1
|
Sun Q, Cui X, Yin D, Li J, Li J, Du L. Molecular mechanisms of UCP1-independent thermogenesis: the role of futile cycles in energy dissipation. J Physiol Biochem 2025:10.1007/s13105-025-01090-x. [PMID: 40380026 DOI: 10.1007/s13105-025-01090-x] [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: 12/14/2024] [Accepted: 05/01/2025] [Indexed: 05/19/2025]
Abstract
Adipose tissue thermogenesis has emerged as a prominent research focus for the treatment of metabolic diseases, particularly through mitochondrial uncoupling, which oxidizes nutrients to produce heat rather than synthesizing ATP. Uncoupling protein 1 (UCP1) has garnered significant attention as a core protein mediating non-shivering thermogenesis(NST). However, recent studies indicate that energy dissipation can also occur via UCP1-independent thermogenesis, partially driven by futile metabolic cycles. These cycles involve ATP depletion coupled with reversible energy reactions, resulting in futile energy expenditure. Unlike classical UCP1-mediated thermogenesis, futile cycling is not confined to brown and beige adipose tissue, suggesting a broader range of therapeutic targets. These findings open new avenues for targeting these pathways to enhance metabolic health. This review explores the characteristics and distinctions of the primary metabolic organs (adipose tissue, liver, and skeletal muscle) involved in the futile cycles of thermogenesis. It further elaborates on the cellular and molecular mechanisms underlying calcium, creatine, and lipid cycling, emphasizing their strengths, limitations, and roles beyond thermogenesis.
Collapse
Affiliation(s)
- Quanhao Sun
- First Clinical School of Medicine, Heilongjiang University of Chinese Medicine, Harbin, 150000, China
| | - Xinyue Cui
- First Clinical School of Medicine, Heilongjiang University of Chinese Medicine, Harbin, 150000, China
| | - Dong Yin
- First Clinical School of Medicine, Heilongjiang University of Chinese Medicine, Harbin, 150000, China
| | - Juan Li
- First Clinical School of Medicine, Heilongjiang University of Chinese Medicine, Harbin, 150000, China
| | - Jiarui Li
- First Clinical School of Medicine, Heilongjiang University of Chinese Medicine, Harbin, 150000, China
| | - Likun Du
- Department of Endocrinology, First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, 150040, China.
| |
Collapse
|
2
|
Wang P, Li J, Li CG, Zhou X, Chen X, Zhu M, Wang H. Restoring Autophagy by Exercise Ameliorates Insulin Resistance Partly via Calcineurin-Driven TFEB Nuclear Translocation. Clin Exp Pharmacol Physiol 2025; 52:e70010. [PMID: 39787618 DOI: 10.1111/1440-1681.70010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 10/21/2024] [Accepted: 10/24/2024] [Indexed: 01/12/2025]
Abstract
Exercise activates autophagy and lysosome system in skeletal muscle, which are known to play an important role in metabolic adaptation. However, the mechanism of exercise-activated autophagy and lysosome system in obese insulin resistance remains covert. In this study, we investigated the role of exercise-induced activation of autophagy and lysosome system in improving glucose metabolism of skeletal muscle. Male C57BL/6 mice were randomly divided into five groups: the chow diet (CD) group, the high-fat diet (HFD) group, the high-fat diet plus exercise (HFD-E) group and the HFD-E treated with calcineurin inhibitor FK506 (HFD-E-F) or saline (HFD-E-S) groups. The mice in exercise groups (HFD-E, HFD-E-F and HFD-E-S) were subjected to aerobic treadmill exercise (speed at 12 m/min for 1 h per session, 0° slope, 5 days per week for 12 weeks). Mice of HFD-E-F group were intraperitoneally administered FK506 (1 mg/kg), once each day for 2 weeks before the end of exercise. Expressions pTFEB, T-TFEB and autophagy-lysosome markers, including Beclin1, LC3, ULK1, SQSTM1, LAMP1, CTSD and CTSL proteins in gastrocnemius muscle were analysed. We demonstrated that HFD induced insulin resistance and decreased autophagy-lysosomal proteins and the exercise significantly increased transcription factor EB (TFEB) translocation from the cytoplasm to the nucleus, restored the impaired autophagy-lysosomal-related protein expressions, and improved glucose metabolism. The increase in TFEB nuclear translocation was partly blocked by the calcineurin inhibitor FK506. Our results suggest that exercise promotes autophagy and lysosome restoration by regulating calcineurin-mediated TFEB nuclear translocation, ultimately alleviating HFD-induced insulin resistance in mice skeletal muscle.
Collapse
Affiliation(s)
- Ping Wang
- School of Physical Education, Hangzhou Normal University, Hangzhou, China
| | - Jiaxin Li
- School of Physical Education, Hangzhou Normal University, Hangzhou, China
| | - Chun Guang Li
- NICM Health Research Institute, Western Sydney University, Westmead, New South Wales, Australia
| | - Xian Zhou
- NICM Health Research Institute, Western Sydney University, Westmead, New South Wales, Australia
| | - Xiaolong Chen
- School of Physical Education, Hangzhou Normal University, Hangzhou, China
| | - Minghua Zhu
- Department of Cardiothoracic Surgery, The Affiliated Hospital of Hangzhou Normal University, Hangzhou, China
| | - Hongjiang Wang
- School of Physical Education, Hangzhou Normal University, Hangzhou, China
| |
Collapse
|
3
|
Salagre D, Bajit H, Fernández-Vázquez G, Dwairy M, Garzón I, Haro-López R, Agil A. Melatonin induces fiber switching by improvement of mitochondrial oxidative capacity and function via NRF2/RCAN/MEF2 in the vastus lateralis muscle from both sex Zücker diabetic fatty rats. Free Radic Biol Med 2025; 227:322-335. [PMID: 39645208 DOI: 10.1016/j.freeradbiomed.2024.12.019] [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: 07/09/2024] [Revised: 11/19/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
The positive role of melatonin in obesity control and skeletal muscle (SKM) preservation is well known. We recently showed that melatonin improves vastus lateralis muscle (VL) fiber oxidative phenotype. However, fiber type characterization, mitochondrial function, and molecular mechanisms that underlie VL fiber switching by melatonin are still undefined. Our study aims to investigate whether melatonin induces fiber switching by NRF2/RCAN/MEF2 pathway activation and mitochondrial oxidative metabolism modulation in the VL of both sex Zücker diabetic fatty (ZDF) rats. 5-Weeks-old male and female ZDF rats (N = 16) and their age-matched lean littermates (ZL) were subdivided into two subgroups: control (C) and orally treated with melatonin (M) (10 mg/kg/day) for 12 weeks. Interestingly, melatonin increased oxidative fibers amounts (Types I and IIa) counteracting the decreased levels found in the VL of obese-diabetic rats, and upregulated NRF2, calcineurin and MEF2 expression. Melatonin also restored the mitochondrial oxidative capacity increasing the respiratory control ratio (RCR) in both sex and phenotype rats through the reduction of the proton leak component of respiration (state 4). Melatonin also improved the VL mitochondrial phosphorylation coefficient and modulated the total oxygen consumption by enhancing complex I, III and IV activity, and fatty acid oxidation (FAO) in both sex obese-diabetic rats, decreasing in male and increasing in female the complex II oxygen consumption. These findings suggest that melatonin treatment induces fiber switching in SKM improving mitochondrial functionality by NRF2/RCAN/MEF2 pathway activation.
Collapse
Affiliation(s)
- Diego Salagre
- Department of Pharmacology, BioHealth Institute Granada (IBs Granada), Neuroscience Institute (CIBM), School of Medicine, University of Granada, 18016, Granada, Spain
| | - Habiba Bajit
- Department of Pharmacology, BioHealth Institute Granada (IBs Granada), Neuroscience Institute (CIBM), School of Medicine, University of Granada, 18016, Granada, Spain
| | | | - Mutaz Dwairy
- Department of Civil Engineering, Yarmuk University, 21163, Irbid, Jordan
| | - Ingrid Garzón
- Tissue Engineering Group, Department of Histology, BioHealth Institute Granada (IBs Granada), School of Medicine, University of Granada, 18016, Granada, Spain
| | - Rocío Haro-López
- Department of Pharmacology, BioHealth Institute Granada (IBs Granada), Neuroscience Institute (CIBM), School of Medicine, University of Granada, 18016, Granada, Spain
| | - Ahmad Agil
- Department of Pharmacology, BioHealth Institute Granada (IBs Granada), Neuroscience Institute (CIBM), School of Medicine, University of Granada, 18016, Granada, Spain.
| |
Collapse
|
4
|
Ribieras AJ, Ortiz YY, Li Y, Le NT, Huerta CT, Voza FA, Shao H, Vazquez-Padron RI, Liu ZJ, Velazquez OC. E-Selectin/AAV Gene Therapy Promotes Myogenesis and Skeletal Muscle Recovery in a Mouse Hindlimb Ischemia Model. Cardiovasc Ther 2023; 2023:6679390. [PMID: 37251271 PMCID: PMC10219778 DOI: 10.1155/2023/6679390] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/25/2023] [Accepted: 05/10/2023] [Indexed: 05/31/2023] Open
Abstract
The response to ischemia in peripheral artery disease (PAD) depends on compensatory neovascularization and coordination of tissue regeneration. Identifying novel mechanisms regulating these processes is critical to the development of nonsurgical treatments for PAD. E-selectin is an adhesion molecule that mediates cell recruitment during neovascularization. Therapeutic priming of ischemic limb tissues with intramuscular E-selectin gene therapy promotes angiogenesis and reduces tissue loss in a murine hindlimb gangrene model. In this study, we evaluated the effects of E-selectin gene therapy on skeletal muscle recovery, specifically focusing on exercise performance and myofiber regeneration. C57BL/6J mice were treated with intramuscular E-selectin/adeno-associated virus serotype 2/2 gene therapy (E-sel/AAV) or LacZ/AAV2/2 (LacZ/AAV) as control and then subjected to femoral artery coagulation. Recovery of hindlimb perfusion was assessed by laser Doppler perfusion imaging and muscle function by treadmill exhaustion and grip strength testing. After three postoperative weeks, hindlimb muscle was harvested for immunofluorescence analysis. At all postoperative time points, mice treated with E-sel/AAV had improved hindlimb perfusion and exercise capacity. E-sel/AAV gene therapy also increased the coexpression of MyoD and Ki-67 in skeletal muscle progenitors and the proportion of Myh7+ myofibers. Altogether, our findings demonstrate that in addition to improving reperfusion, intramuscular E-sel/AAV gene therapy enhances the regeneration of ischemic skeletal muscle with a corresponding benefit on exercise performance. These results suggest a potential role for E-sel/AAV gene therapy as a nonsurgical adjunct in patients with life-limiting PAD.
Collapse
Affiliation(s)
- Antoine J. Ribieras
- Division of Vascular Surgery, DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Yulexi Y. Ortiz
- Division of Vascular Surgery, DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Yan Li
- Division of Vascular Surgery, DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Nga T. Le
- Division of Vascular Surgery, DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Carlos T. Huerta
- Division of Vascular Surgery, DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Francesca A. Voza
- Division of Vascular Surgery, DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Hongwei Shao
- Division of Vascular Surgery, DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Roberto I. Vazquez-Padron
- Division of Vascular Surgery, DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Zhao-Jun Liu
- Division of Vascular Surgery, DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Omaida C. Velazquez
- Division of Vascular Surgery, DeWitt Daughtry Family Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Vascular Biology Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
5
|
Baranowski RW, Braun JL, Vandenboom R, Fajardo VA. Neurogranin inhibits calcineurin in murine soleus muscle: Effects of heterozygous knockdown on muscle adaptations to tenotomy and fatigue resistance. Biochem Biophys Res Commun 2022; 623:89-95. [PMID: 35878428 DOI: 10.1016/j.bbrc.2022.07.062] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 07/14/2022] [Accepted: 07/15/2022] [Indexed: 11/16/2022]
Abstract
Neurogranin (Ng) is a calmodulin (CaM) binding protein that negatively regulates calcineurin - a Ca2+/CaM-dependent phosphatase that can mitigate the slow-to-fast fibre type shift observed with muscle unloading. Here, we questioned whether heterozygous deletion of Ng (Ng+/-) would enhance calcineurin activity, thereby minimizing the slow-to-fast fibre type shift caused by muscle unloading. As expected, soleus muscles from young adult (3-4 months old) Ng± mice had lowered Ng content and enhanced calcineurin activity when compared to soleus muscles obtained from male age-matched wild-type (WT) mice. Two weeks after tenotomy surgery, where the soleus and gastrocnemius tendons were severed, soleus total fibre count were found to be similarly reduced across both genotypes. However, significant reductions in myofibre cross-sectional area were only found in WT mice and not Ng± mice. Furthermore, while soleus muscles from both WT and Ng± mice exhibited a slow-to-fast fibre type shift with tenotomy, soleus muscles from Ng± mice, in both sham and tenotomized conditions, had a greater proportion of oxidative fibres (type I and IIA) compared with that of WT mice. Corresponding well with this, we found that soleus muscles from Ng± mice were more fatigue resistant compared with those obtained from their WT counterparts. Collectively, these findings show that heterozygous Ng deletion increases calcineurin activation, preserves myofibre size in response to unloading, and promotes the oxidative fibre type to ultimately enhance fatigue resistance. This study demonstrates the role of Ng in regulating calcineurin in vivo and its influence on skeletal muscle form and function.
