1
|
Qiu X, Wu W, Zhang S, Huang C, Lin D. 3-Hydroxybutyrate Promotes Myoblast Proliferation and Differentiation through Energy Metabolism and GPR109a-Mediated Ca 2+-NFAT Signaling Pathways. J Proteome Res 2025; 24:2063-2080. [PMID: 40099866 DOI: 10.1021/acs.jproteome.4c01150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Skeletal muscle wasting is a critical clinical problem associated with several diseases that significantly impair patient outcomes due to the progressive loss of muscle mass and function. This study explores the potential of 3-hydroxybutyrate (3-HB) as a therapeutic agent to counteract muscle atrophy by promoting the proliferation and differentiation of C2C12 myoblasts. Using nuclear magnetic resonance (NMR)-based metabolomics analysis, we uncover the underlying mechanisms by which 3-HB exerts its effects. Our findings demonstrate that 3-HB exerts its effects through two distinct mechanisms: as a metabolic substrate and as a signaling molecule. As a metabolic substrate, 3-HB enhances myoblast energy efficiency by stimulating the expression of G protein-coupled receptor 109a (GPR109a), which subsequently upregulates the 3-HB transporters MCT1 and CD147, the utilization enzyme OXCT1, and phosphorylated AMPK, thereby increasing ATP production. As a signaling molecule, 3-HB activates GPR109a, promoting calcium influx, improving calcium homeostasis, and increasing the expression of Ca2+-related proteins such as CAMKK2. This signaling cascade activates calcineurin (CaN), facilitating NFAT translocation to the nucleus and gene expression that drives myoblast proliferation and differentiation. By elucidating the dual regulatory roles of 3-HB in energy metabolism and cellular signaling, this study not only advances our understanding of muscle physiology but also highlights the potential of 3-HB as a novel therapeutic approach for the prevention or treatment of skeletal muscle atrophy.
Collapse
Affiliation(s)
- Xu Qiu
- Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Wenfang Wu
- Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Shuya Zhang
- Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Caihua Huang
- Research and Communication Center of Exercise and Health, Xiamen University of Technology, Xiamen 361024, China
| | - Donghai Lin
- Key Laboratory for Chemical Biology of Fujian Province, MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| |
Collapse
|
2
|
Lian X, Jing Y, Luo T, Guo Y, Lin Y. Pharmacological interventions for the management of anesthesia and sedation in patients with Duchenne muscular dystrophy: a systematic review and meta-analysis. Front Med (Lausanne) 2025; 12:1497538. [PMID: 39931560 PMCID: PMC11808030 DOI: 10.3389/fmed.2025.1497538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 01/06/2025] [Indexed: 02/13/2025] Open
Abstract
Background Patients with duchenne muscular dystrophy (DMD) have an increased risk of complications when they undergo sedation or general anesthesia. However, due to improvements in cardiopulmonary therapies during anesthetic care, patients with DMD are experiencing an unprecedented duration of survival. We performed a systematic analysis to assess the benefits and risks of pharmacological interventions for the management of anesthesia and sedation in DMD patients. Methods We included any type of study reporting any drug intervention to manage anesthesia and sedation in participants previously diagnosed with DMD. Our primary outcomes were the onset time, recovery time, and neurodevelopmental disabilities. Seven electronic databases and three clinical trial registry platforms were searched. Data from the eligible studies were combined to calculate pooled risk ratios or standardized mean differences, and some included studies are presented in a narrative synthesis. Results Forty studies with 196 DMD participants were included in the analysis. Compared with those of the control group, the sensitivity of patients with DMD to neuromuscular blocking agents (NMBAs) may have resulted in a prolonged onset time [MD = -0.96, 95% CI (0.71, 2.60), I 2 = 33%, P < 0.0001] and recovery time [MD = 2.22, 95% CI (1.14, 3.30), I 2 = 76%, P < 0.0001] from anesthesia. The neuromuscular blocking effects showed a significant age dependence in DMD patients, and the safe use of 2 mg/kg sugammadex to antagonize deep neuromuscular blockade and rapid recovery has been reported. Furthermore, DMD patients are at risk of developing malignant hyperpyrexia with general/inhaled anesthesia, and dantrolene is often used for effective rescue. In addition, general anesthesia and central neuraxial blockade in patients with severe DMD are unsafe because respiratory depression and myocardial complications may occur after the administration of volatile anesthetics and depolarizing muscle relaxants (succinylcholine) during the induction of anesthesia. Conclusions Patients with DMD are more sensitive to NMBAs with delayed onset times and prolonged recovery times. Precautions for DMD patients should include quantitative neuromuscular monitoring, electrocardiographic monitoring and rapid airway protection throughout anesthesia. Compared with general anesthesia, regional anesthesia may be a relatively safe option.
Collapse
Affiliation(s)
- Xianghong Lian
- Department of Pharmacy, West China Second University Hospital, Sichuan University, Chengdu, China
- Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Yang Jing
- Department of Pharmacy, West China Second University Hospital, Sichuan University, Chengdu, China
- Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Ting Luo
- Department of Pharmacy, West China Second University Hospital, Sichuan University, Chengdu, China
- Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Yixin Guo
- Department of Pharmacy, West China Second University Hospital, Sichuan University, Chengdu, China
- Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Yunzhu Lin
- Department of Pharmacy, West China Second University Hospital, Sichuan University, Chengdu, China
- Evidence-Based Pharmacy Center, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| |
Collapse
|
3
|
Gupta S, Kishore A, Rishi V, Aggarwal A. Mitochondria and its epigenetic dynamics: Insight into synaptic regulation and synaptopathies. Funct Integr Genomics 2025; 25:26. [PMID: 39849126 DOI: 10.1007/s10142-025-01530-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 01/04/2025] [Accepted: 01/08/2025] [Indexed: 01/25/2025]
Abstract
Mitochondria, the cellular powerhouses, are pivotal to neuronal function and health, particularly through their role in regulating synaptic structure and function. Spine reprogramming, which underlies synapse development, depends heavily on mitochondrial dynamics-such as biogenesis, fission, fusion, and mitophagy as well as functions including ATP production, calcium (Ca2+) regulation, and retrograde signaling. Mitochondria supply the energy necessary for assisting synapse development and plasticity, while also regulating intracellular Ca2+ homeostasis to prevent excitotoxicity and support synaptic neurotransmission. Additionally, the dynamic processes of mitochondria ensure mitochondrial quality and adaptability, which are essential for maintaining effective synaptic activity. Emerging evidence highlights the significant role of epigenetic modifications in regulating mitochondrial dynamics and function. Epigenetic changes influence gene expression, which in turn affects mitochondrial activity, ensuring coordinated responses necessary for synapse development. Furthermore, metabolic changes within mitochondria can impact the epigenetic machinery, thereby modulating gene expression patterns that support synaptic integrity. Altered epigenetic regulation affecting mitochondrial dynamics and functions is linked to several neurological disorders, including Amyotrophic Lateral Sclerosis, Huntington's, Alzheimer's, and Parkinson's diseases, emphasizing its crucial function. The review delves into the molecular machinery involved in mitochondrial dynamics, ATP and Ca2+ regulation, highlighting the role of key proteins that facilitate the processes. Additionally, it also shed light on the emerging epigenetic factors influencing these regulations. It provides a thorough summary on the current understanding of the role of mitochondria in synapse development and emphasizes the importance of both molecular and epigenetic mechanisms in maintaining synaptic integrity.
Collapse
Affiliation(s)
- Shiwangi Gupta
- National Agri-Food and Biomanufacturing Institute, Sector-81, SAS Nagar, Knowledge City, Punjab, India
- Department of Biotechnology, Sector-25, Panjab University, BMS block I, Chandigarh, India
| | - Abhinoy Kishore
- Indian Institute of Science, Bengaluru, India
- Chandigarh Group of Colleges, Landran, Punjab, India
| | - Vikas Rishi
- National Agri-Food and Biomanufacturing Institute, Sector-81, SAS Nagar, Knowledge City, Punjab, India
| | - Aanchal Aggarwal
- National Agri-Food and Biomanufacturing Institute, Sector-81, SAS Nagar, Knowledge City, Punjab, India.
| |
Collapse
|
4
|
García-Puga M, Gerenu G, Bargiela A, Espinosa-Espinosa J, Mosqueira-Martín L, Sagartzazu-Aizpurua M, Aizpurua JM, Vallejo-Illarramendi A, Artero R, López de Munain A, Matheu A. A Novel Class of FKBP12 Ligands Rescues Premature Aging Phenotypes Associated with Myotonic Dystrophy Type 1. Cells 2024; 13:1939. [PMID: 39682688 PMCID: PMC11639790 DOI: 10.3390/cells13231939] [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: 10/09/2024] [Revised: 11/16/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Background: Myotonic dystrophy type 1 (DM1) is an autosomal dominant disorder clinically characterized by progressive muscular weakness and multisystem degeneration, which correlates with the size of CTG expansion and MBLN decrease. These changes induce a calcium and redox homeostasis imbalance in several models that recapitulate the features of premature tissue aging. In this study, we characterized the impact of a new family of FKBP12 ligands (generically named MPs or MP compounds) designed to stabilize FKBP12 binding to the ryanodine receptors and normalize calcium dysregulation under oxidative stress. Methods: Human primary fibroblasts from DM1 patients and control donors, treated with MP compounds or not, were used for functional studies of cell viability, proliferation, and metabolism. The gene expression profile in treated cells was determined using RNA sequencing. The impact of MP compounds in vivo was evaluated in a Drosophila model of the disease using locomotor activity and longevity studies. Results: The treatment with different MP compounds reversed oxidative stress and impaired cell viability and proliferation, mitochondrial activity, and metabolic defects in DM1-derived primary fibroblasts. RNA sequencing analysis confirmed the restoration of molecular pathways related to calcium and redox homeostasis and additional pathways, including the cell cycle and metabolism. This analysis also revealed the rescue of alternative splicing events in DM1 fibroblasts treated with MP compounds. Importantly, treatment with MP compounds significantly extended the lifespan and improved the locomotor activity of a Drosophila model of the DM1 disease, and restored molecular defects characteristic of the disease in vivo. Conclusions: Our results revealed that MP compounds rescue multiple premature aging phenotypes described in DM1 models and decipher the benefits of this new family of compounds in the pre-clinical setting of DM1.
Collapse
Affiliation(s)
- Mikel García-Puga
- Cellular Oncology Group, Biogipuzkoa Health Research Institute, Paseo Dr. Beguiristain s/n, 20014 San Sebastian, Spain;
- Neuroscience Area, Biogipuzkoa Health Research Institute, Biodonostia Institute, 20014 San Sebastian, Spain; (G.G.); (L.M.-M.); (A.V.-I.)
- CIBERNED, CIBER, Carlos III Institute, 28029 Madrid, Spain
- Department of Health Sciences, Public University of Navarre (UPNA), Health Sciences Campus, Avda. de Barañain s/n, 31008 Pamplona, Spain
| | - Gorka Gerenu
- Neuroscience Area, Biogipuzkoa Health Research Institute, Biodonostia Institute, 20014 San Sebastian, Spain; (G.G.); (L.M.-M.); (A.V.-I.)
- CIBERNED, CIBER, Carlos III Institute, 28029 Madrid, Spain
- IKERBASQUE, Basque Foundation for Science, 48009 Bilbao, Spain
| | - Ariadna Bargiela
- Translational Genomics Group, University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, 46100 Burjasot, Spain; (A.B.); (J.E.-E.); (R.A.)
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain
- CIBERER, CIBER, Carlos III Institute, 28029 Madrid, Spain
| | - Jorge Espinosa-Espinosa
- Translational Genomics Group, University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, 46100 Burjasot, Spain; (A.B.); (J.E.-E.); (R.A.)
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain
- CIBERER, CIBER, Carlos III Institute, 28029 Madrid, Spain
| | - Laura Mosqueira-Martín
- Neuroscience Area, Biogipuzkoa Health Research Institute, Biodonostia Institute, 20014 San Sebastian, Spain; (G.G.); (L.M.-M.); (A.V.-I.)
- Group of Neuroscience, Departments of Pediatrics and Neuroscience, Faculty of Medicine and Nursing, University of the Basque Country, 20014 San Sebastian, Spain
| | - Maialen Sagartzazu-Aizpurua
- Joxe Mari Korta R&D Center, Department of Organic Chemistry I, University of the Basque Country, 20014 San Sebastian, Spain; (M.S.-A.); (J.M.A.)
| | - Jesús M. Aizpurua
- Joxe Mari Korta R&D Center, Department of Organic Chemistry I, University of the Basque Country, 20014 San Sebastian, Spain; (M.S.-A.); (J.M.A.)
| | - Ainara Vallejo-Illarramendi
- Neuroscience Area, Biogipuzkoa Health Research Institute, Biodonostia Institute, 20014 San Sebastian, Spain; (G.G.); (L.M.-M.); (A.V.-I.)
