|
21
|
Mahgoub MO, D'Souza C, Al Darmaki RSMH, Baniyas MMYH, Adeghate E. An update on the role of irisin in the regulation of endocrine and metabolic functions. Peptides 2018; 104:15-23. [PMID: 29608940 DOI: 10.1016/j.peptides.2018.03.018] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 03/29/2018] [Accepted: 03/29/2018] [Indexed: 02/07/2023]
Abstract
Irisin is a novel myokine and adipokine that has gained much attention recently due to its mechanisms of action. Irisin is secreted following proteolytic cleavage of its precursor fibronectin type III domain containing 5 (FNDC5). Following its release, irisin exerts its major action by increasing the expression of mitochondrial uncoupling protein 1 (UCP 1), which facilitates the conversion of white adipose tissue (WAT) into beige adipose tissue. Irisin is distributed in various body tissues and several actions have been attributed to its presence in those tissues. It has been suggested that it plays a role in metabolic diseases, ageing, inflammation and neurogenesis. However, the circulating levels of irisin are modulated by several factors such as diet, obesity, exercise, pharmacological agents and different pathological conditions. In this review, we have discussed the mechanisms by which irisin influences the functions of different body systems and how external factors in turn affect the circulating level of irisin. In conclusion, modification of circulating irisin level may help in the management of a variety of endocrine and metabolic disorders.
Collapse
Affiliation(s)
- Mohamed Omer Mahgoub
- Department of Anatomy, College of Medicine & Health Sciences, United Arab Emirates University, Post Box 17666, Al Ain, United Arab Emirates
| | - Crystal D'Souza
- Department of Anatomy, College of Medicine & Health Sciences, United Arab Emirates University, Post Box 17666, Al Ain, United Arab Emirates
| | - Reem S M H Al Darmaki
- Department of Anatomy, College of Medicine & Health Sciences, United Arab Emirates University, Post Box 17666, Al Ain, United Arab Emirates
| | - May M Y H Baniyas
- Department of Anatomy, College of Medicine & Health Sciences, United Arab Emirates University, Post Box 17666, Al Ain, United Arab Emirates
| | - Ernest Adeghate
- Department of Anatomy, College of Medicine & Health Sciences, United Arab Emirates University, Post Box 17666, Al Ain, United Arab Emirates.
| |
Collapse
|
22
|
Gaither MR, Gkafas GA, de Jong M, Sarigol F, Neat F, Regnier T, Moore D, Grӧcke DR, Hall N, Liu X, Kenny J, Lucaci A, Hughes M, Haldenby S, Hoelzel AR. Genomics of habitat choice and adaptive evolution in a deep-sea fish. Nat Ecol Evol 2018; 2:680-687. [DOI: 10.1038/s41559-018-0482-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Accepted: 01/22/2018] [Indexed: 12/30/2022]
|
23
|
Muñoz VR, Gaspar RC, Kuga GK, da Rocha AL, Crisol BM, Botezelli JD, Baptista IL, Mekary RA, da Silva ASR, Cintra DE, de Moura LP, Ropelle ER, Pauli JR. Exercise increases Rho-kinase activity and insulin signaling in skeletal muscle. J Cell Physiol 2018; 233:4791-4800. [PMID: 29219181 DOI: 10.1002/jcp.26278] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 11/14/2017] [Indexed: 12/16/2022]
Affiliation(s)
- Vitor R. Muñoz
- Laboratory of Molecular Biology of Exercise; University of Campinas (UNICAMP); Limeira São Paulo Brazil
| | - Rafael C. Gaspar
- Laboratory of Molecular Biology of Exercise; University of Campinas (UNICAMP); Limeira São Paulo Brazil
| | - Gabriel K. Kuga
- Laboratory of Molecular Biology of Exercise; University of Campinas (UNICAMP); Limeira São Paulo Brazil
| | - Alisson L. da Rocha