Collapse
Affiliation(s)
- Ryan W Baranowski
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, L2S 3A1, Canada; Department of Kinesiology, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Jessica L Braun
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, L2S 3A1, Canada; Department of Kinesiology, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Rene Vandenboom
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, L2S 3A1, Canada; Department of Kinesiology, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Val A Fajardo
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, L2S 3A1, Canada; Department of Kinesiology, Brock University, St. Catharines, ON, L2S 3A1, Canada.
| |
Collapse
|
6
|
Braun JL, Ryoo J, Goodwin K, Copeland EN, Geromella MS, Baranowski RW, MacPherson REK, Fajardo VA. The effects of neurogranin knockdown on SERCA pump efficiency in soleus muscles of female mice fed a high fat diet. Front Endocrinol (Lausanne) 2022; 13:957182. [PMID: 36072929 PMCID: PMC9441848 DOI: 10.3389/fendo.2022.957182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 08/05/2022] [Indexed: 11/26/2022] Open
Abstract
The sarco(endo)plasmic reticulum Ca2+ ATPase (SERCA) pump is responsible for the transport of Ca2+ from the cytosol into the sarcoplasmic reticulum at the expense of ATP, making it a regulator of both muscle relaxation and muscle-based energy expenditure. Neurogranin (Ng) is a small protein that negatively regulates calcineurin signaling. Calcineurin is Ca2+/calmodulin dependent phosphatase that promotes the oxidative fibre type in skeletal muscle and regulates muscle-based energy expenditure. A recent study has shown that calcineurin activation reduces SERCA Ca2+ transport efficiency, ultimately raising energy expenditure. Since the biomedical view of obesity states that it arises as an imbalance between energy intake and expenditure which favors the former, we questioned whether heterozygous Ng deletion (Ng+/- ) would reduce SERCA efficiency and increase energy expenditure in female mice fed a high-fat diet (HFD). Young (3-4-month-old) female wild type (WT) and Ng+/- mice were fed a HFD for 12 weeks with their metabolic profile being analyzed using metabolic cages and DXA scanning, while soleus SERCA efficiency was measured using SERCA specific Ca2+ uptake and ATPase activity assays. Ng+/- mice showed significantly less cage ambulation compared to WT mice but this did not lead to any added weight gain nor changes in daily energy expenditure, glucose or insulin tolerance despite a similar level of food intake. Furthermore, we observed significant reductions in SERCA's apparent coupling ratio which were associated with significant reductions in SERCA1 and phospholamban content. Thus, our results show that Ng regulates SERCA pump efficiency, and future studies should further investigate the potential cellular mechanisms.
Collapse
Affiliation(s)
- Jessica L. Braun
- Department of Kinesiology, Brock University, St. Catharines, ON, Canada
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, Canada
- Centre for Neuroscience, Brock University, St. Catharines, ON, Canada
| | - Jisook Ryoo
- Department of Kinesiology, Brock University, St. Catharines, ON, Canada
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, Canada
| | - Kyle Goodwin
- Department of Kinesiology, Brock University, St. Catharines, ON, Canada
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, Canada
| | - Emily N. Copeland
- Department of Kinesiology, Brock University, St. Catharines, ON, Canada
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, Canada
- Centre for Neuroscience, Brock University, St. Catharines, ON, Canada
| | - Mia S. Geromella
- Department of Kinesiology, Brock University, St. Catharines, ON, Canada
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, Canada
| | - Ryan W. Baranowski
- Department of Kinesiology, Brock University, St. Catharines, ON, Canada
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, Canada
| | - Rebecca E. K. MacPherson
- Centre for Neuroscience, Brock University, St. Catharines, ON, Canada
- Department of Health Sciences, Brock University, St. Catharines, ON, Canada
| | - Val A. Fajardo
- Department of Kinesiology, Brock University, St. Catharines, ON, Canada
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, Canada
- Centre for Neuroscience, Brock University, St. Catharines, ON, Canada
- *Correspondence: Val A. Fajardo,
| |
Collapse
|
7
|
Wang P, Li CG, Zhou X, Ding S. Transcription factor EB enhances autophagy and ameliorates palmitate-induced insulin resistance at least partly via upregulating AMPK activity in skeletal muscle cells. Clin Exp Pharmacol Physiol 2021; 49:302-310. [PMID: 34614219 DOI: 10.1111/1440-1681.13600] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 10/01/2021] [Accepted: 10/01/2021] [Indexed: 12/22/2022]
Abstract
This study aimed to elucidate the role of transcription factor EB (TFEB) in protecting C2C12 myotubes against palmitate (PA)-induced insulin resistance (IR) and explored its mechanism associated with autophagy. PA treatment significantly decreased insulin sensitivity in myotubes and downregulated TFEB protein expression. TFEB overexpression significantly reversed the PA-suppressed glucose transporter 4 (GLUT4) protein expression and improved intracellular glucose uptake and consumption, and also alleviated the decrease of autophagy markers induced by PA. The effect of TFEB overexpression on GLUT4 was also abolished by the autophagy inhibitor 3-MA. In addition, AMPKɑ2-DN inhibited or abolished the effects of TFEB overexpression on upregulation of GLUT4 and PA-induced decrease of autophagy marker expressions. Taken together, our data demonstrated that upregulation of TFEB improved PA-induced IR in C2C12 myotubes by enhancing autophagy and upregulating AMPK activity. TFEB, as a critical regulator of glucose homeostasis in skeletal muscle cells, may be a potential therapeutic target for IR and Type 2 diabetes.
Collapse
Affiliation(s)
- Ping Wang
- School of Physical Education, Hangzhou Normal University, Hangzhou, China
| | - Chun Guang Li
- NICM Health Research Institute, Western Sydney University, Westmead, New South Wales, Australia
| | - Xian Zhou
- NICM Health Research Institute, Western Sydney University, Westmead, New South Wales, Australia
| | - Shuzhe Ding
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention, Ministry of Education, East China Normal University, Shanghai, China.,School of Physical Education & Health, East China Normal University, Shanghai, China
| |
Collapse
|
8
|
Morton SU, Sefton CR, Zhang H, Dai M, Turner DL, Uhler MD, Agrawal PB. microRNA-mRNA Profile of Skeletal Muscle Differentiation and Relevance to Congenital Myotonic Dystrophy. Int J Mol Sci 2021; 22:ijms22052692. [PMID: 33799993 PMCID: PMC7962092 DOI: 10.3390/ijms22052692] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 02/25/2021] [Accepted: 03/04/2021] [Indexed: 01/08/2023] Open
Abstract
microRNAs (miRNAs) regulate messenger RNA (mRNA) abundance and translation during key developmental processes including muscle differentiation. Assessment of miRNA targets can provide insight into muscle biology and gene expression profiles altered by disease. mRNA and miRNA libraries were generated from C2C12 myoblasts during differentiation, and predicted miRNA targets were identified based on presence of miRNA binding sites and reciprocal expression. Seventeen miRNAs were differentially expressed at all time intervals (comparing days 0, 2, and 5) of differentiation. mRNA targets of differentially expressed miRNAs were enriched for functions related to calcium signaling and sarcomere formation. To evaluate this relationship in a disease state, we evaluated the miRNAs differentially expressed in human congenital myotonic dystrophy (CMD) myoblasts and compared with normal control. Seventy-four miRNAs were differentially expressed during healthy human myocyte maturation, of which only 12 were also up- or downregulated in CMD patient cells. The 62 miRNAs that were only differentially expressed in healthy cells were compared with differentiating C2C12 cells. Eighteen of the 62 were conserved in mouse and up- or down-regulated during mouse myoblast differentiation, and their C2C12 targets were enriched for functions related to muscle differentiation and contraction.
Collapse
Affiliation(s)
- Sarah U. Morton
- Division of Newborn Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
- Correspondence: (S.U.M.); (P.B.A.)
| | | | - Huanqing Zhang
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA; (H.Z.); (M.D.); (D.L.T.); (M.D.U.)
| | - Manhong Dai
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA; (H.Z.); (M.D.); (D.L.T.); (M.D.U.)
| | - David L. Turner
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA; (H.Z.); (M.D.); (D.L.T.); (M.D.U.)
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Michael D. Uhler
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI 48109, USA; (H.Z.); (M.D.); (D.L.T.); (M.D.U.)
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Pankaj B. Agrawal
- Division of Newborn Medicine, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA 02115, USA
- Division of Genetics and Genomics, Boston Children’s Hospital, Boston, MA 02115, USA
- The Manton Center for Orphan Disease Research, Boston Children’s Hospital, Boston, MA 02115, USA
- Correspondence: (S.U.M.); (P.B.A.)
| |
Collapse
|
9
|
Moradi F, Copeland EN, Baranowski RW, Scholey AE, Stuart JA, Fajardo VA. Calmodulin-Binding Proteins in Muscle: A Minireview on Nuclear Receptor Interacting Protein, Neurogranin, and Growth-Associated Protein 43. Int J Mol Sci 2020; 21:E1016. [PMID: 32033037 PMCID: PMC7038096 DOI: 10.3390/ijms21031016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/27/2020] [Accepted: 01/31/2020] [Indexed: 01/26/2023] Open
Abstract
Calmodulin (CaM) is an important Ca2+-sensing protein with numerous downstream targets that are either CaM-dependant or CaM-regulated. In muscle, CaM-dependent proteins, which are critical regulators of dynamic Ca2+ handling and contractility, include calcineurin (CaN), CaM-dependant kinase II (CaMKII), ryanodine receptor (RyR), and dihydropyridine receptor (DHPR). CaM-regulated targets include genes associated with oxidative metabolism, muscle plasticity, and repair. Despite its importance in muscle, the regulation of CaM-particularly its availability to bind to and activate downstream targets-is an emerging area of research. In this minireview, we discuss recent studies revealing the importance of small IQ motif proteins that bind to CaM to either facilitate (nuclear receptor interacting protein; NRIP) its activation of downstream targets, or sequester (neurogranin, Ng; and growth-associated protein 43, GAP43) CaM away from their downstream targets. Specifically, we discuss recent studies that have begun uncovering the physiological roles of NRIP, Ng, and GAP43 in skeletal and cardiac muscle, thereby highlighting the importance of endogenously expressed CaM-binding proteins and their regulation of CaM in muscle.
Collapse
Affiliation(s)
- Fereshteh Moradi
- Department of Biological Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada; (F.M.); (J.A.S.)
| | - Emily N. Copeland
- Centre for Neuroscience, Brock University, St. Catharines, ON L2S 3A1, Canada;
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON L2S 3A1, Canada;
| | - Ryan W. Baranowski
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON L2S 3A1, Canada;
- Department of Kinesiology, Brock University, St. Catharines, ON L2S 3A1, Canada;
| | - Aiden E. Scholey
- Department of Kinesiology, Brock University, St. Catharines, ON L2S 3A1, Canada;
| | - Jeffrey A. Stuart
- Department of Biological Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada; (F.M.); (J.A.S.)
| | - Val A. Fajardo
- Centre for Neuroscience, Brock University, St. Catharines, ON L2S 3A1, Canada;
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON L2S 3A1, Canada;
- Department of Kinesiology, Brock University, St. Catharines, ON L2S 3A1, Canada;
| |
Collapse
|
10
|
Andrade-Souza VA, Ghiarone T, Sansonio A, Santos Silva KA, Tomazini F, Arcoverde L, Fyfe J, Perri E, Saner N, Kuang J, Bertuzzi R, Leandro CG, Bishop DJ, Lima-Silva AE. Exercise twice-a-day potentiates markers of mitochondrial biogenesis in men. FASEB J 2019; 34:1602-1619. [PMID: 31914620 DOI: 10.1096/fj.201901207rr] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 10/31/2019] [Accepted: 11/04/2019] [Indexed: 12/18/2022]
Abstract
Endurance exercise begun with reduced muscle glycogen stores seems to potentiate skeletal muscle protein abundance and gene expression. However, it is unknown whether this greater signaling responses is due to performing two exercise sessions in close proximity-as a first exercise session is necessary to reduce the muscle glycogen stores. In the present study, we manipulated the recovery duration between a first muscle glycogen-depleting exercise and a second exercise session, such that the second exercise session started with reduced muscle glycogen in both approaches but was performed either 2 or 15 hours after the first exercise session (so-called "twice-a-day" and "once-daily" approaches, respectively). We found that exercise twice-a-day increased the nuclear abundance of transcription factor EB (TFEB) and nuclear factor of activated T cells (NFAT) and potentiated the transcription of peroxisome proliferator-activated receptor-ɣ coactivator 1-alpha (PGC-1α), peroxisome proliferator-activated receptor-alpha (PPARα), and peroxisome proliferator-activated receptor beta/delta (PPARβ/δ) genes, in comparison with the once-daily exercise. These results suggest that part of the elevated molecular signaling reported with previous "train-low" approaches might be attributed to performing two exercise sessions in close proximity. The twice-a-day approach might be an effective strategy to induce adaptations related to mitochondrial biogenesis and fat oxidation.
Collapse
Affiliation(s)
- Victor Amorim Andrade-Souza
- Department of Physical Education and Sports Science, Academic Center of Vitoria, Federal University of Pernambuco, Vitória de Santo Antão, PE, Brazil
| | - Thaysa Ghiarone
- Department of Physical Education and Sports Science, Academic Center of Vitoria, Federal University of Pernambuco, Vitória de Santo Antão, PE, Brazil
| | - Andre Sansonio
- Department of Physical Education and Sports Science, Academic Center of Vitoria, Federal University of Pernambuco, Vitória de Santo Antão, PE, Brazil
| | - Kleiton Augusto Santos Silva
- Department of Physical Education and Sports Science, Academic Center of Vitoria, Federal University of Pernambuco, Vitória de Santo Antão, PE, Brazil.,Department of Medicine, University of Missouri School of Medicine, Columbia, MI, USA
| | - Fabiano Tomazini
- Department of Physical Education and Sports Science, Academic Center of Vitoria, Federal University of Pernambuco, Vitória de Santo Antão, PE, Brazil
| | - Lucyana Arcoverde
- Department of Physical Education and Sports Science, Academic Center of Vitoria, Federal University of Pernambuco, Vitória de Santo Antão, PE, Brazil
| | - Jackson Fyfe
- School of Exercise and Nutrition Sciences, Faculty of Health, Deakin University, Burwood, VIC, Australia
| | - Enrico Perri
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Nicholas Saner
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Jujiao Kuang
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Romulo Bertuzzi
- Endurance Performance Research Group, School of Physical Education and Sport, University of São Paulo, São Paulo, SP, Brazil
| | - Carol Gois Leandro
- Department of Physical Education and Sports Science, Academic Center of Vitoria, Federal University of Pernambuco, Vitória de Santo Antão, PE, Brazil
| | - David John Bishop
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia.,School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| | - Adriano Eduardo Lima-Silva
- Department of Physical Education and Sports Science, Academic Center of Vitoria, Federal University of Pernambuco, Vitória de Santo Antão, PE, Brazil.,Human Performance Research Group, Academic Department of Physical Education, Technological Federal University of Paraná, Curitiba, PR, Brazil
| |
Collapse
|
11
|
Calcineurin controls gene transcription following stimulation of a Gαq-coupled designer receptor. Exp Cell Res 2019; 383:111553. [DOI: 10.1016/j.yexcr.2019.111553] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 08/07/2019] [Accepted: 08/10/2019] [Indexed: 01/07/2023]
|
12
|
Papadimitriou ID, Eynon N, Yan X, Munson F, Jacques M, Kuang J, Voisin S, North KN, Bishop DJ. A "human knockout" model to investigate the influence of the α-actinin-3 protein on exercise-induced mitochondrial adaptations. Sci Rep 2019; 9:12688. [PMID: 31481717 PMCID: PMC6722100 DOI: 10.1038/s41598-019-49042-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 08/06/2019] [Indexed: 11/09/2022] Open
Abstract
Research in α-actinin-3 knockout mice suggests a novel role for α-actinin-3 as a mediator of cell signalling. We took advantage of naturally-occurring human “knockouts” (lacking α-actinin-3 protein) to investigate the consequences of α-actinin-3 deficiency on exercise-induced changes in mitochondrial-related genes and proteins, as well as endurance training adaptations. At baseline, we observed a compensatory increase of α-actinin-2 protein in ACTN3 XX (α-actinin-3 deficient; n = 18) vs ACTN3 RR (expressing α-actinin-3; n = 19) participants but no differences between genotypes for markers of aerobic fitness or mitochondrial content and function. There was a main effect of genotype, without an interaction, for RCAN1-4 protein content (a marker of calcineurin activity). However, there was no effect of genotype on exercise-induced expression of genes associated with mitochondrial biogenesis, nor post-training physiological changes. In contrast to results in mice, loss of α-actinin-3 is not associated with higher baseline endurance-related phenotypes, or greater adaptations to endurance exercise training in humans.