- CIBERNED, CIBER, Carlos III Institute, 28029 Madrid, Spain
- Group of Neuroscience, Departments of Pediatrics and Neuroscience, Faculty of Medicine and Nursing, University of the Basque Country, 20014 San Sebastian, Spain
| | - Rubén Artero
- Translational Genomics Group, University Institute for Biotechnology and Biomedicine (BIOTECMED), University of Valencia, 46100 Burjasot, Spain; (A.B.); (J.E.-E.); (R.A.)
- INCLIVA Biomedical Research Institute, 46010 Valencia, Spain
- CIBERER, CIBER, Carlos III Institute, 28029 Madrid, Spain
| | - Adolfo López de Munain
- Neuroscience Area, Biogipuzkoa Health Research Institute, Biodonostia Institute, 20014 San Sebastian, Spain; (G.G.); (L.M.-M.); (A.V.-I.)
- CIBERNED, CIBER, Carlos III Institute, 28029 Madrid, Spain
- Group of Neuroscience, Departments of Pediatrics and Neuroscience, Faculty of Medicine and Nursing, University of the Basque Country, 20014 San Sebastian, Spain
- Neurology Department, Donostia University Hospital, Osakidetza, 20014 San Sebastian, Spain
- Department of Internal Medicine, Faculty of Health Sciences, University of Deusto, 48007 Bilbao, Spain
| | - Ander Matheu
- Cellular Oncology Group, Biogipuzkoa Health Research Institute, Paseo Dr. Beguiristain s/n, 20014 San Sebastian, Spain;
- IKERBASQUE, Basque Foundation for Science, 48009 Bilbao, Spain
- CIBERFES, CIBER, Carlos III Institute, 28029 Madrid, Spain
| |
Collapse
|
5
|
Vesga-Castro C, Mosqueira-Martín L, Ubiria-Urkola P, Marco-Moreno P, González-Imaz K, Rendon-Hinestroza J, Vallejo-Illarramendi A, Paredes J. Development of an in vitro platform for the analysis of contractile and calcium dynamics in single human myotubes. LAB ON A CHIP 2024; 24:4741-4754. [PMID: 39264341 DOI: 10.1039/d3lc00442b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
In vitro myotube cultures are widely used as models for studying muscle pathophysiology, but their limited maturation and heterogeneity pose significant challenges for functional analyses. While they remain the gold standard for studying muscle function in vitro, myotube cultures do not fully recapitulate the complexity and native features of muscle fibers, which may compromise their ability to predict in vivo outcomes. To promote maturation and decrease heterogeneity, we have incorporated engineered structures into myotube cultures, based on a PDMS thin layer with micrometer-sized grooves (μGrooves) placed over a glass substrate. Different sizes and shapes of μGrooves were tested for their ability to promote alignment and fusion of myoblasts and enhance their differentiation into myotubes. A 24 hour electrical field stimulation protocol (4 V, 6 ms, 0.1 Hz) was used to further promote myotube maturation, after which several myotube features were assessed, including myotube alignment, width, fusion index, contractile function, and calcium handling. Our results indicate superior calcium and contractile performance in μGrooved myotubes, particularly with the 100 μm-width 700 μm-long geometry (7 : 1). This platform generated homogeneous and isolated myotubes that reproduced native muscle features, such as excitation-contraction coupling and force-frequency responses. Overall, our 2D muscle platform enables robust high-content assays of calcium dynamics and contractile readouts with increased sensitivity and reproducibility compared to traditional myotube cultures, making it particularly suitable for screening therapeutic candidates for different muscle pathologies.
Collapse
Affiliation(s)
- Camila Vesga-Castro
- Tecnun School of Engineering, University of Navarra, Manuel de Lardizábal 13, 20018 San Sebastián, Spain.
- Biomedical Engineering Center, University of Navarra, Campus Universitario, 31080, Pamplona, Spain
| | - Laura Mosqueira-Martín
- Group of Neuroscience, Department of Pediatrics, Faculty of Medicine and Nursing, Donostia Hospital, UPV/EHU, 20014 San Sebastian, Spain.
- Group of Neuromuscular Diseases, IIS Biogipuzkoa, 20014 San Sebastian, Spain
- CIBERNED, Ministry of Science and Innovation, Instituto de Salud Carlos III, 28031 Madrid, Spain
| | - Paul Ubiria-Urkola
- Tecnun School of Engineering, University of Navarra, Manuel de Lardizábal 13, 20018 San Sebastián, Spain.
- Biomedical Engineering Center, University of Navarra, Campus Universitario, 31080, Pamplona, Spain
| | - Pablo Marco-Moreno
- Group of Neuroscience, Department of Pediatrics, Faculty of Medicine and Nursing, Donostia Hospital, UPV/EHU, 20014 San Sebastian, Spain.
- Group of Neuromuscular Diseases, IIS Biogipuzkoa, 20014 San Sebastian, Spain
- CIBERNED, Ministry of Science and Innovation, Instituto de Salud Carlos III, 28031 Madrid, Spain
| | - Klaudia González-Imaz
- Group of Neuroscience, Department of Pediatrics, Faculty of Medicine and Nursing, Donostia Hospital, UPV/EHU, 20014 San Sebastian, Spain.
- Group of Neuromuscular Diseases, IIS Biogipuzkoa, 20014 San Sebastian, Spain
- CIBERNED, Ministry of Science and Innovation, Instituto de Salud Carlos III, 28031 Madrid, Spain
| | - Jorge Rendon-Hinestroza
- Group of Neuroscience, Department of Pediatrics, Faculty of Medicine and Nursing, Donostia Hospital, UPV/EHU, 20014 San Sebastian, Spain.
- Group of Neuromuscular Diseases, IIS Biogipuzkoa, 20014 San Sebastian, Spain
| | - Ainara Vallejo-Illarramendi
- Group of Neuroscience, Department of Pediatrics, Faculty of Medicine and Nursing, Donostia Hospital, UPV/EHU, 20014 San Sebastian, Spain.
- Group of Neuromuscular Diseases, IIS Biogipuzkoa, 20014 San Sebastian, Spain
- CIBERNED, Ministry of Science and Innovation, Instituto de Salud Carlos III, 28031 Madrid, Spain
| | - Jacobo Paredes
- Tecnun School of Engineering, University of Navarra, Manuel de Lardizábal 13, 20018 San Sebastián, Spain.
- Biomedical Engineering Center, University of Navarra, Campus Universitario, 31080, Pamplona, Spain
| |
Collapse
|
6
|
Villani KR, Zhong R, Henley-Beasley CS, Rastelli G, Harris E, Boncompagni S, Barton ER, Wei-LaPierre L. Loss of Calpain 3 dysregulates store-operated calcium entry and its exercise response in mice. FASEB J 2024; 38:e23825. [PMID: 39031532 PMCID: PMC11299996 DOI: 10.1096/fj.202400697r] [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: 03/30/2024] [Revised: 06/18/2024] [Accepted: 07/08/2024] [Indexed: 07/22/2024]
Abstract
Limb-Girdle Muscular Dystrophy R1/2A (LGMD R1/2A) is caused by mutations in the CAPN3 gene encoding Calpain 3, a skeletal-muscle specific, Ca2+-dependent protease. Localization of Calpain 3 within the triad suggests it contributes to Ca2+ homeostasis. Through live-cell Ca2+ measurements, muscle mechanics, immunofluorescence, and electron microscopy (EM) in Capn3 deficient (C3KO) and wild-type (WT) mice, we determined whether loss of Calpain 3 altered Store-Operated Calcium Entry (SOCE) activity. Direct Ca2+ influx measurements revealed loss of Capn3 elicits elevated resting SOCE and increased resting cytosolic Ca2+, supported by high incidence of calcium entry units (CEUs) observed by EM. C3KO and WT mice were subjected to a single bout of treadmill running to elicit SOCE. Within 1HR post-treadmill running, C3KO mice exhibited diminished force production in extensor digitorum longus muscles and a greater decay of Ca2+ transients in flexor digitorum brevis muscle fibers during repetitive stimulation. Striking evidence for impaired exercise-induced SOCE activation in C3KO mice included poor colocalization of key SOCE proteins, stromal-interacting molecule 1 (STIM1) and ORAI1, combined with disappearance of CEUs in C3KO muscles. These results demonstrate that Calpain 3 is a key regulator of SOCE in skeletal muscle and identify SOCE dysregulation as a contributing factor to LGMD R1/2A pathology.
Collapse
Affiliation(s)
- Katelyn R. Villani
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, FL, USA
| | - Renjia Zhong
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, FL, USA
- Department of Emergency Medicine, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - C. Spencer Henley-Beasley
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, FL, USA
- Myology Institute, University of Florida, FL, USA
| | - Giorgia Rastelli
- Center for Advanced Studies and Technology and Department of Neuroscience, Imaging and Clinical Sciences, University G. d’Annunzio of Chieti–Pescara, Chieti, Italy
| | - Erin Harris
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, FL, USA
| | - Simona Boncompagni
- Center for Advanced Studies and Technology and Department of Neuroscience, Imaging and Clinical Sciences, University G. d’Annunzio of Chieti–Pescara, Chieti, Italy
| | - Elisabeth R. Barton
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, FL, USA
- Myology Institute, University of Florida, FL, USA
| | - Lan Wei-LaPierre
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, FL, USA
- Myology Institute, University of Florida, FL, USA
| |
Collapse
|
7
|
Aldharee H. Duchenne muscular dystrophy in Saudi Arabia: a review of the current literature. Front Neurol 2024; 15:1392274. [PMID: 39087004 PMCID: PMC11288836 DOI: 10.3389/fneur.2024.1392274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 07/08/2024] [Indexed: 08/02/2024] Open
Abstract
In the past three decades, significant improvements have occurred in the study of Duchenne muscular dystrophy (DMD). DMD is a rare, severe neuromuscular disease that causes death due to cardiovascular and respiratory complications among affected boys. Since the 1980s, ongoing preclinical and clinical studies have been conducted to explore the disease in depth and discover potential therapeutic strategies. In Saudi Arabia, it is unclear whether health services and research efforts are keeping pace with global achievements. Therefore, this review aims to explore the diagnostic and management strategies and research efforts in Saudi Arabia over the past three decades. I searched the PubMed/Medline, Scopus, and Web of Science databases and included all published articles on the epidemiology, genetics, diagnosis, and management of DMD/BMD in this review. The findings suggest a lack of local standardized diagnostic strategies, a poor understanding of epidemiology and common pathogenic variants, and a critical need for preclinical and clinical research. At the time of writing, no such comprehensive review has been published. Challenges, limitations, and future perspectives are also discussed in this article.
Collapse
Affiliation(s)
- Hitham Aldharee
- Department of Pathology, College of Medicine, Qassim University, Buraidah, Saudi Arabia
| |
Collapse
|
8
|
Gandhi S, Sweeney HL, Hart CC, Han R, Perry CGR. Cardiomyopathy in Duchenne Muscular Dystrophy and the Potential for Mitochondrial Therapeutics to Improve Treatment Response. Cells 2024; 13:1168. [PMID: 39056750 PMCID: PMC11274633 DOI: 10.3390/cells13141168] [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: 01/27/2024] [Revised: 07/05/2024] [Accepted: 07/06/2024] [Indexed: 07/28/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive neuromuscular disease caused by mutations to the dystrophin gene, resulting in deficiency of dystrophin protein, loss of myofiber integrity in skeletal and cardiac muscle, and eventual cell death and replacement with fibrotic tissue. Pathologic cardiac manifestations occur in nearly every DMD patient, with the development of cardiomyopathy-the leading cause of death-inevitable by adulthood. As early cardiac abnormalities are difficult to detect, timely diagnosis and appropriate treatment modalities remain a challenge. There is no cure for DMD; treatment is aimed at delaying disease progression and alleviating symptoms. A comprehensive understanding of the pathophysiological mechanisms is crucial to the development of targeted treatments. While established hypotheses of underlying mechanisms include sarcolemmal weakening, upregulation of pro-inflammatory cytokines, and perturbed ion homeostasis, mitochondrial dysfunction is thought to be a potential key contributor. Several experimental compounds targeting the skeletal muscle pathology of DMD are in development, but the effects of such agents on cardiac function remain unclear. The synergistic integration of small molecule- and gene-target-based drugs with metabolic-, immune-, or ion balance-enhancing compounds into a combinatorial therapy offers potential for treating dystrophin deficiency-induced cardiomyopathy, making it crucial to understand the underlying mechanisms driving the disorder.