- Postgraduate Program in Rehabilitation and Functional Performance,; Ribeirão Preto Medical School, USP; Ribeirão Preto São Paulo Brazil
- School of Physical Education and Sport of Ribeirão Preto; University of São Paulo (USP); Ribeirão Preto São Paulo Brazil
| | - Barbara M. Crisol
- Laboratory of Molecular Biology of Exercise; University of Campinas (UNICAMP); Limeira São Paulo Brazil
| | - José D. Botezelli
- Laboratory of Molecular Biology of Exercise; University of Campinas (UNICAMP); Limeira São Paulo Brazil
| | - Igor L. Baptista
- Laboratory of Cell and Tissue Biology; University of Campinas (UNICAMP); Limeira São Paulo Brazil
| | - Rania A. Mekary
- Department of Nutrition; Harvard T. Chan School of Public Health; Boston Massachusetts
- Department of Pharmaceutical Business and Administrative Sciences; MCPHS University; Boston Massachusetts
| | - Adelino S. R. da Silva
- Laboratory of Cell and Tissue Biology; University of Campinas (UNICAMP); Limeira São Paulo Brazil
| | - Dennys E. Cintra
- Laboratory of Nutritional Genomics; University of Campinas (UNICAMP); Limeira São Paulo Brazil
- OCRC - Obesity and Comorbidities Research Center; University of Campinas (UNICAMP); Campinas São Paulo Brazil
| | - Leandro P. de Moura
- Laboratory of Molecular Biology of Exercise; University of Campinas (UNICAMP); Limeira São Paulo Brazil
- OCRC - Obesity and Comorbidities Research Center; University of Campinas (UNICAMP); Campinas São Paulo Brazil
- CEPECE - Center of Research in Sport Sciences. School of Applied Sciences; University of Campinas (UNICAMP); Limeira São Paulo Brazil
| | - Eduardo R. Ropelle
- Laboratory of Molecular Biology of Exercise; University of Campinas (UNICAMP); Limeira São Paulo Brazil
- OCRC - Obesity and Comorbidities Research Center; University of Campinas (UNICAMP); Campinas São Paulo Brazil
- CEPECE - Center of Research in Sport Sciences. School of Applied Sciences; University of Campinas (UNICAMP); Limeira São Paulo Brazil
| | - José R. Pauli
- Laboratory of Molecular Biology of Exercise; University of Campinas (UNICAMP); Limeira São Paulo Brazil
- OCRC - Obesity and Comorbidities Research Center; University of Campinas (UNICAMP); Campinas São Paulo Brazil
- CEPECE - Center of Research in Sport Sciences. School of Applied Sciences; University of Campinas (UNICAMP); Limeira São Paulo Brazil
| |
Collapse
|
24
|
Giudice J, Taylor JM. Muscle as a paracrine and endocrine organ. Curr Opin Pharmacol 2017; 34:49-55. [PMID: 28605657 PMCID: PMC5808999 DOI: 10.1016/j.coph.2017.05.005] [Citation(s) in RCA: 201] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 05/22/2017] [Accepted: 05/25/2017] [Indexed: 01/05/2023]
Abstract
Skeletal muscle cells are highly abundant and metabolically active and are known to 'communicate' their energy demands to other organs through active secretion. Muscle-derived secretory proteins include a variety of cytokines and peptides collectively referred to as 'myokines' that exert autocrine, paracrine or endocrine effects. Analyses of the skeletal muscle secretome revealed that numerous myokines are secreted in response to contraction or strength training, and that these factors not only regulate energy demand but also contribute to the broad beneficial effects of exercise on cardiovascular, metabolic, and mental health. Herein we review recent studies on the myokines that regulate muscle function and those that mediate cross talk between skeletal muscle and other organs including adipose tissue, liver, pancreas, the cardiovascular system, brain, bones, and skin.
Collapse
Affiliation(s)
- Jimena Giudice
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC 27599, USA; McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA.