Collapse
Affiliation(s)
- I D Papadimitriou
- Institute for Health and Sport (iHeS), Victoria University, Melbourne, Australia.,Department of Physiology, Mahidol University, Bangkok, Thailand
| | - N Eynon
- Institute for Health and Sport (iHeS), Victoria University, Melbourne, Australia.,Murdoch Children's Research Institute, Melbourne, Australia
| | - X Yan
- Institute for Health and Sport (iHeS), Victoria University, Melbourne, Australia
| | - F Munson
- Institute for Health and Sport (iHeS), Victoria University, Melbourne, Australia
| | - M Jacques
- Institute for Health and Sport (iHeS), Victoria University, Melbourne, Australia
| | - J Kuang
- Institute for Health and Sport (iHeS), Victoria University, Melbourne, Australia
| | - S Voisin
- Institute for Health and Sport (iHeS), Victoria University, Melbourne, Australia
| | - K N North
- Murdoch Children's Research Institute, Melbourne, Australia
| | - D J Bishop
- Institute for Health and Sport (iHeS), Victoria University, Melbourne, Australia. .,School of Medical & Health Sciences, Edith Cowan University, Joondalup, Australia.
| |
Collapse
|
13
|
Fajardo VA, Chambers PJ, Juracic ES, Rietze BA, Gamu D, Bellissimo C, Kwon F, Quadrilatero J, Russell Tupling A. Sarcolipin deletion in mdx mice impairs calcineurin signalling and worsens dystrophic pathology. Hum Mol Genet 2019; 27:4094-4102. [PMID: 30137316 DOI: 10.1093/hmg/ddy302] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 08/15/2018] [Indexed: 12/11/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is the most severe form of muscular dystrophy affecting 1 in 3500 live male births. Although there is no cure for DMD, therapeutic strategies aimed at enhancing calcineurin signalling and promoting the slow fibre phenotype have shown promise in mdx mice, which is the classical mouse model for DMD. Sarcolipin (SLN) is a small protein that regulates the sarco(endo)plasmic reticulum Ca2+-ATPase pump and its expression is highly upregulated in dystrophic skeletal muscle. We have recently shown that SLN in skeletal muscle amplifies calcineurin signalling thereby increasing myofibre size and the slow fibre phenotype. Therefore, in the present study we sought to determine the physiological impact of genetic Sln deletion in mdx mice, particularly on calcineurin signalling, fibre-type distribution and size and dystrophic pathology. We generated an mdx/Sln-null (mdx/SlnKO) mouse colony and hypothesized that the soleus and diaphragm muscles from these mice would display blunted calcineurin signalling, smaller myofibre sizes, an increased proportion of fast fibres and worsened dystrophic pathology compared with mdx mice. Our results show that calcineurin signalling was impaired in mdx/SlnKO mice as indicated by reductions in utrophin, stabilin-2 and calcineurin expression. In addition, mdx/SlnKO muscles contained smaller myofibres, exhibited a slow-to-fast fibre-type switch that corresponded with reduced expression of mitochondrial proteins and displayed a worsened dystrophic pathology compared with mdx muscles. Altogether, our findings demonstrate a critical role for SLN upregulation in dystrophic muscles and suggest that SLN can be viewed as a potential therapeutic target.
Collapse
Affiliation(s)
- Val A Fajardo
- Department of Kinesiology, University of Waterloo, Waterloo, ON N2L 3G1 Canada
| | - Paige J Chambers
- Department of Kinesiology, University of Waterloo, Waterloo, ON N2L 3G1 Canada
| | - Emma S Juracic
- Department of Kinesiology, University of Waterloo, Waterloo, ON N2L 3G1 Canada
| | - Bradley A Rietze
- Department of Kinesiology, University of Waterloo, Waterloo, ON N2L 3G1 Canada
| | - Daniel Gamu
- Department of Kinesiology, University of Waterloo, Waterloo, ON N2L 3G1 Canada
| | | | - Frenk Kwon
- Department of Kinesiology, University of Waterloo, Waterloo, ON N2L 3G1 Canada
| | - Joe Quadrilatero
- Department of Kinesiology, University of Waterloo, Waterloo, ON N2L 3G1 Canada
| | - A Russell Tupling
- Department of Kinesiology, University of Waterloo, Waterloo, ON N2L 3G1 Canada
| |
Collapse
|
14
|
Popov DV, Lysenko EA, Bokov RO, Volodina MA, Kurochkina NS, Makhnovskii PA, Vyssokikh MY, Vinogradova OL. Effect of aerobic training on baseline expression of signaling and respiratory proteins in human skeletal muscle. Physiol Rep 2018; 6:e13868. [PMID: 30198217 PMCID: PMC6129775 DOI: 10.14814/phy2.13868] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 08/24/2018] [Indexed: 12/30/2022] Open
Abstract
Most studies examining the molecular mechanisms underlying adaptation of human skeletal muscles to aerobic exercise focused on the response to acute exercise. Here, we examined the effect of a 2-month aerobic training program on baseline parameters in human muscle. Ten untrained males performed a one-legged knee extension exercise for 1 h with the same relative intensity before and after a 2-month aerobic training program. Biopsy samples were taken from vastus lateralis muscle at rest before and after the 2 month training program (baseline samples). Additionally, biopsy samples were taken from the exercised leg 1 and 4 h after the one-legged continuous knee extension exercise. Aerobic training decreases baseline phosphorylation of FOXO1Ser256 , increases that of CaMKIIThr286 , CREB1Ser133 , increases baseline expression of mitochondrial proteins in respiratory complexes I-V, and some regulators of mitochondrial biogenesis (TFAM, NR4A3, and CRTC2). An increase in the baseline content of these proteins was not associated with a change in baseline expression of their genes. The increase in the baseline content of regulators of mitochondrial biogenesis (TFAM and NR4A3) was associated with a transient increase in transcription after acute exercise. Contrariwise, the increase in the baseline content of respiratory proteins does not seem to be regulated at the transcriptional level; rather, it is associated with other mechanisms. Adaptation of human skeletal muscle to regular aerobic exercise is associated not only with transient molecular responses to exercise, but also with changes in baseline phosphorylation and expression of regulatory proteins.
Collapse
Affiliation(s)
- Daniil V. Popov
- Laboratory of Exercise PhysiologyInstitute of Biomedical Problems of the Russian Academy of SciencesMoscowRussia
- Faculty of Fundamental MedicineM.V. Lomonosov Moscow State UniversityMoscowRussia
| | - Evgeny A. Lysenko
- Laboratory of Exercise PhysiologyInstitute of Biomedical Problems of the Russian Academy of SciencesMoscowRussia
- Faculty of Fundamental MedicineM.V. Lomonosov Moscow State UniversityMoscowRussia
| | - Roman O. Bokov
- Laboratory of Exercise PhysiologyInstitute of Biomedical Problems of the Russian Academy of SciencesMoscowRussia
| | - Maria A. Volodina
- Laboratory of Mitochondrial MedicineResearch Center for ObstetricsGynecology and PerinatologyMinistry of Healthcare of the Russian FederationMoscowRussia
| | - Nadia S. Kurochkina
- Laboratory of Exercise PhysiologyInstitute of Biomedical Problems of the Russian Academy of SciencesMoscowRussia
| | - Pavel A. Makhnovskii
- Laboratory of Exercise PhysiologyInstitute of Biomedical Problems of the Russian Academy of SciencesMoscowRussia
| | - Mikhail Y. Vyssokikh
- Laboratory of Mitochondrial MedicineResearch Center for ObstetricsGynecology and PerinatologyMinistry of Healthcare of the Russian FederationMoscowRussia
| | - Olga L. Vinogradova
- Laboratory of Exercise PhysiologyInstitute of Biomedical Problems of the Russian Academy of SciencesMoscowRussia
- Faculty of Fundamental MedicineM.V. Lomonosov Moscow State UniversityMoscowRussia
| |
Collapse
|
15
|
Tallis J, James RS, Seebacher F. The effects of obesity on skeletal muscle contractile function. ACTA ACUST UNITED AC 2018; 221:221/13/jeb163840. [PMID: 29980597 DOI: 10.1242/jeb.163840] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Obesity can cause a decline in contractile function of skeletal muscle, thereby reducing mobility and promoting obesity-associated health risks. We reviewed the literature to establish the current state-of-knowledge of how obesity affects skeletal muscle contraction and relaxation. At a cellular level, the dominant effects of obesity are disrupted calcium signalling and 5'-adenosine monophosphate-activated protein kinase (AMPK) activity. As a result, there is a shift from slow to fast muscle fibre types. Decreased AMPK activity promotes the class II histone deacetylase (HDAC)-mediated inhibition of the myocyte enhancer factor 2 (MEF2). MEF2 promotes slow fibre type expression, and its activity is stimulated by the calcium-dependent phosphatase calcineurin. Obesity-induced attenuation of calcium signalling via its effects on calcineurin, as well as on adiponectin and actinin affects excitation-contraction coupling and excitation-transcription coupling in the myocyte. These molecular changes affect muscle contractile function and phenotype, and thereby in vivo and in vitro muscle performance. In vivo, obesity can increase the absolute force and power produced by increasing the demand on weight-supporting muscle. However, when normalised to body mass, muscle performance of obese individuals is reduced. Isolated muscle preparations show that obesity often leads to a decrease in force produced per muscle cross-sectional area, and power produced per muscle mass. Obesity and ageing have similar physiological consequences. The synergistic effects of obesity and ageing on muscle function may exacerbate morbidity and mortality. Important future research directions include determining: the relationship between time course of weight gain and changes in muscle function; the relative effects of weight gain and high-fat diet feeding per se; the effects of obesity on muscle function during ageing; and if the effects of obesity on muscle function are reversible.
Collapse
Affiliation(s)
- Jason Tallis
- Center for Sport, Exercise and Life Sciences, Science and Health Building, Coventry University, Priory Street, Coventry CV1 5FB, UK
| | - Rob S James
- Center for Sport, Exercise and Life Sciences, Science and Health Building, Coventry University, Priory Street, Coventry CV1 5FB, UK
| | - Frank Seebacher
- School of Life and Environmental Sciences, Heydon Laurence Building A08, University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
16
|
Koo JH, Kim TH, Park SY, Joo MS, Han CY, Choi CS, Kim SG. Gα13 ablation reprograms myofibers to oxidative phenotype and enhances whole-body metabolism. J Clin Invest 2017; 127:3845-3860. [PMID: 28920922 DOI: 10.1172/jci92067] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 08/02/2017] [Indexed: 12/24/2022] Open
Abstract
Skeletal muscle is a key organ in energy homeostasis owing to its high requirement for nutrients. Heterotrimeric G proteins converge signals from cell-surface receptors to potentiate or blunt responses against environmental changes. Here, we show that muscle-specific ablation of Gα13 in mice promotes reprogramming of myofibers to the oxidative type, with resultant increases in mitochondrial biogenesis and cellular respiration. Mechanistically, Gα13 and its downstream effector RhoA suppressed nuclear factor of activated T cells 1 (NFATc1), a chief regulator of myofiber conversion, by increasing Rho-associated kinase 2-mediated (Rock2-mediated) phosphorylation at Ser243. Ser243 phosphorylation of NFATc1 was reduced after exercise, but was higher in obese animals. Consequently, Gα13 ablation in muscles enhanced whole-body energy metabolism and increased insulin sensitivity, thus affording protection from diet-induced obesity and hepatic steatosis. Our results define Gα13 as a switch regulator of myofiber reprogramming, implying that modulations of Gα13 and its downstream effectors in skeletal muscle are a potential therapeutic approach to treating metabolic diseases.
Collapse
Affiliation(s)
- Ja Hyun Koo
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea
| | - Tae Hyun Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea
| | - Shi-Young Park
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University School of Medicine, Incheon, South Korea
| | - Min Sung Joo
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea
| | - Chang Yeob Han
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea
| | - Cheol Soo Choi
- Korea Mouse Metabolic Phenotyping Center, Lee Gil Ya Cancer and Diabetes Institute, Gachon University School of Medicine, Incheon, South Korea.,Endocrinology, Internal Medicine, Gachon University Gil Medical Center, Incheon, South Korea
| | - Sang Geon Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea
| |
Collapse
|
17
|
Barberio MD, Huffman KM, Giri M, Hoffman EP, Kraus WE, Hubal MJ. Pyruvate Dehydrogenase Phosphatase Regulatory Gene Expression Correlates with Exercise Training Insulin Sensitivity Changes. Med Sci Sports Exerc 2017; 48:2387-2397. [PMID: 27846149 DOI: 10.1249/mss.0000000000001041] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE Whole body insulin sensitivity (Si) typically improves after aerobic exercise training; however, individual responses can be highly variable. The purpose of this study was to use global gene expression to identify skeletal muscle genes that correlate with exercise-induced Si changes. METHODS Longitudinal cohorts from the Studies of Targeted Risk Reduction Intervention through Defined Exercise were used as Discovery (Affymetrix) and Confirmation (Illumina) of vastus lateralis gene expression profiles. Discovery (n = 39; 21 men) and Confirmation (n = 42; 19 men) cohorts were matched for age (52 ± 8 vs 51 ± 10 yr), body mass index (30.4 ± 2.8 vs 29.7 ± 2.8 kg·m), and V˙O2max (30.4 ± 2.8 vs 29.7 ± 2.8 mL·kg·min). Si was determined via intravenous glucose tolerance test pretraining and posttraining. Pearson product-moment correlation coefficients determined relationships between a) baseline and b) training-induced changes in gene expression and %ΔSi after training. RESULTS Expression of 2454 (Discovery) and 1778 genes (Confirmation) at baseline were significantly (P < 0.05) correlated to %ΔSi; 112 genes overlapped. Pathway analyses identified Ca signaling-related transcripts in this 112-gene list. Expression changes of 1384 (Discovery) and 1288 genes (Confirmation) after training were significantly (P < 0.05) correlated to %ΔSi; 33 genes overlapped, representing contractile apparatus of skeletal and smooth muscle genes. Pyruvate dehydrogenase phosphatase regulatory subunit expression at baseline (P = 0.01, r = 0.41) and posttraining (P = 0.01, r = 0.43) were both correlated with %ΔSi. CONCLUSIONS Exercise-induced adaptations in skeletal muscle Si are related to baseline levels of Ca-regulating transcripts, which may prime the muscle for adaptation. Relationships between %ΔSi and pyruvate dehydrogenase phosphatase regulatory, a regulatory subunit of the pyruvate dehydrogenase complex, indicate that the Si response is strongly related to key steps in metabolic regulation.