Collapse
Affiliation(s)
- Shivam Gandhi
- School of Kinesiology and Health Science, Muscle Health Research Centre, York University, Toronto, ON M3J 1P3, Canada
| | - H. Lee Sweeney
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, USA; (H.L.S.); (C.C.H.)
- Myology Institute, University of Florida, Gainesville, FL 32610, USA
| | - Cora C. Hart
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, USA; (H.L.S.); (C.C.H.)
- Myology Institute, University of Florida, Gainesville, FL 32610, USA
| | - Renzhi Han
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Christopher G. R. Perry
- School of Kinesiology and Health Science, Muscle Health Research Centre, York University, Toronto, ON M3J 1P3, Canada
| |
Collapse
|
9
|
Oswalia J, Singh S, Gautam V, Arya R. Altered autophagic flux in GNE mutant cells of Indian origin: Potential drug target for GNE myopathy. Exp Cell Res 2024; 440:114118. [PMID: 38852763 DOI: 10.1016/j.yexcr.2024.114118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/11/2024]
Abstract
Autophagy phenomenon in the cell maintains proteostasis balance by eliminating damaged organelles and protein aggregates. Imbalance in autophagic flux may cause accumulation of protein aggregates in various neurodegenerative disorders. Regulation of autophagy by either calcium or chaperone play a key role in the removal of protein aggregates from the cell. The neuromuscular rare genetic disorder, GNE Myopathy, is characterized by accumulation of rimmed vacuoles having protein aggregates of β-amyloid and tau that may result from altered autophagic flux. In the present study, the autophagic flux was deciphered in HEK cell-based model for GNE Myopathy harbouring GNE mutations of Indian origin. The refolding activity of HSP70 chaperone was found to be reduced in GNE mutant cells compared to wild type controls. The autophagic markers LC3II/I ratio was altered with increased number of autophagosome formation in GNE mutant cells compared to wild type cells. The cytosolic calcium levels were also increased in GNE mutant cells of Indian origin. Interestingly, treatment of GNE mutant cells with HSP70 activator, BGP-15, restored the expression and refolding activity of HSP70 along with autophagosome formation. Treatment with calcium chelator, BAPTA-AM restored the cytoplasmic calcium levels and autophagosome formation but not LC3II/I ratio significantly. Our study provides insights towards GNE mutation specific response for autophagy regulation and opens up a therapeutic advancement area in calcium signalling and HSP70 function for GNE related Myopathy.
Collapse
Affiliation(s)
- Jyoti Oswalia
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067, India.
| | - Shagun Singh
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067, India.
| | - Vaishali Gautam
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067, India.
| | - Ranjana Arya
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067, India.
| |
Collapse
|
10
|
Rudolph TE, Roths M, Freestone AD, Yap SQ, Michael A, Rhoads RP, White-Springer SH, Baumgard LH, Selsby JT. Biological sex impacts oxidative stress in skeletal muscle in a porcine heat stress model. Am J Physiol Regul Integr Comp Physiol 2024; 326:R578-R587. [PMID: 38708546 PMCID: PMC11381024 DOI: 10.1152/ajpregu.00268.2023] [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: 11/30/2023] [Revised: 04/03/2024] [Accepted: 04/25/2024] [Indexed: 05/07/2024]
Abstract
Oxidative stress contributes to heat stress (HS)-mediated alterations in skeletal muscle; however, the extent to which biological sex mediates oxidative stress during HS remains unknown. We hypothesized muscle from males would be more resistant to oxidative stress caused by HS than muscle from females. To address this, male and female pigs were housed in thermoneutral conditions (TN; 20.8 ± 1.6°C; 62.0 ± 4.7% relative humidity; n = 8/sex) or subjected to HS (39.4 ± 0.6°C; 33.7 ± 6.3% relative humidity) for 1 (HS1; n = 8/sex) or 7 days (HS7; n = 8/sex) followed by collection of the oxidative portion of the semitendinosus. Although HS increased muscle temperature, by 7 days, muscle from heat-stressed females was cooler than muscle from heat-stressed males (0.3°C; P < 0.05). Relative protein abundance of 4-hydroxynonenal (4-HNE)-modified proteins increased in HS1 females compared with TN (P = 0.05). Furthermore, malondialdehyde (MDA)-modified proteins and 8-hydroxy-2'-deoxyguanosine (8-OHdG) concentration, a DNA damage marker, was increased in HS7 females compared with TN females (P = 0.05). Enzymatic activities of catalase and superoxide dismutase (SOD) remained similar between groups; however, glutathione peroxidase (GPX) activity decreased in HS7 females compared with TN and HS1 females (P ≤ 0.03) and HS7 males (P = 0.02). Notably, HS increased skeletal muscle Ca2+ deposition (P = 0.05) and was greater in HS1 females compared with TN females (P < 0.05). Heat stress increased sarco(endo)plasmic reticulum Ca2+ ATPase (SERCA)2a protein abundance (P < 0.01); however, Ca2+ ATPase activity remained similar between groups. Overall, despite having lower muscle temperature, muscle from heat-stressed females had increased markers of oxidative stress and calcium deposition than muscle from males following identical environmental exposure.NEW & NOTEWORTHY Heat stress is a global threat to human health and agricultural production. We demonstrated that following 7 days of heat stress, skeletal muscle from females was more susceptible to oxidative stress than muscle from males in a porcine model, despite cooler muscle temperatures. The vulnerability to heat stress-induced oxidative stress in females may be driven, at least in part, by decreased antioxidant capacity and calcium dysregulation.
Collapse
Affiliation(s)
- Tori E Rudolph
- Department of Animal Science, Iowa State University, Ames, Iowa, United States
| | - Melissa Roths
- Department of Animal Science, Iowa State University, Ames, Iowa, United States
| | - Alyssa D Freestone
- Department of Animal Science, Iowa State University, Ames, Iowa, United States
| | - Sau Qwan Yap
- Department of Animal Science, Iowa State University, Ames, Iowa, United States
| | - Alyona Michael
- Department of Vet Diagnostic & Production Animal Med, Iowa State University, Ames, Iowa, United States
| | - Robert P Rhoads
- School of Animal Sciences, Virginia Tech, Blacksburg, Virginia, United States
| | - Sarah H White-Springer
- Department of Animal Science, Texas A&M University and Texas A&M AgriLife Research, College Station, Texas, United States
- Department of Kinesiology and Sport Management, Texas A&M University, College Station, Texas, United States
| | - Lance H Baumgard
- Department of Animal Science, Iowa State University, Ames, Iowa, United States
| | - Joshua T Selsby
- Department of Animal Science, Iowa State University, Ames, Iowa, United States
| |
Collapse
|
11
|
Quinn CJ, Cartwright EJ, Trafford AW, Dibb KM. On the role of dysferlin in striated muscle: membrane repair, t-tubules and Ca 2+ handling. J Physiol 2024; 602:1893-1910. [PMID: 38615232 DOI: 10.1113/jp285103] [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: 06/27/2023] [Accepted: 03/05/2024] [Indexed: 04/15/2024] Open
Abstract
Dysferlin is a 237 kDa membrane-associated protein characterised by multiple C2 domains with a diverse role in skeletal and cardiac muscle physiology. Mutations in DYSF are known to cause various types of human muscular dystrophies, known collectively as dysferlinopathies, with some patients developing cardiomyopathy. A myriad of in vitro membrane repair studies suggest that dysferlin plays an integral role in the membrane repair complex in skeletal muscle. In comparison, less is known about dysferlin in the heart, but mounting evidence suggests that dysferlin's role is similar in both muscle types. Recent findings have shown that dysferlin regulates Ca2+ handling in striated muscle via multiple mechanisms and that this becomes more important in conditions of stress. Maintenance of the transverse (t)-tubule network and the tight coordination of excitation-contraction coupling are essential for muscle contractility. Dysferlin regulates the maintenance and repair of t-tubules, and it is suspected that dysferlin regulates t-tubules and sarcolemmal repair through a similar mechanism. This review focuses on the emerging complexity of dysferlin's activity in striated muscle. Such insights will progress our understanding of the proteins and pathways that regulate basic heart and skeletal muscle function and help guide research into striated muscle pathology, especially that which arises due to dysferlin dysfunction.
Collapse
Affiliation(s)
- C J Quinn
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, 3.14 Core Technology Facility, Manchester, UK
| | - E J Cartwright
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, 3.14 Core Technology Facility, Manchester, UK
| | - A W Trafford
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, 3.14 Core Technology Facility, Manchester, UK
| | - K M Dibb
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, 3.14 Core Technology Facility, Manchester, UK
| |
Collapse
|
12
|
Villani KR, Zhong R, Henley-Beasley CS, Rastelli G, Boncompagni S, Barton ER, Wei-LaPierre L. Loss of calpain 3 dysregulates store-operated calcium entry and its exercise response in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.12.575391. [PMID: 38293127 PMCID: PMC10827051 DOI: 10.1101/2024.01.12.575391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Limb-Girdle Muscular Dystrophy 2A (LGMD2A) is caused by mutations in the CAPN3 gene encoding Calpain 3, a skeletal-muscle specific, Ca2+-dependent protease. Localization of Calpain 3 within the triad suggests it contributes to Ca2+ homeostasis. Through live-cell Ca2+ measurements, muscle mechanics, immunofluorescence, and electron microscopy (EM) in Capn3 deficient (C3KO) and wildtype (WT) mice, we determined if loss of Calpain 3 altered Store-Operated Calcium Entry (SOCE) activity. Direct Ca2+ influx measurements revealed loss of Capn3 elicits elevated resting SOCE and increased resting cytosolic Ca2+, supported by high incidence of calcium entry units (CEUs) observed by EM. C3KO and WT mice were subjected to a single bout of treadmill running to elicit SOCE. Within 1HR post-treadmill running, C3KO mice exhibited diminished force production in extensor digitorum longus muscles and a greater decay of Ca2+ transients in flexor digitorum brevis muscle fibers during repetitive stimulation. Striking evidence for impaired exercise-induced SOCE activation in C3KO mice included poor colocalization of key SOCE proteins, stromal-interacting molecule 1 (STIM1) and ORAI1, combined with disappearance of CEUs in C3KO muscles. These results demonstrate that Calpain 3 is a key regulator of SOCE in skeletal muscle and identify SOCE dysregulation as a contributing factor to LGMD2A pathology.
Collapse
Affiliation(s)
- Katelyn R. Villani
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, FL, USA
| | - Renjia Zhong
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, FL, USA
- Department of Emergency Medicine, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - C. Spencer Henley-Beasley
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, FL, USA
- Myology Institute, University of Florida, FL, USA
| | - Giorgia Rastelli
- Center for Advanced Studies and Technology and Department of Neuroscience, Imaging and Clinical Sciences, University G. d’Annunzio of Chieti–Pescara, Chieti, Italy
| | - Simona Boncompagni
- Center for Advanced Studies and Technology and Department of Neuroscience, Imaging and Clinical Sciences, University G. d’Annunzio of Chieti–Pescara, Chieti, Italy
| | - Elisabeth R. Barton
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, FL, USA
- Myology Institute, University of Florida, FL, USA
| | - Lan Wei-LaPierre
- Department of Applied Physiology and Kinesiology, College of Health and Human Performance, University of Florida, FL, USA
- Myology Institute, University of Florida, FL, USA
| |
Collapse
|
13
|
Roman B, Mastoor Y, Zhang Y, Gross D, Springer D, Liu C, Glancy B, Murphy E. Loss of mitochondrial Ca 2+ uptake protein 3 impairs skeletal muscle calcium handling and exercise capacity. J Physiol 2024; 602:113-128. [PMID: 38018177 PMCID: PMC10824360 DOI: 10.1113/jp284894] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 11/06/2023] [Indexed: 11/30/2023] Open
Abstract
Mitochondrial calcium concentration ([Ca2+ ]m ) plays an essential role in bioenergetics, and loss of [Ca2+ ]m homeostasis can trigger diseases and cell death in numerous cell types. Ca2+ uptake into mitochondria occurs via the mitochondrial Ca2+ uniporter (MCU), which is regulated by three mitochondrial Ca2+ uptake (MICU) proteins localized in the intermembrane space, MICU1, 2, and 3. We generated a mouse model of systemic MICU3 ablation and examined its physiological role in skeletal muscle. We found that loss of MICU3 led to impaired exercise capacity. When the muscles were directly stimulated there was a decrease in time to fatigue. MICU3 ablation significantly increased the maximal force of the KO muscle and altered fibre type composition with an increase in the ratio of type IIb (low oxidative capacity) to type IIa (high oxidative capacity) fibres. Furthermore, MICU3-KO mitochondria have reduced uptake of Ca2+ and increased phosphorylation of pyruvate dehydrogenase, indicating that KO animals contain less Ca2+ in their mitochondria. Skeletal muscle from MICU3-KO mice exhibited lower net oxidation of NADH during electrically stimulated muscle contraction compared with wild-type. These data demonstrate that MICU3 plays a role in skeletal muscle physiology by setting the proper threshold for mitochondrial Ca2+ uptake, which is important for matching energy demand and supply in muscle. KEY POINTS: Mitochondrial calcium uptake is an important regulator of bioenergetics and cell death and is regulated by the mitochondrial calcium uniporter (MCU) and three calcium sensitive regulatory proteins (MICU1, 2 and 3). Loss of MICU3 leads to impaired exercise capacity and decreased time to skeletal muscle fatigue. Skeletal muscle from MICU3-KO mice exhibits a net oxidation of NADH during electrically stimulated muscle contractions, suggesting that MICU3 plays a role in skeletal muscle physiology by matching energy demand and supply.