| | - Joan M Taylor
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Pathology, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
25
|
The nuclear phosphatase SCP4 regulates FoxO transcription factors during muscle wasting in chronic kidney disease. Kidney Int 2017; 92:336-348. [PMID: 28506762 DOI: 10.1016/j.kint.2017.02.031] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 02/16/2017] [Accepted: 02/23/2017] [Indexed: 12/21/2022]
Abstract
Chronic kidney disease (CKD) and related inflammatory responses stimulate protein-energy wasting, a complication causing loss of muscle mass. Primarily, muscle wasting results from accelerated protein degradation via autophagic/lysosomal and proteasomal pathways, but mechanisms regulating these proteolysis pathways remain unclear. Since dephosphorylation of FoxOs regulates ubiquitin/proteasome protein metabolism, we tested whether a novel nuclear phosphatase, the small C-terminal domain phosphatase (SCP) 4, regulates FoxOs signaling and, in turn, muscle wasting. In cultured mouse myoblast cells, SCP4 overexpression stimulated proteolysis, while knockdown of SCP4 prevented the proteolysis stimulated by inflammatory cytokines. SCP4 overexpression led to nuclear accumulation of FoxO1/3a followed by increased expression of catabolic factors including myostatin, Atrogin-1, and MuRF-1, and induction of lysosomal-mediated proteolysis. Treatment of C2C12 myotubes with proinflammatory cytokines stimulated SCP4 expression in an NF-κB-dependent manner. In skeletal muscle of mice with CKD, SCP4 expression was up-regulated. Similarly, in skeletal muscle of patients with CKD, SCP4 expression was significantly increased. Knockdown of SCP4 significantly suppressed FoxO1/3a-mediated expression of Atrogin-1 and MuRF-1 and prevented muscle wasting in mice with CKD. Thus, SCP4 is a novel regulator of FoxO transcription factors and promotes cellular proteolysis. Hence, targeting SCP4 may prevent muscle wasting in CKD and possibly other catabolic conditions.
Collapse
|
26
|
Butt ZD, Hackett JD, Volkoff H. Irisin in goldfish (Carassius auratus): Effects of irisin injections on feeding behavior and expression of appetite regulators, uncoupling proteins and lipoprotein lipase, and fasting-induced changes in FNDC5 expression. Peptides 2017; 90:27-36. [PMID: 28219696 DOI: 10.1016/j.peptides.2017.02.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 02/06/2017] [Accepted: 02/13/2017] [Indexed: 12/21/2022]
Abstract
Irisin is a peptide cleaved from the fibronectin type III domain containing protein 5 (FNDC5) gene that is secreted predominantly by muscle cells but also by other tissues including brain and intestine. In mammals, irisin has been shown to have thermogenic actions via the modulation of uncoupling proteins (UCPs) and to affect feeding and energy homeostasis via actions in brain, adipose tissue, liver, muscle and gastrointestinal tract. To examine the role of irisin on feeding and metabolism in fish, the effects of peripheral (intraperitoneal) injections of irisin on feeding behavior, glucose levels and the mRNA expressions of appetite regulators (cocaine and amphetamine regulated transcript CART, agouti related protein AgRP, orexin), UCPs and lipoprotein lipase LPL and brain factors (brain-derived neurotrophic factor , BDNF and tyrosine hydroxylase TH) were assessed in brain, white muscle and intestine. Irisin injections (100ng/g) induced a decrease in food intake and increases in brain orexin, CART1 and CART2, UCP2, BDNF, muscle UCP2 and intestine LPL mRNA expressions but did not affect blood glucose levels, brain AgRP, TH, UCP1, UCP3 and LPL or muscle UCP1, UCP3 and LPL expressions. A partial goldfish FNDC5 cDNA was isolated and the expressions of FDNC5, UCPs, LPL and BDNF were also compared between fed and fasted fish. Fasting induced decreases FNDC5 mRNA expression in the brain and intestine, but not in muscle. Fasting also induced increases in brain BDNF and LPL expressions and increases in UCP1, UCP2, UCP3 and LPL expressions in muscle. Our result suggest that irisin is an anorexigenic factor in fish and its actions might be in part mediated by appetite-regulating factors such as CART and orexins as well as UCP2 and brain factors such as BDNF.
Collapse
Affiliation(s)
- Zahndra Diann Butt
- Departments of Biology and Biochemistry, Memorial University of Newfoundland, St. John's, NL A1 B 3X9, Canada
| | - Jessica Dalton Hackett
- Departments of Biology and Biochemistry, Memorial University of Newfoundland, St. John's, NL A1 B 3X9, Canada
| | - Hélène Volkoff
- Departments of Biology and Biochemistry, Memorial University of Newfoundland, St. John's, NL A1 B 3X9, Canada.
| |
Collapse
|