Collapse
Affiliation(s)
- Matthew D Barberio
- 1Research Center for Genetic Medicine, Children's National Medical Center, Durham, NC; 2Division of Rheumatology, Department of Medicine, Duke University School of Medicine, Durham, NC; 3Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC; 4Department of Exercise and Nutrition Sciences, George Washington University, WASHINGTON, DC; 5Division of Cardiology, Department of Medicine, Duke University School of Medicine, Durham, NC; and 6Department of Integrative Systems Biology, George Washington University, Washington, DC
| | | | | | | | | | | |
Collapse
|
18
|
Cong J, Zhang L, Li J, Wang S, Gao F, Zhou G. Effects of dietary supplementation with carnosine on growth performance, meat quality, antioxidant capacity and muscle fiber characteristics in broiler chickens. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2017; 97:3733-3741. [PMID: 28120335 DOI: 10.1002/jsfa.8236] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 01/09/2017] [Accepted: 01/20/2017] [Indexed: 06/06/2023]
Abstract
BACKGROUND The effects of dietary carnosine were evaluated on the growth performance, meat quality, antioxidant capacity and muscle fiber characteristics in thigh muscle of 256 one-day-old male broilers assigned to four diets - basal diets supplemented with 0, 100, 200 or 400 mg kg-1 carnosine respectively - during a 42 day experiment. RESULTS Carnosine concentration and carnosine synthase expression in thigh muscle were linearly increased (P < 0.05) and the feed/gain ratio was decreased (P < 0.05) in the starter period by carnosine addition. Dietary supplementation with carnosine resulted in linear increases in pH45min , redness and cohesiveness and decreases in drip loss, cooking loss, shear force and hardness (P < 0.05). Carnosine addition elevated the activities of antioxidant enzymes and reduced contents of malondialdehyde and carbonyl compounds (P < 0.05). Dietary carnosine linearly decreased diameters and increased densities of muscle fibers (P < 0.01). The ratios of myosin heavy chain (MyHC) I and IIa were increased while that of MyHC IIb was decreased (P < 0.01). The mRNA expressions of genes related to fiber type transformation were linearly up-regulated (P < 0.05). CONCLUSION These findings indicated that carnosine supplementation was beneficial to improve the growth performance, meat quality, antioxidant capacity and muscle fiber characteristics of broilers. © 2017 Society of Chemical Industry.
Collapse
Affiliation(s)
- Jiahui Cong
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing, China
| | - Lin Zhang
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing, China
| | - Jiaolong Li
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing, China
| | - Shuhao Wang
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing, China
| | - Feng Gao
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing, China
| | - Guanghong Zhou
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
19
|
Mansueto G, Armani A, Viscomi C, D'Orsi L, De Cegli R, Polishchuk EV, Lamperti C, Di Meo I, Romanello V, Marchet S, Saha PK, Zong H, Blaauw B, Solagna F, Tezze C, Grumati P, Bonaldo P, Pessin JE, Zeviani M, Sandri M, Ballabio A. Transcription Factor EB Controls Metabolic Flexibility during Exercise. Cell Metab 2017; 25:182-196. [PMID: 28011087 PMCID: PMC5241227 DOI: 10.1016/j.cmet.2016.11.003] [Citation(s) in RCA: 243] [Impact Index Per Article: 30.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 06/24/2016] [Accepted: 11/05/2016] [Indexed: 11/28/2022]
Abstract
The transcription factor EB (TFEB) is an essential component of lysosomal biogenesis and autophagy for the adaptive response to food deprivation. To address the physiological function of TFEB in skeletal muscle, we have used muscle-specific gain- and loss-of-function approaches. Here, we show that TFEB controls metabolic flexibility in muscle during exercise and that this action is independent of peroxisome proliferator-activated receptor-γ coactivator1α (PGC1α). Indeed, TFEB translocates into the myonuclei during physical activity and regulates glucose uptake and glycogen content by controlling expression of glucose transporters, glycolytic enzymes, and pathways related to glucose homeostasis. In addition, TFEB induces the expression of genes involved in mitochondrial biogenesis, fatty acid oxidation, and oxidative phosphorylation. This coordinated action optimizes mitochondrial substrate utilization, thus enhancing ATP production and exercise capacity. These findings identify TFEB as a critical mediator of the beneficial effects of exercise on metabolism.
Collapse
Affiliation(s)
- Gelsomina Mansueto
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli, Naples, Italy
| | - Andrea Armani
- Department of Biomedical Science, University of Padova, Padova 35121, Italy
| | - Carlo Viscomi
- MRC Mitochondrial Biology Unit, Cambridge CB2 0XY, UK; Fondazione IRCCS Istituto Neurologico "C. Besta," 20133 Milan, Italy
| | - Luca D'Orsi
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli, Naples, Italy
| | - Rossella De Cegli
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli, Naples, Italy
| | - Elena V Polishchuk
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli, Naples, Italy
| | - Costanza Lamperti
- Fondazione IRCCS Istituto Neurologico "C. Besta," 20133 Milan, Italy
| | - Ivano Di Meo
- Fondazione IRCCS Istituto Neurologico "C. Besta," 20133 Milan, Italy
| | - Vanina Romanello
- Department of Biomedical Science, University of Padova, Padova 35121, Italy; Venetian Institute of Molecular Medicine, Padova 35129, Italy
| | - Silvia Marchet
- Fondazione IRCCS Istituto Neurologico "C. Besta," 20133 Milan, Italy
| | - Pradip K Saha
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Haihong Zong
- Hepatobiliary, Pancreatic, and Intestinal Research Institute, North Sichuan Medical College, Nanchong, Sichuan 637000, China; Departments of Medicine and Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Bert Blaauw
- Department of Biomedical Science, University of Padova, Padova 35121, Italy
| | - Francesca Solagna
- Department of Biomedical Science, University of Padova, Padova 35121, Italy
| | - Caterina Tezze
- Department of Biomedical Science, University of Padova, Padova 35121, Italy
| | - Paolo Grumati
- Department of Molecular Medicine, University of Padova, Padova 35121, Italy
| | - Paolo Bonaldo
- Department of Molecular Medicine, University of Padova, Padova 35121, Italy
| | - Jeffrey E Pessin
- Departments of Medicine and Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Massimo Zeviani
- MRC Mitochondrial Biology Unit, Cambridge CB2 0XY, UK; Fondazione IRCCS Istituto Neurologico "C. Besta," 20133 Milan, Italy
| | - Marco Sandri
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli, Naples, Italy; Department of Biomedical Science, University of Padova, Padova 35121, Italy; Venetian Institute of Molecular Medicine, Padova 35129, Italy.
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine (TIGEM), Via Campi Flegrei 34, 80078 Pozzuoli, Naples, Italy; Medical Genetics, Department of Pediatrics, Federico II University, Via Pansini 5, 80131 Naples, Italy; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030, USA.
| |
Collapse
|
20
|
Pant M, Bal NC, Periasamy M. Sarcolipin: A Key Thermogenic and Metabolic Regulator in Skeletal Muscle. Trends Endocrinol Metab 2016; 27:881-892. [PMID: 27637585 PMCID: PMC5424604 DOI: 10.1016/j.tem.2016.08.006] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 08/19/2016] [Accepted: 08/22/2016] [Indexed: 12/14/2022]
Abstract
Skeletal muscle constitutes ∼40% of body mass and has the capacity to play a major role as thermogenic, metabolic, and endocrine organ. In addition to shivering, muscle also contributes to nonshivering thermogenesis via futile sarcoplasmic/endoplasmic reticulum Ca2+ ATPase (SERCA) activity. Sarcolipin (SLN), a regulator of SERCA activity in muscle, plays an important role in regulating muscle thermogenesis and metabolism. Uncoupling of SERCA by SLN increases ATP hydrolysis and heat production, and contributes to temperature homeostasis. SLN also affects whole-body metabolism and weight gain in mice, and is upregulated in various muscle diseases including muscular dystrophy, suggesting a role for SLN during increased metabolic demand. In this review we also highlight the physiological roles of skeletal muscle beyond contraction.
Collapse
Affiliation(s)
- Meghna Pant
- Department of Physiology and Cell Biology, Ohio State University, Columbus, OH, USA
| | - Naresh C Bal
- Sanford Burnham Medical Research Institute at Lake Nona, Orlando, FL, USA; School of Biotechnology, KIIT University, Bhubaneswar, Odisha, India
| | - Muthu Periasamy
- Department of Physiology and Cell Biology, Ohio State University, Columbus, OH, USA; Sanford Burnham Medical Research Institute at Lake Nona, Orlando, FL, USA.
| |
Collapse
|
21
|
Matsa E, Burridge PW, Yu KH, Ahrens JH, Termglinchan V, Wu H, Liu C, Shukla P, Sayed N, Churko JM, Shao N, Woo NA, Chao AS, Gold JD, Karakikes I, Snyder MP, Wu JC. Transcriptome Profiling of Patient-Specific Human iPSC-Cardiomyocytes Predicts Individual Drug Safety and Efficacy Responses In Vitro. Cell Stem Cell 2016; 19:311-25. [PMID: 27545504 PMCID: PMC5087997 DOI: 10.1016/j.stem.2016.07.006] [Citation(s) in RCA: 123] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 04/20/2016] [Accepted: 07/15/2016] [Indexed: 01/24/2023]
Abstract
Understanding individual susceptibility to drug-induced cardiotoxicity is key to improving patient safety and preventing drug attrition. Human induced pluripotent stem cells (hiPSCs) enable the study of pharmacological and toxicological responses in patient-specific cardiomyocytes (CMs) and may serve as preclinical platforms for precision medicine. Transcriptome profiling in hiPSC-CMs from seven individuals lacking known cardiovascular disease-associated mutations and in three isogenic human heart tissue and hiPSC-CM pairs showed greater inter-patient variation than intra-patient variation, verifying that reprogramming and differentiation preserve patient-specific gene expression, particularly in metabolic and stress-response genes. Transcriptome-based toxicology analysis predicted and risk-stratified patient-specific susceptibility to cardiotoxicity, and functional assays in hiPSC-CMs using tacrolimus and rosiglitazone, drugs targeting pathways predicted to produce cardiotoxicity, validated inter-patient differential responses. CRISPR/Cas9-mediated pathway correction prevented drug-induced cardiotoxicity. Our data suggest that hiPSC-CMs can be used in vitro to predict and validate patient-specific drug safety and efficacy, potentially enabling future clinical approaches to precision medicine.
Collapse
Affiliation(s)
- Elena Matsa
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Departments of Medicine and Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Paul W Burridge
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Departments of Medicine and Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Kun-Hsing Yu
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA; Biomedical Informatics Training Program, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - John H Ahrens
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Vittavat Termglinchan
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Departments of Medicine and Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Haodi Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Departments of Medicine and Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Chun Liu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Departments of Medicine and Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Praveen Shukla
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Departments of Medicine and Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nazish Sayed
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Departments of Medicine and Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jared M Churko
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Departments of Medicine and Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ningyi Shao
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Departments of Medicine and Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nicole A Woo
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Alexander S Chao
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Joseph D Gold
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ioannis Karakikes
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Departments of Medicine and Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael P Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Departments of Medicine and Radiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute of Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
22
|
Wang X, Wang G, Shi Y, Sun L, Gorczynski R, Li YJ, Xu Z, Spaner DE. PPAR-delta promotes survival of breast cancer cells in harsh metabolic conditions. Oncogenesis 2016; 5:e232. [PMID: 27270614 PMCID: PMC4945742 DOI: 10.1038/oncsis.2016.41] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 05/03/2016] [Indexed: 12/27/2022] Open
Abstract
Expression of the nuclear receptor peroxisome proliferator activated receptor delta (PPARδ) in breast cancer cells is negatively associated with patient survival, but the underlying mechanisms are not clear. High PPARδ protein levels in rat breast adenocarcinomas were found to be associated with increased growth in soft agar and mice. Transgenic expression of PPARδ increased the ability of human breast cancer cell lines to migrate in vitro and form lung metastases in mice. PPARδ also conferred the ability to grow in exhausted tissue culture media and survive in low-glucose and other endoplasmic reticulum stress conditions such as hypoxia. Upregulation of PPARδ by glucocorticoids or synthetic agonists also protected human breast cancer cells from low glucose. Survival in low glucose was related to increased antioxidant defenses mediated in part by catalase and also to late AKT phosphorylation, which is associated with the prolonged glucose-deprivation response. Synthetic antagonists reversed the survival benefits conferred by PPARδ in vitro. These findings suggest that PPARδ conditions breast cancer cells to survive in harsh microenvironmental conditions by reducing oxidative stress and enhancing survival signaling responses. Drugs that target PPARδ may have a role in the treatment of breast cancer.