Collapse
Affiliation(s)
| | | | - Yingfan Zhang
- Muscle Energetics, NHLBI, and NIAMS, NIH, Bethesda, MD, USA
| | - Dennis Gross
- Cardiac Physiology, NHLBI, NIH, Bethesda, MD, USA
| | | | - Chengyu Liu
- Transgenic Core, NHLBI, NIH, Bethesda, MD, USA
| | - Brian Glancy
- Muscle Energetics, NHLBI, and NIAMS, NIH, Bethesda, MD, USA
- Transgenic Core, NHLBI, NIH, Bethesda, MD, USA
| | | |
Collapse
|
14
|
Rawls A, Diviak BK, Smith CI, Severson GW, Acosta SA, Wilson-Rawls J. Pharmacotherapeutic Approaches to Treatment of Muscular Dystrophies. Biomolecules 2023; 13:1536. [PMID: 37892218 PMCID: PMC10605463 DOI: 10.3390/biom13101536] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Muscular dystrophies are a heterogeneous group of genetic muscle-wasting disorders that are subdivided based on the region of the body impacted by muscle weakness as well as the functional activity of the underlying genetic mutations. A common feature of the pathophysiology of muscular dystrophies is chronic inflammation associated with the replacement of muscle mass with fibrotic scarring. With the progression of these disorders, many patients suffer cardiomyopathies with fibrosis of the cardiac tissue. Anti-inflammatory glucocorticoids represent the standard of care for Duchenne muscular dystrophy, the most common muscular dystrophy worldwide; however, long-term exposure to glucocorticoids results in highly adverse side effects, limiting their use. Thus, it is important to develop new pharmacotherapeutic approaches to limit inflammation and fibrosis to reduce muscle damage and promote repair. Here, we examine the pathophysiology, genetic background, and emerging therapeutic strategies for muscular dystrophies.
Collapse
Affiliation(s)
- Alan Rawls
- School of Life Sciences, Arizona State University, Tempe, AZ 85287-4501, USA; (B.K.D.); (C.I.S.); (G.W.S.); (S.A.A.)
| | - Bridget K. Diviak
- School of Life Sciences, Arizona State University, Tempe, AZ 85287-4501, USA; (B.K.D.); (C.I.S.); (G.W.S.); (S.A.A.)
- Molecular and Cellular Biology Graduate Program, School of Life Sciences, Tempe, AZ 85287 4501, USA
| | - Cameron I. Smith
- School of Life Sciences, Arizona State University, Tempe, AZ 85287-4501, USA; (B.K.D.); (C.I.S.); (G.W.S.); (S.A.A.)
- Molecular and Cellular Biology Graduate Program, School of Life Sciences, Tempe, AZ 85287 4501, USA
| | - Grant W. Severson
- School of Life Sciences, Arizona State University, Tempe, AZ 85287-4501, USA; (B.K.D.); (C.I.S.); (G.W.S.); (S.A.A.)
- Molecular and Cellular Biology Graduate Program, School of Life Sciences, Tempe, AZ 85287 4501, USA
| | - Sofia A. Acosta
- School of Life Sciences, Arizona State University, Tempe, AZ 85287-4501, USA; (B.K.D.); (C.I.S.); (G.W.S.); (S.A.A.)
- Molecular and Cellular Biology Graduate Program, School of Life Sciences, Tempe, AZ 85287 4501, USA
| | - Jeanne Wilson-Rawls
- School of Life Sciences, Arizona State University, Tempe, AZ 85287-4501, USA; (B.K.D.); (C.I.S.); (G.W.S.); (S.A.A.)
| |
Collapse
|
15
|
Chapoy Villanueva H, Sung JH, Stevens JA, Zhang MJ, Nelson PM, Denduluri LS, Feng F, O'Connell TD, Townsend D, Liu JC. Distinct effects of cardiac mitochondrial calcium uniporter inactivation via EMRE deletion in the short and long term. J Mol Cell Cardiol 2023; 181:33-45. [PMID: 37230379 PMCID: PMC10524693 DOI: 10.1016/j.yjmcc.2023.05.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 05/13/2023] [Accepted: 05/18/2023] [Indexed: 05/27/2023]
Abstract
Transport of Ca2+ into mitochondria is thought to stimulate the production of ATP, a critical process in the heart's fight or flight response, but excess Ca2+ can trigger cell death. The mitochondrial Ca2+ uniporter complex is the primary route of Ca2+ transport into mitochondria, in which the channel-forming protein MCU and the regulatory protein EMRE are essential for activity. In previous studies, chronic Mcu or Emre deletion differed from acute cardiac Mcu deletion in response to adrenergic stimulation and ischemia/reperfusion (I/R) injury, despite equivalent inactivation of rapid mitochondrial Ca2+ uptake. To explore this discrepancy between chronic and acute loss of uniporter activity, we compared short-term and long-term Emre deletion using a novel conditional cardiac-specific, tamoxifen-inducible mouse model. After short-term Emre deletion (3 weeks post-tamoxifen) in adult mice, cardiac mitochondria were unable to take up Ca2+, had lower basal mitochondrial Ca2+ levels, and displayed attenuated Ca2+-induced ATP production and mPTP opening. Moreover, short-term EMRE loss blunted cardiac response to adrenergic stimulation and improved maintenance of cardiac function in an ex vivo I/R model. We then tested whether the long-term absence of EMRE (3 months post-tamoxifen) in adulthood would lead to distinct outcomes. After long-term Emre deletion, mitochondrial Ca2+ handling and function, as well as cardiac response to adrenergic stimulation, were similarly impaired as in short-term deletion. Interestingly, however, protection from I/R injury was lost in the long-term. These data suggest that several months without uniporter function are insufficient to restore bioenergetic response but are sufficient to restore susceptibility to I/R.
Collapse
Affiliation(s)
- Hector Chapoy Villanueva
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Jae Hwi Sung
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Jackie A Stevens
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Michael J Zhang
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA; Cardiovascular Division, Department of Medicine, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Peyton M Nelson
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Lalitha S Denduluri
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Feng Feng
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Timothy D O'Connell
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - DeWayne Townsend
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Julia C Liu
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA.
| |
Collapse
|
16
|
Hu C, Yan L, Li P, Yu Y. Identification of calcium metabolism related score associated with the poor outcome in papillary thyroid carcinoma. Front Oncol 2023; 13:1108773. [PMID: 37056339 PMCID: PMC10086330 DOI: 10.3389/fonc.2023.1108773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 03/09/2023] [Indexed: 03/30/2023] Open
Abstract
IntroductionPapillary thyroid carcinoma is a type of thyroid cancer that exhibits significant variability in prognosis. Extensive research indicates that the impaired signaling of 1,25(OH)2D3-VDR may be a crucial factor in the development and progression of PTC.MethodsTo investigate this further, Integrated analysis mRNA expression information from The Cancer Genome Atlas and GEO, we compared gene expression in cancer and normal tissues and identified differentially expressed genes (DEGs). Through this analysis, we identified DEGs and calculated risk estimates for seven genetic markers.ResultsSubsequently, we constructed predictive models using LASSO-Cox regression to test the predictive value of these markers. Our results revealed that 64 calcium metabolism-related genes showed significant differences between tumor and normal tissues. Ten of the identified DEGs were significantly associated with overall survival, indicating their potential role in disease progression. Using the average risk score for the seven genetic markers, we divided patients into high- and low-risk groups. We found that patients in the low-risk group had significantly better overall survival than those in the high-risk group, highlighting the importance of these genetic markers in predicting prognosis. Further analysis using Cox regression demonstrated that the risk levels had independent predictive power. Additionally, we conducted functional analysis of the identified genetic markers, which showed significant differences in immune status between the two patient groups. We also investigated the effect of these calcium metabolism-related genes on thyroid cancer biological functions, immune microenvironment, and drug resistance.DiscussionOur findings provide evidence of a novel genetic signature associated with calcium metabolism, which can predict prognosis in patients with PTC. These results may have significant implications for the development of new diagnostic and therapeutic approaches to improve outcomes for PTC patients.
Collapse
Affiliation(s)
- Chuanxiang Hu
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
| | - Lijuan Yan
- State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, Collaborative Innovation Center for Biotherapy, Nankai University, Tianjin, China
- Tianjin Key Laboratory of Protein Sciences, National Demonstration Center for Experimental Biology Education and College of Life Sciences, Nankai University, Tianjin, China
| | - Peng Li
- State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Center of Tianjin for Medical Epigenetics, Collaborative Innovation Center for Biotherapy, Nankai University, Tianjin, China
- Tianjin Key Laboratory of Protein Sciences, National Demonstration Center for Experimental Biology Education and College of Life Sciences, Nankai University, Tianjin, China
- *Correspondence: Peng Li, ; Yang Yu,
| | - Yang Yu
- Department of Thyroid and Neck Tumor, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- *Correspondence: Peng Li, ; Yang Yu,
| |
Collapse
|
17
|
Dubuisson N, Versele R, Planchon C, Selvais CM, Noel L, Abou-Samra M, Davis-López de Carrizosa MA. Histological Methods to Assess Skeletal Muscle Degeneration and Regeneration in Duchenne Muscular Dystrophy. Int J Mol Sci 2022; 23:16080. [PMID: 36555721 PMCID: PMC9786356 DOI: 10.3390/ijms232416080] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/09/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive disease caused by the loss of function of the protein dystrophin. This protein contributes to the stabilisation of striated cells during contraction, as it anchors the cytoskeleton with components of the extracellular matrix through the dystrophin-associated protein complex (DAPC). Moreover, absence of the functional protein affects the expression and function of proteins within the DAPC, leading to molecular events responsible for myofibre damage, muscle weakening, disability and, eventually, premature death. Presently, there is no cure for DMD, but different treatments help manage some of the symptoms. Advances in genetic and exon-skipping therapies are the most promising intervention, the safety and efficiency of which are tested in animal models. In addition to in vivo functional tests, ex vivo molecular evaluation aids assess to what extent the therapy has contributed to the regenerative process. In this regard, the later advances in microscopy and image acquisition systems and the current expansion of antibodies for immunohistological evaluation together with the development of different spectrum fluorescent dyes have made histology a crucial tool. Nevertheless, the complexity of the molecular events that take place in dystrophic muscles, together with the rise of a multitude of markers for each of the phases of the process, makes the histological assessment a challenging task. Therefore, here, we summarise and explain the rationale behind different histological techniques used in the literature to assess degeneration and regeneration in the field of dystrophinopathies, focusing especially on those related to DMD.