Collapse
Affiliation(s)
- X Wang
- Biology Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - G Wang
- Biology Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Y Shi
- Biology Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - L Sun
- Biology Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - R Gorczynski
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada.,Transplant Research Division, Toronto General Hospital, Toronto, Ontario, Canada
| | - Y-J Li
- Biology Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Department of Anatomy, Norman Bethune College of Medicine, Jilin University, Changchun, China
| | - Z Xu
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - D E Spaner
- Biology Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada.,Department of Immunology, University of Toronto, Toronto, Ontario, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.,Sunnybrook Odette Cancer Center, Toronto, Ontario, Canada.,Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
23
|
Peiris H, Duffield MD, Fadista J, Jessup CF, Kashmir V, Genders AJ, McGee SL, Martin AM, Saiedi M, Morton N, Carter R, Cousin MA, Kokotos AC, Oskolkov N, Volkov P, Hough TA, Fisher EMC, Tybulewicz VLJ, Busciglio J, Coskun PE, Becker A, Belichenko PV, Mobley WC, Ryan MT, Chan JY, Laybutt DR, Coates PT, Yang S, Ling C, Groop L, Pritchard MA, Keating DJ. A Syntenic Cross Species Aneuploidy Genetic Screen Links RCAN1 Expression to β-Cell Mitochondrial Dysfunction in Type 2 Diabetes. PLoS Genet 2016; 12:e1006033. [PMID: 27195491 PMCID: PMC4873152 DOI: 10.1371/journal.pgen.1006033] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 04/18/2016] [Indexed: 12/20/2022] Open
Abstract
Type 2 diabetes (T2D) is a complex metabolic disease associated with obesity, insulin resistance and hypoinsulinemia due to pancreatic β-cell dysfunction. Reduced mitochondrial function is thought to be central to β-cell dysfunction. Mitochondrial dysfunction and reduced insulin secretion are also observed in β-cells of humans with the most common human genetic disorder, Down syndrome (DS, Trisomy 21). To identify regions of chromosome 21 that may be associated with perturbed glucose homeostasis we profiled the glycaemic status of different DS mouse models. The Ts65Dn and Dp16 DS mouse lines were hyperglycemic, while Tc1 and Ts1Rhr mice were not, providing us with a region of chromosome 21 containing genes that cause hyperglycemia. We then examined whether any of these genes were upregulated in a set of ~5,000 gene expression changes we had identified in a large gene expression analysis of human T2D β-cells. This approach produced a single gene, RCAN1, as a candidate gene linking hyperglycemia and functional changes in T2D β-cells. Further investigations demonstrated that RCAN1 methylation is reduced in human T2D islets at multiple sites, correlating with increased expression. RCAN1 protein expression was also increased in db/db mouse islets and in human and mouse islets exposed to high glucose. Mice overexpressing RCAN1 had reduced in vivo glucose-stimulated insulin secretion and their β-cells displayed mitochondrial dysfunction including hyperpolarised membrane potential, reduced oxidative phosphorylation and low ATP production. This lack of β-cell ATP had functional consequences by negatively affecting both glucose-stimulated membrane depolarisation and ATP-dependent insulin granule exocytosis. Thus, from amongst the myriad of gene expression changes occurring in T2D β-cells where we had little knowledge of which changes cause β-cell dysfunction, we applied a trisomy 21 screening approach which linked RCAN1 to β-cell mitochondrial dysfunction in T2D.
Collapse
Affiliation(s)
- Heshan Peiris
- Department of Human Physiology and Centre for Neuroscience, Flinders University, Adelaide, South Australia, Australia
| | - Michael D. Duffield
- Department of Human Physiology and Centre for Neuroscience, Flinders University, Adelaide, South Australia, Australia
| | | | - Claire F. Jessup
- Islet Biology Laboratory, Department of Anatomy and Histology and Centre for Neuroscience, Flinders University, Adelaide, South Australia, Australia
- Discipline of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - Vinder Kashmir
- Department of Human Physiology and Centre for Neuroscience, Flinders University, Adelaide, South Australia, Australia
| | - Amanda J. Genders
- Metabolic Remodelling Laboratory, Metabolic Research Unit, School of Medicine, Deakin University, Geelong, Australia
| | - Sean L. McGee
- Metabolic Remodelling Laboratory, Metabolic Research Unit, School of Medicine, Deakin University, Geelong, Australia
- Metabolism and Inflammation Program, Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Alyce M. Martin
- Department of Human Physiology and Centre for Neuroscience, Flinders University, Adelaide, South Australia, Australia
| | - Madiha Saiedi
- Department of Human Physiology and Centre for Neuroscience, Flinders University, Adelaide, South Australia, Australia
| | - Nicholas Morton
- Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Roderick Carter
- Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Michael A. Cousin
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
| | - Alexandros C. Kokotos
- Centre for Integrative Physiology, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Petr Volkov
- Lund University Diabetes Centre, Malmö, Sweden
| | - Tertius A. Hough
- Mary Lyon Centre Pathology, MRC Harwell, Harwell Oxford Science Park, Oxford, United Kingdom
| | - Elizabeth M. C. Fisher
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, United Kingdom
| | - Victor L. J. Tybulewicz
- Francis Crick Institute, Mill Hill, London, United Kingdom
- Department of Medicine, Imperial College London, London, United Kingdom
| | - Jorge Busciglio
- Department of Neurobiology and Behaviour, University of California, Irvine, Irvine, California, United States of America
| | - Pinar E. Coskun
- Department of Neurobiology and Behaviour, University of California, Irvine, Irvine, California, United States of America
| | - Ann Becker
- Department of Neurosciences School of Medicine, University of California, San Diego, San Diego, California, United States of America
| | - Pavel V. Belichenko
- Department of Neurosciences School of Medicine, University of California, San Diego, San Diego, California, United States of America
| | - William C. Mobley
- Department of Neurosciences School of Medicine, University of California, San Diego, San Diego, California, United States of America
| | - Michael T. Ryan
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, Australia
| | - Jeng Yie Chan
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Darlinghurst, Sydney, New South Wales, Australia
| | - D. Ross Laybutt
- Diabetes and Metabolism Division, Garvan Institute of Medical Research, Darlinghurst, Sydney, New South Wales, Australia
| | - P. Toby Coates
- Clinical and Experimental Transplantation Group, Royal Adelaide Hospital, North Terrace, Adelaide, South Australia, Australia
| | - Sijun Yang
- Animal Experiment Center, Animal Biosafety Level-III Laboratory, Wuhan University, Wuhan, China
| | | | - Leif Groop
- Lund University Diabetes Centre, Malmö, Sweden
| | - Melanie A. Pritchard
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, Victoria, Australia
| | - Damien J. Keating
- Department of Human Physiology and Centre for Neuroscience, Flinders University, Adelaide, South Australia, Australia
- South Australian Health and Medical Research Institute, Adelaide, Australia
| |
Collapse
|
24
|
Xia L, Cheung KK, Yeung SS, Yeung EW. The involvement of transient receptor potential canonical type 1 in skeletal muscle regrowth after unloading-induced atrophy. J Physiol 2016; 594:3111-26. [PMID: 26752511 DOI: 10.1113/jp271705] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 01/06/2016] [Indexed: 02/05/2023] Open
Abstract
KEY POINTS Decreased mechanical loading results in skeletal muscle atrophy. The transient receptor potential canonical type 1 (TRPC1) protein is implicated in this process. Investigation of the regulation of TRPC1 in vivo has rarely been reported. In the present study, we employ the mouse hindlimb unloading and reloading model to examine the involvement of TRPC1 in the regulation of muscle atrophy and regrowth, respectively. We establish the physiological relevance of the concept that manipulation of TRPC1 could interfere with muscle regrowth processes following an atrophy-inducing event. Specifically, we show that suppressing TRPC1 expression during reloading impairs the recovery of the muscle mass and slow myosin heavy chain profile. Calcineurin appears to be part of the signalling pathway involved in the regulation of TRPC1 expression during muscle regrowth. These results provide new insights concerning the function of TRPC1. Interventions targeting TRPC1 or its downstream or upstream pathways could be useful for promoting muscle regeneration. ABSTRACT Decreased mechanical loading, such as bed rest, results in skeletal muscle atrophy. The functional consequences of decreased mechanical loading include a loss of muscle mass and decreased muscle strength, particularly in anti-gravity muscles. The purpose of this investigation was to clarify the regulatory role of the transient receptor potential canonical type 1 (TRPC1) protein during muscle atrophy and regrowth. Mice were subjected to 14 days of hindlimb unloading followed by 3, 7, 14 and 28 days of reloading. Weight-bearing mice were used as controls. TRPC1 expression in the soleus muscle decreased significantly and persisted at 7 days of reloading. Small interfering RNA (siRNA)-mediated downregulation of TRPC1 in weight-bearing soleus muscles resulted in a reduced muscle mass and a reduced myofibre cross-sectional area (CSA). Microinjecting siRNA into soleus muscles in vivo after 7 days of reloading provided further evidence for the role of TRPC1 in regulating muscle regrowth. Myofibre CSA, as well as the percentage of slow myosin heavy chain-positive myofibres, was significantly lower in TRPC1-siRNA-expressing muscles than in control muscles after 14 days of reloading. Additionally, inhibition of calcineurin (CaN) activity downregulated TRPC1 expression in both weight-bearing and reloaded muscles, suggesting a possible association between CaN and TRPC1 during skeletal muscle regrowth.
Collapse
Affiliation(s)
- Lu Xia
- Muscle Physiology Laboratory, Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong.,Rehabilitation Key Laboratory of Sichuan Province, Department of Rehabilitation, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kwok-Kuen Cheung
- Muscle Physiology Laboratory, Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Simon S Yeung
- Muscle Physiology Laboratory, Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| | - Ella W Yeung
- Muscle Physiology Laboratory, Department of Rehabilitation Sciences, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong
| |
Collapse
|
25
|
Wang K, Liu D, Hernandez-Sanchez J, Chen J, Liu C, Wu Z, Fang M, Li N. Genome Wide Association Analysis Reveals New Production Trait Genes in a Male Duroc Population. PLoS One 2015; 10:e0139207. [PMID: 26418247 PMCID: PMC4587933 DOI: 10.1371/journal.pone.0139207] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 09/10/2015] [Indexed: 01/07/2023] Open
Abstract
In this study, 796 male Duroc pigs were used to identify genomic regions controlling growth traits. Three production traits were studied: food conversion ratio, days to 100 KG, and average daily gain, using a panel of 39,436 single nucleotide polymorphisms. In total, we detected 11 genome-wide and 162 chromosome-wide single nucleotide polymorphism trait associations. The Gene ontology analysis identified 14 candidate genes close to significant single nucleotide polymorphisms, with growth-related functions: six for days to 100 KG (WT1, FBXO3, DOCK7, PPP3CA, AGPAT9, and NKX6-1), seven for food conversion ratio (MAP2, TBX15, IVL, ARL15, CPS1, VWC2L, and VAV3), and one for average daily gain (COL27A1). Gene ontology analysis indicated that most of the candidate genes are involved in muscle, fat, bone or nervous system development, nutrient absorption, and metabolism, which are all either directly or indirectly related to growth traits in pigs. Additionally, we found four haplotype blocks composed of suggestive single nucleotide polymorphisms located in the growth trait-related quantitative trait loci and further narrowed down the ranges, the largest of which decreased by ~60 Mb. Hence, our results could be used to improve pig production traits by increasing the frequency of favorable alleles via artificial selection.
Collapse
Affiliation(s)
- Kejun Wang
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, People’s Republic of China
| | - Dewu Liu
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, People’s Republic of China
| | - Jules Hernandez-Sanchez
- Research Methods Group| Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), 60 Musk Ave/cnr. Blamey St, Kelvin Grove, QLD 4059, Australia
| | - Jie Chen
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, People’s Republic of China
| | - Chengkun Liu
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, People’s Republic of China
| | - Zhenfang Wu
- Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, 510642, People’s Republic of China
| | - Meiying Fang
- Department of Animal Genetics and Breeding, National Engineering Laboratory for Animal Breeding, MOA Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, People’s Republic of China
- * E-mail:
| | - Ning Li
- State Key Laboratory for Agrobiotechnology, China Agricultural University, Beijing, 100094, People’s Republic of China
| |
Collapse
|
26
|
Murphy ACH, Young PW. The actinin family of actin cross-linking proteins - a genetic perspective. Cell Biosci 2015; 5:49. [PMID: 26312134 PMCID: PMC4550062 DOI: 10.1186/s13578-015-0029-7] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 06/22/2015] [Accepted: 06/23/2015] [Indexed: 01/08/2023] Open
Abstract
Actinins are one of the major actin cross-linking proteins found in virtually all cell types and are the ancestral proteins of a larger family that includes spectrin, dystrophin and utrophin. Invertebrates have a single actinin-encoding ACTN gene, while mammals have four. Mutations in all four human genes have now been linked to heritable diseases or traits. ACTN1 mutations cause macrothrombocytopenia, a platelet disorder characterized by excessive bleeding. ACTN2 mutations have been linked to a range of cardiomyopathies, and ACTN4 mutations cause a kidney condition called focal segmental glomerulosclerosis. Intriguingly, approximately 16 % of people worldwide are homozygous for a nonsense mutation in ACTN3 that abolishes actinin-3 protein expression. This ACTN3 null allele has undergone recent positive selection in specific human populations, which may be linked to improved endurance and adaptation to colder climates. In this review we discuss the human genetics of the ACTN gene family, as well as ACTN gene knockout studies in several model organisms. Observations from both of these areas provide insights into the evolution and cellular functions of actinins.
Collapse
Affiliation(s)
- Anita C H Murphy
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Paul W Young
- School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| |
Collapse
|
27
|
Shu J, Li H, Shan Y, Xu W, Chen W, Song C, Song W. Expression profile of IGF-I-calcineurin-NFATc3-dependent pathway genes in skeletal muscle during early development between duck breeds differing in growth rates. Dev Genes Evol 2015; 225:139-48. [PMID: 25963597 DOI: 10.1007/s00427-015-0501-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 04/30/2015] [Indexed: 11/25/2022]
Abstract
The insulin-like growth factor I (IGF-I)-calcineurin (CaN)-NFATc signaling pathways have been implicated in the regulation of myocyte hypertrophy and fiber-type specificity. In the present study, the expression of the CnAα, NFATc3, and IGF-I genes was quantified by RT-PCR for the first time in the breast muscle (BM) and leg muscle (LM) on days 13, 17, 21, 25, and 27 of embryonic development, as well as at 7 days posthatching (PH), in Gaoyou and Jinding ducks, which differ in their muscle growth rates. Consistent expression patterns of CnAα, NFATc3, and IGF-I were found in the same anatomical location at different development stages in both duck breeds, showing significant differences in an age-specific fashion. However, the three genes were differentially expressed in the two different anatomical locations (BM and LM). CnAα, NFATc3, and IGF-I messenger RNA (mRNA) could be detected as early as embryonic day 13 (ED13), and the highest level appeared at this stage in both BM and LM. Significant positive relationships were observed in the expression of the studied genes in the BM and LM of both duck breeds. Also, the expression of these three genes showed a positive relationship with the percentage of type IIb fibers and a negative relationship with the percentage of type I fibers and type IIa fibers. Our data indicate differential expression and coordinated developmental regulation of the selected genes involved in the IGF-I-calcineurin-NFATc3 pathway in duck skeletal muscle during embryonic and early PH growth and development; these data also indicate that this signaling pathway might play a role in the regulation of myofiber type transition.