Collapse
Affiliation(s)
- Nicolas Dubuisson
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium
- Neuromuscular Reference Center, Cliniques Universitaires Saint-Luc (CUSL), Avenue Hippocrate 10, 1200 Brussels, Belgium
| | - Romain Versele
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium
| | - Chloé Planchon
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium
| | - Camille M. Selvais
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium
| | - Laurence Noel
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium
| | - Michel Abou-Samra
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium
| | - María A. Davis-López de Carrizosa
- Endocrinology, Diabetes and Nutrition Unit, Institute of Experimental and Clinical Research, Medical Sector, Université Catholique de Louvain (UCLouvain), Avenue Hippocrate 55, 1200 Brussels, Belgium
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, 41012 Seville, Spain
| |
Collapse
|
18
|
A Large-Scale High-Throughput Screen for Modulators of SERCA Activity. Biomolecules 2022; 12:biom12121789. [PMID: 36551215 PMCID: PMC9776381 DOI: 10.3390/biom12121789] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/21/2022] [Accepted: 11/25/2022] [Indexed: 12/03/2022] Open
Abstract
The sarco/endoplasmic reticulum Ca-ATPase (SERCA) is a P-type ion pump that transports Ca2+ from the cytosol into the endoplasmic/sarcoplasmic reticulum (ER/SR) in most mammalian cells. It is critically important in muscle, facilitating relaxation and enabling subsequent contraction. Increasing SERCA expression or specific activity can alleviate muscle dysfunction, most notably in the heart, and we seek to develop small-molecule drug candidates that activate SERCA. Therefore, we adapted an NADH-coupled assay, measuring Ca-dependent ATPase activity of SERCA, to high-throughput screening (HTS) format, and screened a 46,000-compound library of diverse chemical scaffolds. This HTS platform yielded numerous hits that reproducibly alter SERCA Ca-ATPase activity, with few false positives. The top 19 activating hits were further tested for effects on both Ca-ATPase and Ca2+ transport, in both cardiac and skeletal SR. Nearly all hits increased Ca2+ uptake in both cardiac and skeletal SR, with some showing isoform specificity. Furthermore, dual analysis of both activities identified compounds with a range of effects on Ca2+-uptake and ATPase, which fit into distinct classifications. Further study will be needed to identify which classifications are best suited for therapeutic use. These results reinforce the need for robust secondary assays and criteria for selection of lead compounds, before undergoing HTS on a larger scale.
Collapse
|
19
|
Hu M, Ge MR, Li HX, Zhang B, Li G. Identification of DAPK1 as an autophagy-related biomarker for myotonic dystrophy type 1. Front Genet 2022; 13:1022640. [PMID: 36338967 PMCID: PMC9634726 DOI: 10.3389/fgene.2022.1022640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 10/07/2022] [Indexed: 11/24/2022] Open
Abstract
Myotonic dystrophy type I (DM1), a CTG repeat expansion hereditary disorder, is primarily characterized by myotonia. Several studies have reported that abnormal autophagy pathway has a close relationship with DM1. However, the underlying key regulatory molecules dictating autophagy disturbance still remains elusive. Previous studies mainly focused on finding targeted therapies for DM1, but the clinical heterogeneity of the DM1 is rarely addressed. Herein, to identify potential regulator genes related to autophagy and cross-correlation among clinical symptoms, we performed weighted gene co-expression network analysis (WGCNA) to construct the co-expression network and screened out 7 core autophagy-related genes (DAPK1, KLHL4, ERBB3, SESN3, ATF4, MEG3, and COL1A1) by overlapping within differentially expressed genes (DEG), cytoHubba, gene significance (GS) and module membership (MM) score. Meanwhile, we here analyzed autophagy-related molecular subtypes of DM1 in relation to the clinical phenotype. Our results show that three genes (DAPK1, SESN3, and MEG3) contribute to distinguish these two molecular subtypes of DM1. We then develop an analysis of RNA-seq data from six human skin fibroblasts (3 DM1, 3 healthy donors). Intriguingly, of the 7 hallmark genes obtained, DAPK1 is the only confirmed gene, and finally identified in vitro by RT-PCR. Furthermore, we assessed the DAPK1 accuracy diagnosis of DM1 by plotting a receiver operating characteristic curve (ROC) (AUC = 0.965). In this study, we first validated autophagy status of DM1 individuals exhibits a clearly heterogeneity. Our study identified and validated DAPK1 serve as a novel autophagy-related biomarker that correlate with the progression of DM1.
Collapse
Affiliation(s)
| | | | | | - Bei Zhang
- *Correspondence: Bei Zhang, ; Gang Li,
| | - Gang Li
- *Correspondence: Bei Zhang, ; Gang Li,
| |
Collapse
|
20
|
Gosselin MRF, Mournetas V, Borczyk M, Verma S, Occhipinti A, Róg J, Bozycki L, Korostynski M, Robson SC, Angione C, Pinset C, Gorecki DC. Loss of full-length dystrophin expression results in major cell-autonomous abnormalities in proliferating myoblasts. eLife 2022; 11:e75521. [PMID: 36164827 PMCID: PMC9514850 DOI: 10.7554/elife.75521] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 09/02/2022] [Indexed: 12/05/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) affects myofibers and muscle stem cells, causing progressive muscle degeneration and repair defects. It was unknown whether dystrophic myoblasts-the effector cells of muscle growth and regeneration-are affected. Using transcriptomic, genome-scale metabolic modelling and functional analyses, we demonstrate, for the first time, convergent abnormalities in primary mouse and human dystrophic myoblasts. In Dmdmdx myoblasts lacking full-length dystrophin, the expression of 170 genes was significantly altered. Myod1 and key genes controlled by MyoD (Myog, Mymk, Mymx, epigenetic regulators, ECM interactors, calcium signalling and fibrosis genes) were significantly downregulated. Gene ontology analysis indicated enrichment in genes involved in muscle development and function. Functionally, we found increased myoblast proliferation, reduced chemotaxis and accelerated differentiation, which are all essential for myoregeneration. The defects were caused by the loss of expression of full-length dystrophin, as similar and not exacerbated alterations were observed in dystrophin-null Dmdmdx-βgeo myoblasts. Corresponding abnormalities were identified in human DMD primary myoblasts and a dystrophic mouse muscle cell line, confirming the cross-species and cell-autonomous nature of these defects. The genome-scale metabolic analysis in human DMD myoblasts showed alterations in the rate of glycolysis/gluconeogenesis, leukotriene metabolism, and mitochondrial beta-oxidation of various fatty acids. These results reveal the disease continuum: DMD defects in satellite cells, the myoblast dysfunction affecting muscle regeneration, which is insufficient to counteract muscle loss due to myofiber instability. Contrary to the established belief, our data demonstrate that DMD abnormalities occur in myoblasts, making these cells a novel therapeutic target for the treatment of this lethal disease.
Collapse
Affiliation(s)
- Maxime RF Gosselin
- School of Pharmacy and Biomedical Sciences, University of PortsmouthPortsmouthUnited Kingdom
| | | | - Malgorzata Borczyk
- Laboratory of Pharmacogenomics, Maj Institute of Pharmacology PASKrakowPoland
| | - Suraj Verma
- School of Computing, Engineering and Digital Technologies, Teesside UniversityMiddlesbroughUnited Kingdom
| | - Annalisa Occhipinti
- School of Computing, Engineering and Digital Technologies, Teesside UniversityMiddlesbroughUnited Kingdom
| | - Justyna Róg
- School of Pharmacy and Biomedical Sciences, University of PortsmouthPortsmouthUnited Kingdom
- Laboratory of Cellular Metabolism, Nencki Institute of Experimental BiologyWarsawPoland
| | - Lukasz Bozycki
- School of Pharmacy and Biomedical Sciences, University of PortsmouthPortsmouthUnited Kingdom
- Laboratory of Cellular Metabolism, Nencki Institute of Experimental BiologyWarsawPoland
| | - Michal Korostynski
- Laboratory of Pharmacogenomics, Maj Institute of Pharmacology PASKrakowPoland
| | - Samuel C Robson
- School of Pharmacy and Biomedical Sciences, University of PortsmouthPortsmouthUnited Kingdom
- Centre for Enzyme Innovation, University of PortsmouthPortsmouthUnited Kingdom
| | - Claudio Angione
- School of Computing, Engineering and Digital Technologies, Teesside UniversityMiddlesbroughUnited Kingdom
| | | | - Dariusz C Gorecki
- School of Pharmacy and Biomedical Sciences, University of PortsmouthPortsmouthUnited Kingdom
| |
Collapse
|
21
|
Boulinguiez A, Duhem C, Mayeuf-Louchart A, Pourcet B, Sebti Y, Kondratska K, Montel V, Delhaye S, Thorel Q, Beauchamp J, Hebras A, Gimenez M, Couvelaere M, Zecchin M, Ferri L, Prevarskaya N, Forand A, Gentil C, Ohana J, Piétri-Rouxel F, Bastide B, Staels B, Duez H, Lancel S. NR1D1 controls skeletal muscle calcium homeostasis through myoregulin repression. JCI Insight 2022; 7:153584. [PMID: 35917173 PMCID: PMC9536258 DOI: 10.1172/jci.insight.153584] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 07/27/2022] [Indexed: 11/17/2022] Open
Abstract
The sarcoplasmic reticulum (SR) plays an important role in calcium homeostasis. SR calcium mishandling is described in pathological conditions such as myopathies. Here, we investigated whether the nuclear receptor subfamily 1 group D member (NR1D1, also called REV-ERBα) regulates skeletal muscle SR calcium homeostasis. Our data demonstrate that NR1D1 deficiency in mice impairs SERCA-dependent SR calcium uptake. NR1D1 acts on calcium homeostasis by repressing the SERCA inhibitor myoregulin through direct binding to its promoter. Restoration of myoregulin counteracts the effects of NR1D1 overexpression on SR calcium content. Interestingly, myoblasts from Duchenne myopathy patients display lower NR1D1 expression, whereas pharmacological NR1D1 activation ameliorates SR calcium homeostasis, and improves muscle structure and function in dystrophic mdx/Utr+/- mice. Our findings demonstrate that NR1D1 regulates muscle SR calcium homeostasis, pointing to its therapeutic interest for mitigating myopathy.
Collapse
Affiliation(s)
- Alexis Boulinguiez
- U1011-EGID, University Lille, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Christian Duhem
- U1011-EGID, University Lille, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Alicia Mayeuf-Louchart
- U1011-EGID, University Lille, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Benoit Pourcet
- U1011-EGID, University Lille, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Yasmine Sebti
- U1011-EGID, University Lille, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Kateryna Kondratska
- U1003 - PHYCEL - Physiologie Cellulaire, University Lille, Inserm,, Villeneuve d'Ascq, France
| | - Valérie Montel
- URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, Univ. Lille, Univ. Artois, Univ. Littoral Côte d'Opale,, Lille, France
| | - Stéphane Delhaye
- U1011-EGID, University Lille, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Quentin Thorel
- U1011-EGID, University Lille, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Justine Beauchamp
- U1011-EGID, University Lille, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Aurore Hebras
- U1011-EGID, University Lille, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Marion Gimenez
- U1011-EGID, University Lille, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Marie Couvelaere
- U1011-EGID, University Lille, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Mathilde Zecchin
- U1011-EGID, University Lille, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Lise Ferri
- U1011-EGID, University Lille, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Natalia Prevarskaya
- U1003 - PHYCEL - Physiologie Cellulaire, University Lille, Inserm, Villeneuve d'Ascq, France
| | - Anne Forand
- INSERM U845, Université Paris Descartes, Paris, France
| | | | - Jessica Ohana
- MyoLine, Sorbonne Université-UMRS974-Inserm-Institut de Myologie, Paris, France
| | | | - Bruno Bastide
- URePSSS - Unité de Recherche Pluridisciplinaire Sport Santé Société, Univ. Lille, Univ. Artois, Univ. Littoral Côte d'Opale,, Lille, France
| | - Bart Staels
- U1011-EGID, University Lille, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Helene Duez
- U1011-EGID, University Lille, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Steve Lancel
- U1011-EGID, University Lille, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| |
Collapse
|
22
|
Pingel J, Vandenrijt J, Kampmann ML, Andersen JD. Altered gene expression levels of genes related to muscle function in adults with cerebral palsy. Tissue Cell 2022; 76:101744. [DOI: 10.1016/j.tice.2022.101744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 01/06/2022] [Accepted: 01/25/2022] [Indexed: 11/30/2022]
|
23
|
Vesga-Castro C, Aldazabal J, Vallejo-Illarramendi A, Paredes J. Contractile force assessment methods for in vitro skeletal muscle tissues. eLife 2022; 11:e77204. [PMID: 35604384 PMCID: PMC9126583 DOI: 10.7554/elife.77204] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 04/27/2022] [Indexed: 02/06/2023] Open
Abstract
Over the last few years, there has been growing interest in measuring the contractile force (CF) of engineered muscle tissues to evaluate their functionality. However, there are still no standards available for selecting the most suitable experimental platform, measuring system, culture protocol, or stimulation patterns. Consequently, the high variability of published data hinders any comparison between different studies. We have identified that cantilever deflection, post deflection, and force transducers are the most commonly used configurations for CF assessment in 2D and 3D models. Additionally, we have discussed the most relevant emerging technologies that would greatly complement CF evaluation with intracellular and localized analysis. This review provides a comprehensive analysis of the most significant advances in CF evaluation and its critical parameters. In order to compare contractile performance across experimental platforms, we have used the specific force (sF, kN/m2), CF normalized to the calculated cross-sectional area (CSA). However, this parameter presents a high variability throughout the different studies, which indicates the need to identify additional parameters and complementary analysis suitable for proper comparison. We propose that future contractility studies in skeletal muscle constructs report detailed information about construct size, contractile area, maturity level, sarcomere length, and, ideally, the tetanus-to-twitch ratio. These studies will hopefully shed light on the relative impact of these variables on muscle force performance of engineered muscle constructs. Prospective advances in muscle tissue engineering, particularly in muscle disease models, will require a joint effort to develop standardized methodologies for assessing CF of engineered muscle tissues.