Collapse
Affiliation(s)
- Jingting Shu
- Key Laboratory for Poultry Genetics and Breeding of Jiangsu Province, Institute of Poultry Science, Chinese Academy of Agricultural Science, Yangzhou, 225125, China
| | | | | | | | | | | | | |
Collapse
|
28
|
Altered Ca2+ kinetics associated with α-actinin-3 deficiency may explain positive selection for ACTN3 null allele in human evolution. PLoS Genet 2015; 11:e1004862. [PMID: 25590636 PMCID: PMC4295894 DOI: 10.1371/journal.pgen.1004862] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 10/29/2014] [Indexed: 11/28/2022] Open
Abstract
Over 1.5 billion people lack the skeletal muscle fast-twitch fibre protein α-actinin-3 due to homozygosity for a common null polymorphism (R577X) in the ACTN3 gene. α-Actinin-3 deficiency is detrimental to sprint performance in elite athletes and beneficial to endurance activities. In the human genome, it is very difficult to find single-gene loss-of-function variants that bear signatures of positive selection, yet intriguingly, the ACTN3 null variant has undergone strong positive selection during recent evolution, appearing to provide a survival advantage where food resources are scarce and climate is cold. We have previously demonstrated that α-actinin-3 deficiency in the Actn3 KO mouse results in a shift in fast-twitch fibres towards oxidative metabolism, which would be more “energy efficient” in famine, and beneficial to endurance performance. Prolonged exposure to cold can also induce changes in skeletal muscle similar to those observed with endurance training, and changes in Ca2+ handling by the sarcoplasmic reticulum (SR) are a key factor underlying these adaptations. On this basis, we explored the effects of α-actinin-3 deficiency on Ca2+ kinetics in single flexor digitorum brevis muscle fibres from Actn3 KO mice, using the Ca2+-sensitive dye fura-2. Compared to wild-type, fibres of Actn3 KO mice showed: (i) an increased rate of decay of the twitch transient; (ii) a fourfold increase in the rate of SR Ca2+ leak; (iii) a threefold increase in the rate of SR Ca2+ pumping; and (iv) enhanced maintenance of tetanic Ca2+ during fatigue. The SR Ca2+ pump, SERCA1, and the Ca2+-binding proteins, calsequestrin and sarcalumenin, showed markedly increased expression in muscles of KO mice. Together, these changes in Ca2+ handling in the absence of α-actinin-3 are consistent with cold acclimatisation and thermogenesis, and offer an additional explanation for the positive selection of the ACTN3 577X null allele in populations living in cold environments during recent evolution. α-Actinin-3 is a protein found inside the muscles of most people around the world. It is encoded by a gene called ACTN3, popularly known as “the gene for speed.” In 1.5 billion people, a certain variation in the genetic sequence of their ACTN3 gene causes their muscles to produce no α-actinin-3 protein at all. These people have no muscle disease; however, in elite athletes, a lack of α-actinin-3 seems to be beneficial for endurance activities and detrimental to sprinting activities. Intriguingly, α-actinin-3 deficiency varies in frequency across the globe, being most common in European and Asian populations and least common in African populations. During recent human evolution, there appears to have been strong positive selection for α-actinin-3 deficiency in places where food resources are relatively scarce and climate is cold. We have previously demonstrated that α-actinin-3 deficiency in the Actn3 knockout (KO) mouse causes a shift towards more “energy efficient” forms of muscle metabolism which would enhance survival in times of famine, and benefit endurance performance. Our present study, using single muscle fibres from Actn3 KO mice, demonstrates changes in calcium handling that would adapt muscles to cold environments and provide a survival advantage in cold climates.
Collapse
|
29
|
Eilers W, Jaspers RT, de Haan A, Ferrié C, Valdivieso P, Flück M. CaMKII content affects contractile, but not mitochondrial, characteristics in regenerating skeletal muscle. BMC PHYSIOLOGY 2014; 14:7. [PMID: 25515219 PMCID: PMC4277655 DOI: 10.1186/s12899-014-0007-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 10/23/2014] [Indexed: 11/21/2022]
Abstract
Background The multi-meric calcium/calmodulin-dependent protein kinase II (CaMKII) is the main CaMK in skeletal muscle and its expression increases with endurance training. CaMK family members are implicated in contraction-induced regulation of calcium handling, fast myosin type IIA expression and mitochondrial biogenesis. The objective of this study was to investigate the role of an increased CaMKII content for the expression of the contractile and mitochondrial phenotype in vivo. Towards this end we attempted to co-express alpha- and beta-CaMKII isoforms in skeletal muscle and characterised the effect on the contractile and mitochondrial phenotype. Results Fast-twitch muscle m. gastrocnemius (GM) and slow-twitch muscle m. soleus (SOL) of the right leg of 3-month old rats were transfected via electro-transfer of injected expression plasmids for native α/β CaMKII. Effects were identified from the comparison to control-transfected muscles of the contralateral leg and non-transfected muscles. α/β CaMKII content in muscle fibres was 4-5-fold increased 7 days after transfection. The transfection rate was more pronounced in SOL than GM muscle (i.e. 12.6 vs. 3.5%). The overexpressed α/β CaMKII was functional as shown through increased threonine 287 phosphorylation of β-CaMKII after isometric exercise and down-regulated transcripts COXI, COXIV, SDHB after high-intensity exercise in situ. α/β CaMKII overexpression under normal cage activity accelerated excitation-contraction coupling and relaxation in SOL muscle in association with increased SERCA2, ANXV and fast myosin type IIA/X content but did not affect mitochondrial protein content. These effects were observed on a background of regenerating muscle fibres. Conclusion Elevated CaMKII content promotes a slow-to-fast type fibre shift in regenerating muscle but is not sufficient to stimulate mitochondrial biogenesis in the absence of an endurance stimulus.
Collapse
Affiliation(s)
- Wouter Eilers
- Institute for Biomedical Research into Human Movement and Health, Manchester Metropolitan University, John Dalton Building, Oxford Road, M1 5GD, Manchester, United Kingdom.
| | - Richard T Jaspers
- Laboratory for Myology, MOVE Research Institute Amsterdam, Faculty of Human Movement Sciences, VU University Amsterdam, Van der Boechorststraat 7, 1081 BT, Amsterdam, The Netherlands.
| | - Arnold de Haan
- Institute for Biomedical Research into Human Movement and Health, Manchester Metropolitan University, John Dalton Building, Oxford Road, M1 5GD, Manchester, United Kingdom. .,Laboratory for Myology, MOVE Research Institute Amsterdam, Faculty of Human Movement Sciences, VU University Amsterdam, Van der Boechorststraat 7, 1081 BT, Amsterdam, The Netherlands.
| | - Céline Ferrié
- Laboratory for Muscle Plasticity, Department of Orthopaedics, University of Zurich, Balgrist University Hospital, Forchstrasse 340, 8008, Zurich, Switzerland.
| | - Paola Valdivieso
- Laboratory for Muscle Plasticity, Department of Orthopaedics, University of Zurich, Balgrist University Hospital, Forchstrasse 340, 8008, Zurich, Switzerland.
| | - Martin Flück
- Institute for Biomedical Research into Human Movement and Health, Manchester Metropolitan University, John Dalton Building, Oxford Road, M1 5GD, Manchester, United Kingdom. .,Laboratory for Muscle Plasticity, Department of Orthopaedics, University of Zurich, Balgrist University Hospital, Forchstrasse 340, 8008, Zurich, Switzerland.
| |
Collapse
|
30
|
Gestational diabetes is characterized by reduced mitochondrial protein expression and altered calcium signaling proteins in skeletal muscle. PLoS One 2014; 9:e106872. [PMID: 25216282 PMCID: PMC4162568 DOI: 10.1371/journal.pone.0106872] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 08/04/2014] [Indexed: 12/14/2022] Open
Abstract
The rising prevalence of gestational diabetes mellitus (GDM) affects up to 18% of pregnant women with immediate and long-term metabolic consequences for both mother and infant. Abnormal glucose uptake and lipid oxidation are hallmark features of GDM prompting us to use an exploratory proteomics approach to investigate the cellular mechanisms underlying differences in skeletal muscle metabolism between obese pregnant women with GDM (OGDM) and obese pregnant women with normal glucose tolerance (ONGT). Functional validation was performed in a second cohort of obese OGDM and ONGT pregnant women. Quantitative proteomic analysis in rectus abdominus skeletal muscle tissue collected at delivery revealed reduced protein content of mitochondrial complex I (C-I) subunits (NDUFS3, NDUFV2) and altered content of proteins involved in calcium homeostasis/signaling (calcineurin A, α1-syntrophin, annexin A4) in OGDM (n = 6) vs. ONGT (n = 6). Follow-up analyses showed reduced enzymatic activity of mitochondrial complexes C-I, C-III, and C-IV (−60–75%) in the OGDM (n = 8) compared with ONGT (n = 10) subjects, though no differences were observed for mitochondrial complex protein content. Upstream regulators of mitochondrial biogenesis and oxidative phosphorylation were not different between groups. However, AMPK phosphorylation was dramatically reduced by 75% in the OGDM women. These data suggest that GDM is associated with reduced skeletal muscle oxidative phosphorylation and disordered calcium homeostasis. These relationships deserve further attention as they may represent novel risk factors for development of GDM and may have implications on the effectiveness of physical activity interventions on both treatment strategies for GDM and for prevention of type 2 diabetes postpartum.
Collapse
|
31
|
Dwyer DS, Weeks K, Aamodt EJ. Drug discovery based on genetic and metabolic findings in schizophrenia. Expert Rev Clin Pharmacol 2014; 1:773-89. [PMID: 24410607 DOI: 10.1586/17512433.1.6.773] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Recent progress in the genetics of schizophrenia provides the rationale for re-evaluating causative factors and therapeutic strategies for this disease. Here, we review the major candidate susceptibility genes and relate the aberrant function of these genes to defective regulation of energy metabolism in the schizophrenic brain. Disturbances in energy metabolism potentially lead to neurodevelopmental deficits, impaired function of the mature nervous system and failure to maintain neurites/dendrites and synaptic connections. Current antipsychotic drugs do not specifically address these underlying deficits; therefore, a new generation of more effective medications is urgently needed. Novel targets for future drug discovery are identified in this review. The coordinated application of structure-based drug design, systems biology and research on model organisms may greatly facilitate the search for next-generation antipsychotic drugs.
Collapse
Affiliation(s)
- Donard S Dwyer
- Professor and Director of Basic Research, Departments of Psychiatry and Pharmacology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA.
| | | | | |
Collapse
|
32
|
Dietary stimulators of the PGC-1 superfamily and mitochondrial biosynthesis in skeletal muscle. A mini-review. J Physiol Biochem 2013; 70:271-84. [DOI: 10.1007/s13105-013-0301-4] [Citation(s) in RCA: 260] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 11/21/2013] [Indexed: 11/26/2022]
|
33
|
Holloszy JO. Regulation of mitochondrial biogenesis and GLUT4 expression by exercise. Compr Physiol 2013; 1:921-40. [PMID: 23737207 DOI: 10.1002/cphy.c100052] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Endurance exercise training can induce large increases mitochondria and the GLUT4 isoform of the glucose transporter in skeletal muscle. For a long time after the discovery in the 1960s that exercise results in an increase in muscle mitochondria, there was no progress in elucidation of the mechanisms involved. The reason for this lack of progress was that nothing was known regarding how expression of the genes-encoding mitochondrial proteins is coordinately regulated. This situation changed rapidly after discovery of transcription factors that control transcription of genes-encoding mitochondrial proteins and, most importantly, the discovery of peroxisome proliferator-gamma coactivator-1α (PGC-1α). This transcription coactivator binds to and activates transcription factors that regulate transcription of genes-encoding mitochondrial proteins. Thus, PGC-1α activates and coordinates mitochondrial biogenesis. It is now known that exercise rapidly activates and induces increased expression of PGC-1α. The exercise-generated signals that lead to PGC-1α activation and increased expression are the increases in cytosolic Ca(2+) and decreases in ATP and creatine phosphate (∼P). Ca(2+) mediates its effect by activating CAMKII, while the decrease in ∼P mediates its effect via activation of AMPK. Expression of the GLUT4 isoform of the glucose transporter is regulated in parallel with mitochondrial biogenesis via the same signaling pathways. This review describes what is known regarding the regulation of mitochondrial biogenesis and GLUT4 expression by exercise. A major component of this review deals with the physiological and metabolic consequences of the exercise-induced increase in mitochondria and GLUT4.
Collapse
Affiliation(s)
- John O Holloszy
- Division of Geriatrics and Nutritional Sciences, Washington University School of Medicine, St. Louis, Missouri, USA.
| |
Collapse
|
34
|
Seto JT, Quinlan KGR, Lek M, Zheng XF, Garton F, MacArthur DG, Hogarth MW, Houweling PJ, Gregorevic P, Turner N, Cooney GJ, Yang N, North KN. ACTN3 genotype influences muscle performance through the regulation of calcineurin signaling. J Clin Invest 2013; 123:4255-63. [PMID: 24091322 DOI: 10.1172/jci67691] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Accepted: 07/19/2013] [Indexed: 02/02/2023] Open
Abstract
α-Actinin-3 deficiency occurs in approximately 16% of the global population due to homozygosity for a common nonsense polymorphism in the ACTN3 gene. Loss of α-actinin-3 is associated with reduced power and enhanced endurance capacity in elite athletes and nonathletes due to "slowing" of the metabolic and physiological properties of fast fibers. Here, we have shown that α-actinin-3 deficiency results in increased calcineurin activity in mouse and human skeletal muscle and enhanced adaptive response to endurance training. α-Actinin-2, which is differentially expressed in α-actinin-3-deficient muscle, has higher binding affinity for calsarcin-2, a key inhibitor of calcineurin activation. We have further demonstrated that α-actinin-2 competes with calcineurin for binding to calsarcin-2, resulting in enhanced calcineurin signaling and reprogramming of the metabolic phenotype of fast muscle fibers. Our data provide a mechanistic explanation for the effects of the ACTN3 genotype on skeletal muscle performance in elite athletes and on adaptation to changing physical demands in the general population. In addition, we have demonstrated that the sarcomeric α-actinins play a role in the regulation of calcineurin signaling.