Collapse
Affiliation(s)
- Camila Vesga-Castro
- University of Navarra, Tecnun School of Engineering, Manuel de LardizábalSan SebastianSpain
- University of Navarra, Biomedical Engineering Center, Campus UniversitarioPamplonaSpain
- Group of Neurosciences, Department of Pediatrics, UPV/EHU, Hospital Donostia - IIS BiodonostiaSan SebastianSpain
| | - Javier Aldazabal
- University of Navarra, Tecnun School of Engineering, Manuel de LardizábalSan SebastianSpain
- University of Navarra, Biomedical Engineering Center, Campus UniversitarioPamplonaSpain
| | - Ainara Vallejo-Illarramendi
- Group of Neurosciences, Department of Pediatrics, UPV/EHU, Hospital Donostia - IIS BiodonostiaSan SebastianSpain
- CIBERNED, Instituto de Salud Carlos III, Ministry of Science, Innovation, and UniversitiesMadridSpain
| | - Jacobo Paredes
- University of Navarra, Tecnun School of Engineering, Manuel de LardizábalSan SebastianSpain
- University of Navarra, Biomedical Engineering Center, Campus UniversitarioPamplonaSpain
| |
Collapse
|
24
|
Lasa-Elgarresta J, Mosqueira-Martín L, González-Imaz K, Marco-Moreno P, Gerenu G, Mamchaoui K, Mouly V, López de Munain A, Vallejo-Illarramendi A. Targeting the Ubiquitin-Proteasome System in Limb-Girdle Muscular Dystrophy With CAPN3 Mutations. Front Cell Dev Biol 2022; 10:822563. [PMID: 35309930 PMCID: PMC8924035 DOI: 10.3389/fcell.2022.822563] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 02/03/2022] [Indexed: 12/26/2022] Open
Abstract
LGMDR1 is caused by mutations in the CAPN3 gene that encodes calpain 3 (CAPN3), a non-lysosomal cysteine protease necessary for proper muscle function. Our previous findings show that CAPN3 deficiency leads to reduced SERCA levels through increased protein degradation. This work investigates the potential contribution of the ubiquitin-proteasome pathway to increased SERCA degradation in LGMDR1. Consistent with our previous results, we observed that CAPN3-deficient human myotubes exhibit reduced SERCA protein levels and high cytosolic calcium concentration. Treatment with the proteasome inhibitor bortezomib (Velcade) increased SERCA2 protein levels and normalized intracellular calcium levels in CAPN3-deficient myotubes. Moreover, bortezomib was able to recover mutated CAPN3 protein in a patient carrying R289W and R546L missense mutations. We found that CAPN3 knockout mice (C3KO) presented SERCA deficits in skeletal muscle in the early stages of the disease, prior to the manifestation of muscle deficits. However, treatment with bortezomib (0.8 mg/kg every 72 h) for 3 weeks did not rescue SERCA levels. No change in muscle proteasome activity was observed in bortezomib-treated animals, suggesting that higher bortezomib doses are needed to rescue SERCA levels in this model. Overall, our results lay the foundation for exploring inhibition of the ubiquitin-proteasome as a new therapeutic target to treat LGMDR1 patients. Moreover, patients carrying missense mutations in CAPN3 and presumably other genes may benefit from proteasome inhibition by rescuing mutant protein levels. Further studies in suitable models will be necessary to demonstrate the therapeutic efficacy of proteasome inhibition for different missense mutations.
Collapse
Affiliation(s)
- Jaione Lasa-Elgarresta
- Group of Neuroscience, Departments of Pediatrics and Neuroscience, Faculty of Medicine and Nursing, Hospital Donostia, UPV/EHU, San Sebastian, Spain.,IIS Biodonostia, Neurosciences Area, Group of Neuromuscular Diseases, San Sebastian, Spain
| | - Laura Mosqueira-Martín
- Group of Neuroscience, Departments of Pediatrics and Neuroscience, Faculty of Medicine and Nursing, Hospital Donostia, UPV/EHU, San Sebastian, Spain.,IIS Biodonostia, Neurosciences Area, Group of Neuromuscular Diseases, San Sebastian, Spain
| | - Klaudia González-Imaz
- IIS Biodonostia, Neurosciences Area, Group of Neuromuscular Diseases, San Sebastian, Spain
| | - Pablo Marco-Moreno
- IIS Biodonostia, Neurosciences Area, Group of Neuromuscular Diseases, San Sebastian, Spain
| | - Gorka Gerenu
- IIS Biodonostia, Neurosciences Area, Group of Neuromuscular Diseases, San Sebastian, Spain.,CIBERNED, Instituto de Salud Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain.,Department of Physiology, Faculty of Medicine and Nursing, UPV/EHU, Leioa, Spain
| | - Kamel Mamchaoui
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Vincent Mouly
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Adolfo López de Munain
- Group of Neuroscience, Departments of Pediatrics and Neuroscience, Faculty of Medicine and Nursing, Hospital Donostia, UPV/EHU, San Sebastian, Spain.,IIS Biodonostia, Neurosciences Area, Group of Neuromuscular Diseases, San Sebastian, Spain.,CIBERNED, Instituto de Salud Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain
| | - Ainara Vallejo-Illarramendi
- Group of Neuroscience, Departments of Pediatrics and Neuroscience, Faculty of Medicine and Nursing, Hospital Donostia, UPV/EHU, San Sebastian, Spain.,IIS Biodonostia, Neurosciences Area, Group of Neuromuscular Diseases, San Sebastian, Spain.,CIBERNED, Instituto de Salud Carlos III, Ministry of Economy and Competitiveness, Madrid, Spain
| |
Collapse
|
25
|
Cai B, Ma M, Zhang J, Wang Z, Kong S, Zhou Z, Lian L, Zhang J, Li J, Wang Y, Li H, Zhang X, Nie Q. LncEDCH1 improves mitochondrial function to reduce muscle atrophy by interacting with SERCA2. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 27:319-334. [PMID: 35024244 PMCID: PMC8717430 DOI: 10.1016/j.omtn.2021.12.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 12/07/2021] [Indexed: 12/25/2022]
Abstract
Skeletal muscle is a regulator of the body's energy expenditure and metabolism. Abnormal regulation of skeletal muscle-specific genes leads to various muscle diseases. Long non-coding RNAs (lncRNAs) have been demonstrated to play important roles in muscle growth and muscle atrophy. To explore the potential function of muscle-associated lncRNA, we analyzed our previous RNA-sequencing data and selected the lncRNA (LncEDCH1) as the research object. In this study, we report that LncEDCH1 is specifically enriched in skeletal muscle, and its transcriptional activity is positively regulated by transcription factor SP1. LncEDCH1 regulates myoblast proliferation and differentiation in vitro. In vivo, LncEDCH1 reduces intramuscular fat deposition, activates slow-twitch muscle phenotype, and inhibits muscle atrophy. Mechanistically, LncEDCH1 binds to sarcoplasmic/ER calcium ATPase 2 (SERCA2) protein to enhance SERCA2 protein stability and increase SERCA2 activity. Meanwhile, LncEDCH1 improves mitochondrial efficiency possibly through a SERCA2-mediated activation of the AMPK pathway. Our findings provide a strategy for using LncEDCH1 as an effective regulator for the treatment of muscle atrophy and energy metabolism.
Collapse
Affiliation(s)
- Bolin Cai
- Lingnan Guangdong Laboratory of Modern Agriculture & State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, Guangdong 510642, China
| | - Manting Ma
- Lingnan Guangdong Laboratory of Modern Agriculture & State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, Guangdong 510642, China
| | - Jing Zhang
- Lingnan Guangdong Laboratory of Modern Agriculture & State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, Guangdong 510642, China
| | - Zhijun Wang
- Lingnan Guangdong Laboratory of Modern Agriculture & State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, Guangdong 510642, China
| | - Shaofen Kong
- Lingnan Guangdong Laboratory of Modern Agriculture & State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, Guangdong 510642, China
| | - Zhen Zhou
- Lingnan Guangdong Laboratory of Modern Agriculture & State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, Guangdong 510642, China
| | - Ling Lian
- National Engineering Laboratory for Animal Breeding and MOA Key Laboratory of Animal Genetics and Breeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Jiannan Zhang
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, China
| | - Juan Li
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, China
| | - Yajun Wang
- Key Laboratory of Bio-Resources and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610065, China
| | - Hongmei Li
- Lingnan Guangdong Laboratory of Modern Agriculture & State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, Guangdong 510642, China
| | - Xiquan Zhang
- Lingnan Guangdong Laboratory of Modern Agriculture & State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, Guangdong 510642, China
| | - Qinghua Nie
- Lingnan Guangdong Laboratory of Modern Agriculture & State Key Laboratory for Conservation and Utilization of Subtropical Agro-bioresources, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong 510642, China.,Guangdong Provincial Key Lab of Agro-Animal Genomics and Molecular Breeding, Key Laboratory of Chicken Genetics, Breeding and Reproduction, Ministry of Agriculture, Guangzhou, Guangdong 510642, China
| |
Collapse
|
26
|
Bhardwaj G, Penniman CM, Klaus K, Weatherford ET, Pan H, Dreyfuss JM, Nair KS, Kahn CR, O’Neill BT. Transcriptomic Regulation of Muscle Mitochondria and Calcium Signaling by Insulin/IGF-1 Receptors Depends on FoxO Transcription Factors. Front Physiol 2022; 12:779121. [PMID: 35185597 PMCID: PMC8855073 DOI: 10.3389/fphys.2021.779121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 12/23/2021] [Indexed: 02/02/2023] Open
Abstract
Insulin and IGF-1, acting through the insulin receptor (IR) and IGF-1 receptor (IGF1R), maintain muscle mass and mitochondrial function, at least part of which occurs via their action to regulate gene expression. Here, we show that while muscle-specific deletion of IR or IGF1R individually results in only modest changes in the muscle transcriptome, combined deletion of IR/IGF1R (MIGIRKO) altered > 3000 genes, including genes involved in mitochondrial dysfunction, fibrosis, cardiac hypertrophy, and pathways related to estrogen receptor, protein kinase A (PKA), and calcium signaling. Functionally, this was associated with decreased mitochondrial respiration and increased ROS production in MIGIRKO muscle. To determine the role of FoxOs in these changes, we performed RNA-Seq on mice with muscle-specific deletion of FoxO1/3/4 (M-FoxO TKO) or combined deletion of IR, IGF1R, and FoxO1/3/4 in a muscle quintuple knockout (M-QKO). This revealed that among IR/IGF1R regulated genes, >97% were FoxO-dependent, and their expression was normalized in M-FoxO TKO and M-QKO muscle. FoxO-dependent genes were related to oxidative phosphorylation, inflammatory signaling, and TCA cycle. Metabolomic analysis showed accumulation of TCA cycle metabolites in MIGIRKO, which was reversed in M-QKO muscle. Likewise, calcium signaling genes involved in PKA signaling and sarcoplasmic reticulum calcium homeostasis were markedly altered in MIGIRKO muscle but normalized in M-QKO. Thus, combined loss of insulin and IGF-1 action in muscle transcriptionally alters mitochondrial function and multiple regulatory and signaling pathways, and these changes are mediated by FoxO transcription factors.
Collapse
Affiliation(s)
- Gourav Bhardwaj
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Christie M. Penniman
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Katherine Klaus
- Division of Endocrinology and Metabolism, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - Eric T. Weatherford
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Hui Pan
- Bioinformatics and Biostatistics Core, Joslin Diabetes Center, Harvard Medical School, Boston, MA, United States
| | - Jonathan M. Dreyfuss
- Bioinformatics and Biostatistics Core, Joslin Diabetes Center, Harvard Medical School, Boston, MA, United States
| | - K. Sreekumaran Nair
- Division of Endocrinology and Metabolism, Mayo Clinic College of Medicine and Science, Rochester, MN, United States
| | - C. Ronald Kahn
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, United States
| | - Brian T. O’Neill
- Fraternal Order of Eagles Diabetes Research Center and Division of Endocrinology and Metabolism, Roy J. and Lucille A. Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Veterans Affairs Health Care System, Iowa City, IA, United States
| |
Collapse
|
27
|
Talmon M, Massara E, Pruonto G, Quaregna M, Boccafoschi F, Riva B, Fresu LG. Characterization of a functional Ca2+ toolkit in urine-derived stem cells and derived skeletal muscle cells. Cell Calcium 2022; 103:102548. [DOI: 10.1016/j.ceca.2022.102548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 01/11/2022] [Accepted: 02/02/2022] [Indexed: 11/16/2022]
|
28
|
Meyer P, Notarnicola C, Meli AC, Matecki S, Hugon G, Salvador J, Khalil M, Féasson L, Cances C, Cottalorda J, Desguerre I, Cuisset JM, Sabouraud P, Lacampagne A, Chevassus H, Rivier F, Carnac G. Skeletal Ryanodine Receptors Are Involved in Impaired Myogenic Differentiation in Duchenne Muscular Dystrophy Patients. Int J Mol Sci 2021; 22:12985. [PMID: 34884796 PMCID: PMC8657486 DOI: 10.3390/ijms222312985] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/24/2021] [Accepted: 11/29/2021] [Indexed: 11/17/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is characterized by progressive muscle wasting following repeated muscle damage and inadequate regeneration. Impaired myogenesis and differentiation play a major role in DMD as well as intracellular calcium (Ca2+) mishandling. Ca2+ release from the sarcoplasmic reticulum is mostly mediated by the type 1 ryanodine receptor (RYR1) that is required for skeletal muscle differentiation in animals. The study objective was to determine whether altered RYR1-mediated Ca2+ release contributes to myogenic differentiation impairment in DMD patients. The comparison of primary cultured myoblasts from six boys with DMD and five healthy controls highlighted delayed myoblast differentiation in DMD. Silencing RYR1 expression using specific si-RNA in a healthy control induced a similar delayed differentiation. In DMD myotubes, resting intracellular Ca2+ concentration was increased, but RYR1-mediated Ca2+ release was not changed compared with control myotubes. Incubation with the RYR-calstabin interaction stabilizer S107 decreased resting Ca2+ concentration in DMD myotubes to control values and improved calstabin1 binding to the RYR1 complex. S107 also improved myogenic differentiation in DMD. Furthermore, intracellular Ca2+ concentration was correlated with endomysial fibrosis, which is the only myopathologic parameter associated with poor motor outcome in patients with DMD. This suggested a potential relationship between RYR1 dysfunction and motor impairment. Our study highlights RYR1-mediated Ca2+ leakage in human DMD myotubes and its key role in myogenic differentiation impairment. RYR1 stabilization may be an interesting adjunctive therapeutic strategy in DMD.
Collapse
Affiliation(s)
- Pierre Meyer
- PhyMedExp, University of Montpellier, Inserm, CNRS, 34295 Montpellier, France; (C.N.); (A.C.M.); (S.M.); (G.H.); (J.S.); (A.L.); (F.R.); (G.C.)
- Reference Centre for Neuromuscular Diseases AOC, Clinical Investigation Centre, Pediatric Neurology Department, Montpellier University Hospital, 34000 Montpellier, France
| | - Cécile Notarnicola
- PhyMedExp, University of Montpellier, Inserm, CNRS, 34295 Montpellier, France; (C.N.); (A.C.M.); (S.M.); (G.H.); (J.S.); (A.L.); (F.R.); (G.C.)
| | - Albano C. Meli
- PhyMedExp, University of Montpellier, Inserm, CNRS, 34295 Montpellier, France; (C.N.); (A.C.M.); (S.M.); (G.H.); (J.S.); (A.L.); (F.R.); (G.C.)
| | - Stefan Matecki
- PhyMedExp, University of Montpellier, Inserm, CNRS, 34295 Montpellier, France; (C.N.); (A.C.M.); (S.M.); (G.H.); (J.S.); (A.L.); (F.R.); (G.C.)
| | - Gérald Hugon
- PhyMedExp, University of Montpellier, Inserm, CNRS, 34295 Montpellier, France; (C.N.); (A.C.M.); (S.M.); (G.H.); (J.S.); (A.L.); (F.R.); (G.C.)
| | - Jérémy Salvador
- PhyMedExp, University of Montpellier, Inserm, CNRS, 34295 Montpellier, France; (C.N.); (A.C.M.); (S.M.); (G.H.); (J.S.); (A.L.); (F.R.); (G.C.)
| | - Mirna Khalil
- Clinical Investigation Center, Montpellier University Hospital, 34000 Montpellier, France; (M.K.); (H.C.)
| | - Léonard Féasson
- Myology Unit, Reference Center for Neuromuscular Diseases Euro-NmD, Inter-University Laboratory of Human Movement Sciences—EA7424, University Hospital of Saint-Etienne, 42055 Saint-Etienne, France;
| | - Claude Cances
- Reference Center for Neuromuscular Diseases AOC, Pediatric Neurology Department, Toulouse University Hospital, 3100 Toulouse, France;
- Pediatric Clinical Research Unit, Pediatric Multi-thematic Module CIC 1436, Toulouse Children’s Hospital, 31300 Toulouse, France
| | - Jérôme Cottalorda
- Pediatric Orthopedic and Plastic Surgery Department, Montpellier University Hospital, 34295 Montpellier, France;
| | - Isabelle Desguerre
- Reference Center for Neuromuscular Diseases Paris Nord-Ile-de-France-Est, Pediatric Neurology Department, Necker Enfant Malades University Hospital, Assistance Publique des Hôpitaux de Paris Centre, Paris University, 75019 Paris, France;
| | - Jean-Marie Cuisset
- Reference Center for Neuromuscular Diseases Nord-Ile-de-France-Est, Pediatric Neurology Department, Lille University Hospital, 59000 Lille, France;
| | - Pascal Sabouraud
- Reference Center for Neuromuscular Diseases Nord-Ile-de-France-Est, Pediatric Neurology Department, Reims University Hospital, 51100 Reims, France;
| | - Alain Lacampagne
- PhyMedExp, University of Montpellier, Inserm, CNRS, 34295 Montpellier, France; (C.N.); (A.C.M.); (S.M.); (G.H.); (J.S.); (A.L.); (F.R.); (G.C.)
| | - Hugues Chevassus
- Clinical Investigation Center, Montpellier University Hospital, 34000 Montpellier, France; (M.K.); (H.C.)
| | - François Rivier
- PhyMedExp, University of Montpellier, Inserm, CNRS, 34295 Montpellier, France; (C.N.); (A.C.M.); (S.M.); (G.H.); (J.S.); (A.L.); (F.R.); (G.C.)
- Reference Centre for Neuromuscular Diseases AOC, Clinical Investigation Centre, Pediatric Neurology Department, Montpellier University Hospital, 34000 Montpellier, France
| | - Gilles Carnac
- PhyMedExp, University of Montpellier, Inserm, CNRS, 34295 Montpellier, France; (C.N.); (A.C.M.); (S.M.); (G.H.); (J.S.); (A.L.); (F.R.); (G.C.)
| |
Collapse
|
29
|
A nutrient-responsive hormonal circuit mediates an inter-tissue program regulating metabolic homeostasis in adult Drosophila. Nat Commun 2021; 12:5178. [PMID: 34462441 PMCID: PMC8405823 DOI: 10.1038/s41467-021-25445-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 08/04/2021] [Indexed: 02/07/2023] Open
Abstract
Animals maintain metabolic homeostasis by modulating the activity of specialized organs that adjust internal metabolism to external conditions. However, the hormonal signals coordinating these functions are incompletely characterized. Here we show that six neurosecretory cells in the Drosophila central nervous system respond to circulating nutrient levels by releasing Capa hormones, homologs of mammalian neuromedin U, which activate the Capa receptor (CapaR) in peripheral tissues to control energy homeostasis. Loss of Capa/CapaR signaling causes intestinal hypomotility and impaired nutrient absorption, which gradually deplete internal nutrient stores and reduce organismal lifespan. Conversely, increased Capa/CapaR activity increases fluid and waste excretion. Furthermore, Capa/CapaR inhibits the release of glucagon-like adipokinetic hormone from the corpora cardiaca, which restricts energy mobilization from adipose tissue to avoid harmful hyperglycemia. Our results suggest that the Capa/CapaR circuit occupies a central node in a homeostatic program that facilitates the digestion and absorption of nutrients and regulates systemic energy balance.
Collapse
|
30
|
Barefield DY, Sell JJ, Tahtah I, Kearns SD, McNally EM, Demonbreun AR. Loss of dysferlin or myoferlin results in differential defects in excitation-contraction coupling in mouse skeletal muscle. Sci Rep 2021; 11:15865. [PMID: 34354129 PMCID: PMC8342512 DOI: 10.1038/s41598-021-95378-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 07/26/2021] [Indexed: 11/25/2022] Open
Abstract
Muscular dystrophies are disorders characterized by progressive muscle loss and weakness that are both genotypically and phenotypically heterogenous. Progression of muscle disease arises from impaired regeneration, plasma membrane instability, defective membrane repair, and calcium mishandling. The ferlin protein family, including dysferlin and myoferlin, are calcium-binding, membrane-associated proteins that regulate membrane fusion, trafficking, and tubule formation. Mice lacking dysferlin (Dysf), myoferlin (Myof), and both dysferlin and myoferlin (Fer) on an isogenic inbred 129 background were previously demonstrated that loss of both dysferlin and myoferlin resulted in more severe muscle disease than loss of either gene alone. Furthermore, Fer mice had disordered triad organization with visibly malformed transverse tubules and sarcoplasmic reticulum, suggesting distinct roles of dysferlin and myoferlin. To assess the physiological role of disorganized triads, we now assessed excitation contraction (EC) coupling in these models. We identified differential abnormalities in EC coupling and ryanodine receptor disruption in flexor digitorum brevis myofibers isolated from ferlin mutant mice. We found that loss of dysferlin alone preserved sensitivity for EC coupling and was associated with larger ryanodine receptor clusters compared to wildtype myofibers. Loss of myoferlin alone or together with a loss of dysferlin reduced sensitivity for EC coupling, and produced disorganized and smaller ryanodine receptor cluster size compared to wildtype myofibers. These data reveal impaired EC coupling in Myof and Fer myofibers and slightly potentiated EC coupling in Dysf myofibers. Despite high homology, dysferlin and myoferlin have differential roles in regulating sarcotubular formation and maintenance resulting in unique impairments in calcium handling properties.
Collapse
Affiliation(s)
- David Y Barefield
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, 303 E Superior Lurie 5-500, Chicago, IL, 60611, USA. .,Department of Cell and Molecular Physiology, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL, 60153, USA.
| | - Jordan J Sell
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, 303 E Superior Lurie 5-500, Chicago, IL, 60611, USA
| | - Ibrahim Tahtah
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, 303 E Superior Lurie 5-500, Chicago, IL, 60611, USA
| | - Samuel D Kearns
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, 303 E Superior Lurie 5-500, Chicago, IL, 60611, USA
| | - Elizabeth M McNally
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, 303 E Superior Lurie 5-500, Chicago, IL, 60611, USA
| | - Alexis R Demonbreun
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, 303 E Superior Lurie 5-500, Chicago, IL, 60611, USA. .,Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA. .,Center for Genetic Medicine, Northwestern University, 303 E Superior Lurie 5-512, Chicago, IL, 60611, USA.
| |
Collapse
|
31
|
Keil C, Klein J, Schmitt F, Zorlu Y, Haase H, Yücesan G. Arylphosphonate-Tethered Porphyrins: Fluorescence Silencing Speaks a Metal Language in Living Enterocytes*. Chembiochem 2021; 22:1925-1931. [PMID: 33554446 PMCID: PMC8252553 DOI: 10.1002/cbic.202100031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/04/2021] [Indexed: 12/22/2022]
Abstract
We report the application of a highly versatile and engineerable novel sensor platform to monitor biologically significant and toxic metal ions in live human Caco-2 enterocytes. The extended conjugation between the fluorescent porphyrin core and metal ions through aromatic phenylphosphonic acid tethers generates a unique turn off and turn on fluorescence and, in addition, shifts in absorption and emission spectra for zinc, cobalt, cadmium and mercury. The reported fluorescent probes p-H8 TPPA and m-H8 TPPA can monitor a wide range of metal ion concentrations via fluorescence titration and also via fluorescence decay curves. Cu- and Zn-induced turn off fluorescence can be differentially reversed by the addition of common chelators. Both p-H8 TPPA and m-H8 TPPA readily pass the mammalian cellular membrane due to their amphipathic character as confirmed by confocal microscopic imaging of living enterocytes.