Collapse
|
35
|
Calcineurin: a poorly understood regulator of muscle mass. Int J Biochem Cell Biol 2013; 45:2173-8. [PMID: 23838168 DOI: 10.1016/j.biocel.2013.06.029] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 06/26/2013] [Accepted: 06/28/2013] [Indexed: 01/14/2023]
Abstract
This review will discuss the existing literature that has examined the role of calcineurin (CnA) in the regulation of skeletal muscle mass in conditions associated with hypertrophic growth or atrophy. Muscle mass is determined by the balance between protein synthesis and degradation which is controlled by a number of intracellular signaling pathways, most notably the insulin/IGF/phosphatidylinositol 3-kinase (PI3K)/Akt system. Despite being activated by IGF-1 and having well-described functions in the determination of muscle fiber phenotypes, calcineurin (CnA), a Ca(2+)-activated serine/threonine phosphatase, and its downstream signaling partners have garnered little attention as a regulator of muscle mass. Compared to other signaling pathways, the relatively few studies that have examined the role of CnA in the regulation of muscle size have produced discordant results. The reasons for these differences is not obvious but may be due to the selective nature of the genetic models studied, fluctuations in the endogenous level of CnA activity in various muscles, and the variable use of CnA inhibitors to inhibit CnA signaling. Despite the inconsistent nature of the outcomes, there is sufficient direct and indirect evidence to conclude that CnA plays a role in the regulation of skeletal muscle mass. This article is part of a Directed Issue entitled: Molecular basis of muscle wasting.
Collapse
|
36
|
Majumdar G, Adris P, Bhargava N, Chen H, Raghow R. Pan-histone deacetylase inhibitors regulate signaling pathways involved in proliferative and pro-inflammatory mechanisms in H9c2 cells. BMC Genomics 2012; 13:709. [PMID: 23249388 PMCID: PMC3561284 DOI: 10.1186/1471-2164-13-709] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 12/12/2012] [Indexed: 01/19/2023] Open
Abstract
Background We have shown previously that pan-HDAC inhibitors (HDACIs) m-carboxycinnamic acid bis-hydroxamide (CBHA) and trichostatin A (TSA) attenuated cardiac hypertrophy in BALB/c mice by inducing hyper-acetylation of cardiac chromatin that was accompanied by suppression of pro-inflammatory gene networks. However, it was not feasible to determine the precise contribution of the myocytes- and non-myocytes to HDACI-induced gene expression in the intact heart. Therefore, the current study was undertaken with a primary goal of elucidating temporal changes in the transcriptomes of cardiac myocytes exposed to CBHA and TSA. Results We incubated H9c2 cardiac myocytes in growth medium containing either of the two HDACIs for 6h and 24h and analyzed changes in gene expression using Illumina microarrays. H9c2 cells exposed to TSA for 6h and 24h led to differential expression of 468 and 231 genes, respectively. In contrast, cardiac myocytes incubated with CBHA for 6h and 24h elicited differential expression of 768 and 999 genes, respectively. We analyzed CBHA- and TSA-induced differentially expressed genes by Ingenuity Pathway (IPA), Kyoto Encyclopedia of Genes and Genomes (KEGG) and Core_TF programs and discovered that CBHA and TSA impinged on several common gene networks. Thus, both HDACIs induced a repertoire of signaling kinases (PTEN-PI3K-AKT and MAPK) and transcription factors (Myc, p53, NFkB and HNF4A) representing canonical TGFβ, TNF-α, IFNγ and IL-6 specific networks. An overrepresentation of E2F, AP2, EGR1 and SP1 specific motifs was also found in the promoters of the differentially expressed genes. Apparently, TSA elicited predominantly TGFβ- and TNF-α-intensive gene networks regardless of the duration of treatment. In contrast, CBHA elicited TNF-α and IFNγ specific networks at 6 h, followed by elicitation of IL-6 and IFNγ-centered gene networks at 24h. Conclusions Our data show that both CBHA and TSA induced similar, but not identical, time-dependent, gene networks in H9c2 cardiac myocytes. Initially, both HDACIs impinged on numerous genes associated with adipokine signaling, intracellular metabolism and energetics, and cell cycle. A continued exposure to either CBHA or TSA led to the emergence of a number of apoptosis- and inflammation-specific gene networks that were apparently suppressed by both HDACIs. Based on these data we posit that the anti-inflammatory and anti-proliferative actions of HDACIs are myocyte-intrinsic. These findings advance our understanding of the mechanisms of actions of HDACIs on cardiac myocytes and reveal potential signaling pathways that may be targeted therapeutically.
Collapse
Affiliation(s)
- Gipsy Majumdar
- Department of Veterans Affairs Medical Center, 1030 Jefferson Avenue, Memphis, TN 38104, USA
| | | | | | | | | |
Collapse
|
37
|
Pronounced effects of acute endurance exercise on gene expression in resting and exercising human skeletal muscle. PLoS One 2012; 7:e51066. [PMID: 23226462 PMCID: PMC3511348 DOI: 10.1371/journal.pone.0051066] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Accepted: 10/29/2012] [Indexed: 12/21/2022] Open
Abstract
Regular physical activity positively influences whole body energy metabolism and substrate handling in exercising muscle. While it is recognized that the effects of exercise extend beyond exercising muscle, it is unclear to what extent exercise impacts non-exercising muscles. Here we investigated the effects of an acute endurance exercise bouts on gene expression in exercising and non-exercising human muscle. To that end, 12 male subjects aged 44–56 performed one hour of one-legged cycling at 50% Wmax. Muscle biopsies were taken from the exercising and non-exercising leg before and immediately after exercise and analyzed by microarray. One-legged cycling raised plasma lactate, free fatty acids, cortisol, noradrenalin, and adrenalin levels. Surprisingly, acute endurance exercise not only caused pronounced gene expression changes in exercising muscle but also in non-exercising muscle. In the exercising leg the three most highly induced genes were all part of the NR4A family. Remarkably, many genes induced in non-exercising muscle were PPAR targets or related to PPAR signalling, including PDK4, ANGPTL4 and SLC22A5. Pathway analysis confirmed this finding. In conclusion, our data indicate that acute endurance exercise elicits pronounced changes in gene expression in non-exercising muscle, which are likely mediated by changes in circulating factors such as free fatty acids. The study points to a major influence of exercise beyond the contracting muscle.
Collapse
|
38
|
Wu HT, Chen W, Cheng KC, Ku PM, Yeh CH, Cheng JT. Oleic acid activates peroxisome proliferator-activated receptor δ to compensate insulin resistance in steatotic cells. J Nutr Biochem 2012; 23:1264-70. [PMID: 22209682 DOI: 10.1016/j.jnutbio.2011.07.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2011] [Revised: 07/09/2011] [Accepted: 07/14/2011] [Indexed: 01/22/2023]
|
39
|
Donaldson AV, Maddocks M, Martolini D, Polkey MI, Man WDC. Muscle function in COPD: a complex interplay. Int J Chron Obstruct Pulmon Dis 2012; 7:523-35. [PMID: 22973093 PMCID: PMC3430120 DOI: 10.2147/copd.s28247] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The skeletal muscles play an essential role in life, providing the mechanical basis for respiration and movement. Skeletal muscle dysfunction is prevalent in all stages of chronic obstructive pulmonary disease (COPD), and significantly influences symptoms, functional capacity, health related quality of life, health resource usage and even mortality. Furthermore, in contrast to the lungs, the skeletal muscles are potentially remedial with existing therapy, namely exercise-training. This review summarizes clinical and laboratory observations of the respiratory and peripheral skeletal muscles (in particular the diaphragm and quadriceps), and current understanding of the underlying etiological processes. As further progress is made in the elucidation of the molecular mechanisms of skeletal muscle dysfunction, new pharmacological therapies are likely to emerge to treat this important extra-pulmonary manifestation of COPD.
Collapse
Affiliation(s)
- Anna V Donaldson
- NIHR Respiratory Biomedical, Research Unit, Royal Brompton, and Harefield NHS Foundation, Trust and Imperial College, London
| | | | | | | | | |
Collapse
|
40
|
Chou MT, Lo SH, Cheng KC, Li YX, Chen LJ, Cheng JT. Activation of β-adrenoceptors by dobutamine may induce a higher expression of peroxisome proliferator-activated receptors δ (PPARδ) in neonatal rat cardiomyocytes. ScientificWorldJournal 2012; 2012:248320. [PMID: 22666095 PMCID: PMC3362020 DOI: 10.1100/2012/248320] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2012] [Accepted: 02/28/2012] [Indexed: 11/25/2022] Open
Abstract
Recent evidence showed the role of peroxisome proliferator-activated receptors (PPARs) in cardiac function. Cardiac contraction induced by various agents is critical in restoring the activity of peroxisome proliferator-activated receptors δ (PPARδ) in cardiac myopathy. Because dobutamine is an agent widely used to treat heart failure in emergency setting, this study is aimed to investigate the change of PPARδ in response to dobutamine. Neonatal rat cardiomyocytes were used to examine the effects of dobutamine on PPARδ expression levels and cardiac troponin I (cTnI) phosphorylation via Western blotting analysis. We show that treatment with dobutamine increased PPARδ expression and cTnI phosphorylation in a time- and dose-dependent manner in neonatal rat cardiomyocytes. These increases were blocked by the antagonist of β1-adrenoceptors. Also, the action of dobutamine was related to the increase of calcium ions and diminished by chelating intracellular calcium. Additionally, dobutamine-induced action was reduced by the inhibition of downstream messengers involved in this calcium-related pathway. Moreover, deletion of PPARδ using siRNA generated the reduction of cTnI phosphorylation in cardiomyocytes treated with dobutamine. Thus, we concluded that PPARδ is increased by dobutamine in cardiac cells.
Collapse
Affiliation(s)
- Ming-Ting Chou
- Department of Cardiology and Department of Medical Research, Chi-Mei Medical Center, Yong Kang, Tainan 73101, Taiwan
| | | | | | | | | | | |
Collapse
|
41
|
Kusudo T, Wang Z, Mizuno A, Suzuki M, Yamashita H. TRPV4 deficiency increases skeletal muscle metabolic capacity and resistance against diet-induced obesity. J Appl Physiol (1985) 2012; 112:1223-32. [DOI: 10.1152/japplphysiol.01070.2011] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Transient receptor potential channel V4 (TRPV4) functions as a nonselective cation channel in various cells and plays physiological roles in osmotic and thermal sensation. However, the function of TRPV4 in energy metabolism is unknown. Here, we report that TRPV4 deficiency results in increased muscle oxidative capacity and resistance to diet-induced obesity in mice. Although no difference in body weight was observed between wild-type and Trpv4−/− mice when fed a standard chow diet, obesity phenotypes induced by a high-fat diet were significantly improved in Trpv4−/− mice, without any change in food intake. Quantitative analysis of mRNA revealed the constitutive upregulation of many genes, including those for transcription factors such as peroxisome proliferator-activated receptor α and for metabolic enzymes such as phosphoenolpyruvate carboxykinase. These upregulated genes were especially prominent in oxidative skeletal muscle, in which the activity of Ca2+-dependent phosphatase calcineurin was elevated, suggesting that other Ca2+ channels function in the skeletal muscle of Trpv4−/− mice. Indeed, gene expressions for TRPC3 and TRPC6 increased in the muscles of Trpv4−/− mice compared with those of wild-type mice. The number of oxidative type I fiber also increased in the mutant muscles following myogenin gene induction. These results strongly suggested that inactivation of Trpv4 induces compensatory increases in TRPC3 and TRPC6 production, and elevation of calcineurin activity, affecting energy metabolism through increased expression of genes involved in fuel oxidation in skeletal muscle and thereby contributing to increased energy expenditure and protection from diet-induced obesity in mice.
Collapse
Affiliation(s)
- Tatsuya Kusudo
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai; and
| | - Zhonghua Wang
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai; and
| | - Atsuko Mizuno
- Department of Pharmacology, Division of Molecular Pharmacology, Jichi Medical University, Shimotsuke, Japan
| | - Makoto Suzuki
- Department of Pharmacology, Division of Molecular Pharmacology, Jichi Medical University, Shimotsuke, Japan
| | - Hitoshi Yamashita
- Department of Biomedical Sciences, College of Life and Health Sciences, Chubu University, Kasugai; and
| |
Collapse
|
42
|
Plattner H, Sehring IM, Mohamed IK, Miranda K, De Souza W, Billington R, Genazzani A, Ladenburger EM. Calcium signaling in closely related protozoan groups (Alveolata): non-parasitic ciliates (Paramecium, Tetrahymena) vs. parasitic Apicomplexa (Plasmodium, Toxoplasma). Cell Calcium 2012; 51:351-82. [PMID: 22387010 DOI: 10.1016/j.ceca.2012.01.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Revised: 01/10/2012] [Accepted: 01/12/2012] [Indexed: 12/20/2022]
Abstract
The importance of Ca2+-signaling for many subcellular processes is well established in higher eukaryotes, whereas information about protozoa is restricted. Recent genome analyses have stimulated such work also with Alveolates, such as ciliates (Paramecium, Tetrahymena) and their pathogenic close relatives, the Apicomplexa (Plasmodium, Toxoplasma). Here we compare Ca2+ signaling in the two closely related groups. Acidic Ca2+ stores have been characterized in detail in Apicomplexa, but hardly in ciliates. Two-pore channels engaged in Ca2+-release from acidic stores in higher eukaryotes have not been stingently characterized in either group. Both groups are endowed with plasma membrane- and endoplasmic reticulum-type Ca2+-ATPases (PMCA, SERCA), respectively. Only recently was it possible to identify in Paramecium a number of homologs of ryanodine and inositol 1,3,4-trisphosphate receptors (RyR, IP3R) and to localize them to widely different organelles participating in vesicle trafficking. For Apicomplexa, physiological experiments suggest the presence of related channels although their identity remains elusive. In Paramecium, IP3Rs are constitutively active in the contractile vacuole complex; RyR-related channels in alveolar sacs are activated during exocytosis stimulation, whereas in the parasites the homologous structure (inner membrane complex) may no longer function as a Ca2+ store. Scrutinized comparison of the two closely related protozoan phyla may stimulate further work and elucidate adaptation to parasitic life. See also "Conclusions" section.
Collapse
Affiliation(s)
- H Plattner
- Department of Biology, University of Konstanz, P.O. Box 5560, 78457 Konstanz, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Chen ZC, Yu BC, Chen LJ, Cheng KC, Lin HJ, Cheng JT. Characterization of the mechanisms of the increase in PPARδ expression induced by digoxin in the heart using the H9c2 cell line. Br J Pharmacol 2011; 163:390-8. [PMID: 21232041 DOI: 10.1111/j.1476-5381.2011.01212.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND AND PURPOSE Digoxin has been used as an inotropic agent in heart failure for a long time. Troponin I (TnI) phosphorylation is related to cardiac contractility, and the genes are regulated by peroxisome proliferator-activated receptors (PPARs). Our previous studies indicated that cardiac abnormality related to the depressed expression of PPARδ in the hearts of STZ rats is reversed by digoxin. However, the cellular mechanisms for this effect of digoxin have not been elucidated. The aim of the present study was to investigate possible mechanisms for this effect of digoxin using the H9c2 cell line cultured in high glucose (HG) conditions. METHODS The effects of digoxin on PPARδ expression, intracellular calcium and TnI phosphorylation were investigated in cultured H9c2 cells, maintained in a HG medium, by using Western blot analysis. RESULTS Digoxin increased PPARδ expression in H9c2 cells subjected to HG conditions, and increase the intracellular calcium concentration. This effect of digoxin was blocked by BAPTA-AM at concentrations sufficient to chelate calcium ions. In addition, the calcineurin inhibitor cyclosporine A and KN93, an inhibitor of calcium/calmodulin-dependent protein kinase, inhibited this action. Digoxin also increased TnI phosphorylation and this was inhibited when PPARδ was silenced by the addition of RNAi to the cells. Similar changes were observed on the contraction of H9c2 cells. CONCLUSION The results suggest that digoxin appears, through calcium-triggered signals, to reverse the reduced expression of PPARδ in H9c2 cells caused by HG treatment.
Collapse
Affiliation(s)
- Zhih-Cherng Chen
- Department of Cardiology, Chi-Mei Medical Center, Yong Kang City, Tainan County, Taiwan
| | | | | | | | | | | |
Collapse
|
44
|
Kozubowski L, Thompson JW, Cardenas ME, Moseley MA, Heitman J. Association of calcineurin with the COPI protein Sec28 and the COPII protein Sec13 revealed by quantitative proteomics. PLoS One 2011; 6:e25280. [PMID: 21984910 PMCID: PMC3184950 DOI: 10.1371/journal.pone.0025280] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Accepted: 08/30/2011] [Indexed: 11/29/2022] Open
Abstract
Calcineurin is a calcium-calmodulin-dependent serine/threonine specific protein phosphatase operating in key cellular processes governing responses to extracellular cues. Calcineurin is essential for growth at high temperature and virulence of the human fungal pathogen Cryptococcus neoformans but the underlying mechanism is unknown. We performed a mass spectrometry analysis to identify proteins that associate with the calcineurin A catalytic subunit (Cna1) in C. neoformans cells grown under non-stress and high temperature stress conditions. A novel prioritization strategy for mass spectrometry data from immunoprecipitation experiments identified putative substrates and proteins potentially operating with calcineurin in common pathways. Cna1 co-purified with proteins involved in membrane trafficking including the COPI component Sec28 and the COPII component Sec13. The association of Cna1 with Sec28 and Sec13 was confirmed by co-immunoprecipitation. Cna1 exhibited a dramatic change in subcellular localization during high temperature stress from diffuse cytoplasmic to ER-associated puncta and the mother-bud neck and co-localized with Sec28 and Sec13.
Collapse
Affiliation(s)
- Lukasz Kozubowski
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, United States of America.
| | | | | | | | | |
Collapse
|
45
|
Abstract
Mammalian skeletal muscle comprises different fiber types, whose identity is first established during embryonic development by intrinsic myogenic control mechanisms and is later modulated by neural and hormonal factors. The relative proportion of the different fiber types varies strikingly between species, and in humans shows significant variability between individuals. Myosin heavy chain isoforms, whose complete inventory and expression pattern are now available, provide a useful marker for fiber types, both for the four major forms present in trunk and limb muscles and the minor forms present in head and neck muscles. However, muscle fiber diversity involves all functional muscle cell compartments, including membrane excitation, excitation-contraction coupling, contractile machinery, cytoskeleton scaffold, and energy supply systems. Variations within each compartment are limited by the need of matching fiber type properties between different compartments. Nerve activity is a major control mechanism of the fiber type profile, and multiple signaling pathways are implicated in activity-dependent changes of muscle fibers. The characterization of these pathways is raising increasing interest in clinical medicine, given the potentially beneficial effects of muscle fiber type switching in the prevention and treatment of metabolic diseases.
Collapse
Affiliation(s)
- Stefano Schiaffino
- Venetian Institute of Molecular Medicine, Department of Biomedical Sciences, University of Padova, Consiglio Nazionale delle Ricerche Institute of Neurosciences, and Department of Human Anatomy and Physiology, University of Padova, Padova, Italy
| | - Carlo Reggiani
- Venetian Institute of Molecular Medicine, Department of Biomedical Sciences, University of Padova, Consiglio Nazionale delle Ricerche Institute of Neurosciences, and Department of Human Anatomy and Physiology, University of Padova, Padova, Italy
| |
Collapse
|
46
|
Majumdar G, Rooney RJ, Johnson IM, Raghow R. Panhistone deacetylase inhibitors inhibit proinflammatory signaling pathways to ameliorate interleukin-18-induced cardiac hypertrophy. Physiol Genomics 2011; 43:1319-33. [PMID: 21954451 DOI: 10.1152/physiolgenomics.00048.2011] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
We investigated the genome-wide consequences of pan-histone deacetylase inhibitors (HDACIs) trichostatin A (TSA) and m-carboxycinnamic acid bis-hydroxamide (CBHA) in the hearts of BALB/c mice eliciting hypertrophy in response to interleukin-18 (IL-18). Both TSA and CBHA profoundly altered cardiac chromatin structure that occurred concomitantly with normalization of IL-18-induced gene expression and amelioration of cardiac hypertrophy. The hearts of mice exposed to IL-18+/-TSA or CBHA elicited distinct gene expression profiles. Of 184 genes that were differentially regulated by IL-18 and TSA, 33 were regulated in an opposite manner. The hearts of mice treated with IL-18 and/or CBHA elicited 147 differentially expressed genes (DEGs), a third of which were oppositely regulated by IL-18 and CBHA. Ingenuity Pathways and Kyoto Encyclopedia of Genes and Genomes analyses of DEGs showed that IL-18 impinged on TNF-α- and IFNγ-specific gene networks relegated to controlling immunity and inflammation, cardiac metabolism and energetics, and cell proliferation and apoptosis. These TNF-α- and IFNγ-specific gene networks, extensively connected with PI3K, MAPK, and NF-κB signaling pathways, were oppositely regulated by IL-18 and pan-HDACIs. Evidently, both TSA and CBHA caused a two- to fourfold induction of phosphatase and tensin homolog expression to counteract IL-18-induced proinflammatory signaling and cardiac hypertrophy.
Collapse
Affiliation(s)
- Gipsy Majumdar
- Department of Veterans Affairs Medical Center, University of Tennessee Health Science Center, Memphis, TN 38104, USA
| | | | | | | |
Collapse
|
47
|
Giulivi C, Ross-Inta C, Omanska-Klusek A, Napoli E, Sakaguchi D, Barrientos G, Allen PD, Pessah IN. Basal bioenergetic abnormalities in skeletal muscle from ryanodine receptor malignant hyperthermia-susceptible R163C knock-in mice. J Biol Chem 2011; 286:99-113. [PMID: 20978128 PMCID: PMC3013050 DOI: 10.1074/jbc.m110.153247] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2010] [Revised: 10/13/2010] [Indexed: 12/25/2022] Open
Abstract
Malignant hyperthermia (MH) and central core disease in humans have been associated with mutations in the skeletal ryanodine receptor (RyR1). Heterozygous mice expressing the human MH/central core disease RyR1 R163C mutation exhibit MH when exposed to halothane or heat stress. Considering that many MH symptoms resemble those that could ensue from a mitochondrial dysfunction (e.g. metabolic acidosis and hyperthermia) and that MH-susceptible mice or humans have a higher than normal cytoplasmic Ca(2+) concentration at rest, we evaluated the role of mitochondria in skeletal muscle from R163C compared with wild type mice under basal (untriggered) conditions. R163C skeletal muscle exhibited a significant increase in matrix Ca(2+), increased reactive oxygen species production, lower expression of mitochondrial proteins, and higher mtDNA copy number. These changes, in conjunction with lower myoglobin and glycogen contents, Myh4 and GAPDH transcript levels, GAPDH activity, and lower glucose utilization suggested a switch to a compromised bioenergetic state characterized by both low oxidative phosphorylation and glycolysis. The shift in bioenergetic state was accompanied by a dysregulation of Ca(2+)-responsive signaling pathways regulated by calcineurin and ERK1/2. Chronically elevated resting Ca(2+) in R163C skeletal muscle elicited the maintenance of a fast-twitch fiber program and the development of insulin resistance-like phenotype as part of a metabolic adaptation to the R163C RyR1 mutation.
Collapse
Affiliation(s)
- Cecilia Giulivi
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, California 95616, USA.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Seebacher F, Brand MD, Else PL, Guderley H, Hulbert AJ, Moyes CD. Plasticity of oxidative metabolism in variable climates: molecular mechanisms. Physiol Biochem Zool 2010; 83:721-32. [PMID: 20586603 DOI: 10.1086/649964] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Converting food to chemical energy (ATP) that is usable by cells is a principal requirement to sustain life. The rate of ATP production has to be sufficient for housekeeping functions, such as protein synthesis and maintaining membrane potentials, as well as for growth and locomotion. Energy metabolism is temperature sensitive, and animals respond to environmental variability at different temporal levels, from within-individual to evolutionary timescales. Here we review principal molecular mechanisms that underlie control of oxidative ATP production in response to climate variability. Nuclear transcription factors and coactivators control expression of mitochondrial proteins and abundance of mitochondria. Fatty acid and phospholipid concentrations of membranes influence the activity of membrane-bound proteins as well as the passive leak of protons across the mitochondrial membrane. Passive proton leak as well as protein-mediated proton leak across the inner mitochondrial membrane determine the efficacy of ATP production but are also instrumental in endothermic heat production and as a defense against reactive oxygen species. Both transcriptional mechanisms and membrane composition interact with environmental temperature and diet, and this interaction between diet and temperature in determining mitochondrial function links the two major environmental variables that are affected by changing climates. The limits to metabolic plasticity could be set by the production of reactive oxygen species leading to cellular damage, limits to substrate availability in mitochondria, and a disproportionally large increase in proton leak over ATP production.
Collapse
Affiliation(s)
- Frank Seebacher
- School of Biological Sciences, University of Sydney, Sydney, New South Wales 2006, Australia.
| | | | | | | | | | | |
Collapse
|
49
|
Yan Z, Okutsu M, Akhtar YN, Lira VA. Regulation of exercise-induced fiber type transformation, mitochondrial biogenesis, and angiogenesis in skeletal muscle. J Appl Physiol (1985) 2010; 110:264-74. [PMID: 21030673 DOI: 10.1152/japplphysiol.00993.2010] [Citation(s) in RCA: 232] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Skeletal muscle exhibits superb plasticity in response to changes in functional demands. Chronic increases of skeletal muscle contractile activity, such as endurance exercise, lead to a variety of physiological and biochemical adaptations in skeletal muscle, including mitochondrial biogenesis, angiogenesis, and fiber type transformation. These adaptive changes are the basis for the improvement of physical performance and other health benefits. This review focuses on recent findings in genetically engineered animal models designed to elucidate the mechanisms and functions of various signal transduction pathways and gene expression programs in exercise-induced skeletal muscle adaptations.
Collapse
Affiliation(s)
- Zhen Yan
- Department of Medicine, University of Virginia, Charlottesville, Virginia, USA.
| | | | | | | |
Collapse
|
50
|
Gundersen K. Excitation-transcription coupling in skeletal muscle: the molecular pathways of exercise. Biol Rev Camb Philos Soc 2010; 86:564-600. [PMID: 21040371 PMCID: PMC3170710 DOI: 10.1111/j.1469-185x.2010.00161.x] [Citation(s) in RCA: 176] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Muscle fibres have different properties with respect to force, contraction speed, endurance, oxidative/glycolytic capacity etc. Although adult muscle fibres are normally post-mitotic with little turnover of cells, the physiological properties of the pre-existing fibres can be changed in the adult animal upon changes in usage such as after exercise. The signal to change is mainly conveyed by alterations in the patterns of nerve-evoked electrical activity, and is to a large extent due to switches in the expression of genes. Thus, an excitation-transcription coupling must exist. It is suggested that changes in nerve-evoked muscle activity lead to a variety of activity correlates such as increases in free intracellular Ca2+ levels caused by influx across the cell membrane and/or release from the sarcoplasmatic reticulum, concentrations of metabolites such as lipids and ADP, hypoxia and mechanical stress. Such correlates are detected by sensors such as protein kinase C (PKC), calmodulin, AMP-activated kinase (AMPK), peroxisome proliferator-activated receptor δ (PPARδ), and oxygen dependent prolyl hydroxylases that trigger intracellular signaling cascades. These complex cascades involve several transcription factors such as nuclear factor of activated T-cells (NFAT), myocyte enhancer factor 2 (MEF2), myogenic differentiation factor (myoD), myogenin, PPARδ, and sine oculis homeobox 1/eyes absent 1 (Six1/Eya1). These factors might act indirectly by inducing gene products that act back on the cascade, or as ultimate transcription factors binding to and transactivating/repressing genes for the fast and slow isoforms of various contractile proteins and of metabolic enzymes. The determination of size and force is even more complex as this involves not only intracellular signaling within the muscle fibres, but also muscle stem cells called satellite cells. Intercellular signaling substances such as myostatin and insulin-like growth factor 1 (IGF-1) seem to act in a paracrine fashion. Induction of hypertrophy is accompanied by the satellite cells fusing to myofibres and thereby increasing the capacity for protein synthesis. These extra nuclei seem to remain part of the fibre even during subsequent atrophy as a form of muscle memory facilitating retraining. In addition to changes in myonuclear number during hypertrophy, changes in muscle fibre size seem to be caused by alterations in transcription, translation (per nucleus) and protein degradation.
Collapse
Affiliation(s)
- Kristian Gundersen
- Department of Molecular Biosciences, University of Oslo, P.O. Box 1041, Blindern, N-0316 Oslo, Norway.
| |
Collapse
|