Collapse
Affiliation(s)
- Claudia Keil
- Technische Universität BerlinChair of Food Chemistry and ToxicologyStraße des 17. Juni 13510623BerlinGermany
| | - Julia Klein
- Technische Universität BerlinChair of Food Chemistry and ToxicologyStraße des 17. Juni 13510623BerlinGermany
| | - Franz‐Josef Schmitt
- Martin-Luther-Universität Halle-WittenbergDepartment of Physicsvon-Danckelmann-Platz 306120Halle/SaaleGermany
| | - Yunus Zorlu
- Department of ChemistryFaculty of ScienceGebze Technical University41400Gebze-KocaeliTurkey
| | - Hajo Haase
- Technische Universität BerlinChair of Food Chemistry and ToxicologyStraße des 17. Juni 13510623BerlinGermany
| | - Gündoğ Yücesan
- Technische Universität BerlinChair of Food Chemistry and ToxicologyStraße des 17. Juni 13510623BerlinGermany
| |
Collapse
|
32
|
Memme JM, Slavin M, Moradi N, Hood DA. Mitochondrial Bioenergetics and Turnover during Chronic Muscle Disuse. Int J Mol Sci 2021; 22:ijms22105179. [PMID: 34068411 PMCID: PMC8153634 DOI: 10.3390/ijms22105179] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 05/07/2021] [Accepted: 05/11/2021] [Indexed: 12/15/2022] Open
Abstract
Periods of muscle disuse promote marked mitochondrial alterations that contribute to the impaired metabolic health and degree of atrophy in the muscle. Thus, understanding the molecular underpinnings of muscle mitochondrial decline with prolonged inactivity is of considerable interest. There are translational applications to patients subjected to limb immobilization following injury, illness-induced bed rest, neuropathies, and even microgravity. Studies in these patients, as well as on various pre-clinical rodent models have elucidated the pathways involved in mitochondrial quality control, such as mitochondrial biogenesis, mitophagy, fission and fusion, and the corresponding mitochondrial derangements that underlie the muscle atrophy that ensues from inactivity. Defective organelles display altered respiratory function concurrent with increased accumulation of reactive oxygen species, which exacerbate myofiber atrophy via degradative pathways. The preservation of muscle quality and function is critical for maintaining mobility throughout the lifespan, and for the prevention of inactivity-related diseases. Exercise training is effective in preserving muscle mass by promoting favourable mitochondrial adaptations that offset the mitochondrial dysfunction, which contributes to the declines in muscle and whole-body metabolic health. This highlights the need for further investigation of the mechanisms in which mitochondria contribute to disuse-induced atrophy, as well as the specific molecular targets that can be exploited therapeutically.
Collapse
Affiliation(s)
| | | | | | - David A. Hood
- Correspondence: ; Tel.: +1-(416)-736-2100 (ext. 66640)
| |
Collapse
|
33
|
Ellwood RA, Piasecki M, Szewczyk NJ. Caenorhabditis elegans as a Model System for Duchenne Muscular Dystrophy. Int J Mol Sci 2021; 22:ijms22094891. [PMID: 34063069 PMCID: PMC8125261 DOI: 10.3390/ijms22094891] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 04/26/2021] [Accepted: 04/26/2021] [Indexed: 02/07/2023] Open
Abstract
The nematode worm Caenorhabditis elegans has been used extensively to enhance our understanding of the human neuromuscular disorder Duchenne Muscular Dystrophy (DMD). With new arising clinically relevant models, technologies and treatments, there is a need to reconcile the literature and collate the key findings associated with this model.
Collapse
Affiliation(s)
- Rebecca A. Ellwood
- Medical Research Council (MRC) Versus Arthritis, Centre for Musculoskeletal Ageing Research, Royal Derby Hospital, University of Nottingham, Derby DE22 3DT, UK; (R.A.E.); (M.P.)
- National Institute for Health Research, Nottingham Biomedical Research Centre, Derby DE22 3DT, UK
| | - Mathew Piasecki
- Medical Research Council (MRC) Versus Arthritis, Centre for Musculoskeletal Ageing Research, Royal Derby Hospital, University of Nottingham, Derby DE22 3DT, UK; (R.A.E.); (M.P.)
- National Institute for Health Research, Nottingham Biomedical Research Centre, Derby DE22 3DT, UK
| | - Nathaniel J. Szewczyk
- Medical Research Council (MRC) Versus Arthritis, Centre for Musculoskeletal Ageing Research, Royal Derby Hospital, University of Nottingham, Derby DE22 3DT, UK; (R.A.E.); (M.P.)
- National Institute for Health Research, Nottingham Biomedical Research Centre, Derby DE22 3DT, UK
- Ohio Musculoskeletal and Neurologic Institute, Ohio University, Athens, OH 45701, USA
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
- Correspondence:
| |
Collapse
|
34
|
Sarco/Endoplasmic Reticulum Ca 2+ Transporting ATPase (SERCA) Modulates Autophagic, Inflammatory, and Mitochondrial Responses during Influenza A Virus Infection in Human Lung Cells. J Virol 2021; 95:JVI.00217-21. [PMID: 33692207 PMCID: PMC8139658 DOI: 10.1128/jvi.00217-21] [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] [Indexed: 01/04/2023] Open
Abstract
Influenza A virus is an important human pathogen causing significant morbidity and mortality. Numerous host factors and cellular responses are dysregulated during influenza A virus infection. This includes arrest of autophagic flux dependent on the influenza M2 ion channel, but little is known which host factors participate in this autophagic dysfunction. Sarco/endoplasmic reticulum calcium ATPase (SERCA) is known to regulate transport of calcium ions between the cytoplasm and the sarco/endoplasmic reticulum, and has been positively correlated with autophagic flux. Herein, we found that SERCA activity was suppressed in influenza A virus infected human lung cells (H1395) and that CDN1163, an activator of SERCA, restored autophagic flux and thus reduced autophagosome accumulation caused by the influenza A virus. Activating SERCA activity with CDN1163 also decreased expression of inflammatory cytokines and chemokines and attenuated mitochondrial dysfunction in IAV-infected H1395 cells. Conversely, SERCA inhibition or genetic ablation aggravated the autophagy dysfunction, mitochondria, and inflammatory responses in the cells infected with influenza A virus. Further study showed that SERCA might regulate the inflammatory response by modulating phosphorylation of MAPK-JNK pathway. These findings showed that the influenza A virus induced autophagic flux blocking, inflammatory response and mitochondrial dysfunction by inhibiting SERCA activity. This study provides further understanding of the host-viral interactions between the influenza virus, SERCA activity, autophagy, inflammatory response, and mitochondrial function. SERCA may be a potential host target for decreasing inflammatory and superoxide injury during influenza A virus infection.IMPORTANCE:IAV is a major cause of infectious respiratory diseases, characterized by a marked respiratory tract inflammatory response that causes morbidity and mortality in seasonal epidemics, or pandemic outbreaks. SERCA is a critical component in maintaining cellular calcium levels, and is positively correlated with autophagic flux. Here, we discovered that SERCA is suppressed in IAV-infected human lung cells and influenza A virus induces blocking of autophagic flux, inflammatory response and mitochondrial dysfunction by inhibiting SERCA. We posit that the pharmacological activation of SERCA can be a powerful intervention strategy to prevent autophagy arrest, inflammatory response, and mitochondrial dysfunction in IAV-infected cells. Therefore, SERCA activity modulation could be a potential therapeutic strategy for managing clinical symptoms of severe influenza disease.
Collapse
|
35
|
Extra-skeletal effects of dietary calcium: Impact on the cardiovascular system, obesity, and cancer. ADVANCES IN FOOD AND NUTRITION RESEARCH 2021; 96:1-25. [PMID: 34112350 DOI: 10.1016/bs.afnr.2021.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Calcium is well known to be integral to bone and muscle health, with deleterious effects such as osteoporosis associated with inadequate calcium intake. Recent studies have also highlighted the significant effects of calcium in extra-musculoskeletal functioning, including the cardiovascular system, obesity, and cancer. Calcium impacts the cardiovascular system as an antagonist associated with a reduction in hypertension, increase vasodilation, and improvement in blood vessel function when obtained in the diet as an organic source, through food. However, the inorganic source of calcium, found in supplements, may be negatively associated with the cardiovascular system due to plaque deposits and atherogenesis when taken in excess. Some studies suggest that calcium intake may impact obesity by regulation of adipogenesis and reducing fat deposits with resulting weight loss. The pathogenesis of calcium for reducing obesity is thought to be related in part to its impact on gut microbiota profile, with the suggestion that calcium may have prebiotic properties. Animal and some human studies propose that calcium may also have a role in cancer prevention and/or treatment due to its function in the cell proliferation process and the impact on hormonal regulation, and thus warrants more investigations in the human population. Some prospective and small clinical studies suggest that calcium may be beneficial for colorectal cancer. Overall, emerging research in various areas continues to highlight the essentiality of dietary calcium for functioning at the molecular and biochemical level toward improvement in health and some chronic disease conditions.
Collapse
|
36
|
Valera IC, Wacker AL, Hwang HS, Holmes C, Laitano O, Landstrom AP, Parvatiyar MS. Essential roles of the dystrophin-glycoprotein complex in different cardiac pathologies. Adv Med Sci 2021; 66:52-71. [PMID: 33387942 DOI: 10.1016/j.advms.2020.12.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 12/12/2020] [Accepted: 12/17/2020] [Indexed: 12/20/2022]
Abstract
The dystrophin-glycoprotein complex (DGC), situated at the sarcolemma dynamically remodels during cardiac disease. This review examines DGC remodeling as a common denominator in diseases affecting heart function and health. Dystrophin and the DGC serve as broad cytoskeletal integrators that are critical for maintaining stability of muscle membranes. The presence of pathogenic variants in genes encoding proteins of the DGC can cause absence of the protein and/or alterations in other complex members leading to muscular dystrophies. Targeted studies have allowed the individual functions of affected proteins to be defined. The DGC has demonstrated its dynamic function, remodeling under a number of conditions that stress the heart. Beyond genetic causes, pathogenic processes also impinge on the DGC, causing alterations in the abundance of dystrophin and associated proteins during cardiac insult such as ischemia-reperfusion injury, mechanical unloading, and myocarditis. When considering new therapeutic strategies, it is important to assess DGC remodeling as a common factor in various heart diseases. The DGC connects the internal F-actin-based cytoskeleton to laminin-211 of the extracellular space, playing an important role in the transmission of mechanical force to the extracellular matrix. The essential functions of dystrophin and the DGC have been long recognized. DGC based therapeutic approaches have been primarily focused on muscular dystrophies, however it may be a beneficial target in a number of disorders that affect the heart. This review provides an account of what we now know, and discusses how this knowledge can benefit persistent health conditions in the clinic.
Collapse
Affiliation(s)
- Isela C Valera
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL, USA
| | - Amanda L Wacker
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL, USA
| | - Hyun Seok Hwang
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL, USA
| | - Christina Holmes
- Department of Chemical and Biomedical Engineering, Florida A&M University-Florida State University College of Engineering, Tallahassee, FL, USA
| | - Orlando Laitano
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL, USA
| | - Andrew P Landstrom
- Department of Pediatrics, Division of Cardiology, Duke University School of Medicine, Durham, NC, USA; Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA
| | - Michelle S Parvatiyar
- Department of Nutrition, Food and Exercise Sciences, Florida State University, Tallahassee, FL, USA.
| |
Collapse
|
37
|
Rodriguez-Gonzalez M, Lubian-Gutierrez M, Cascales-Poyatos HM, Perez-Reviriego AA, Castellano-Martinez A. Role of the Renin-Angiotensin-Aldosterone System in Dystrophin-Deficient Cardiomyopathy. Int J Mol Sci 2020; 22:356. [PMID: 33396334 PMCID: PMC7796305 DOI: 10.3390/ijms22010356] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 12/27/2020] [Accepted: 12/28/2020] [Indexed: 12/16/2022] Open
Abstract
Dystrophin-deficient cardiomyopathy (DDC) is currently the leading cause of death in patients with dystrophinopathies. Targeting myocardial fibrosis (MF) has become a major therapeutic goal in order to prevent the occurrence of DDC. We aimed to review and summarize the current evidence about the role of the renin-angiotensin-aldosterone system (RAAS) in the development and perpetuation of MF in DCC. We conducted a comprehensive search of peer-reviewed English literature on PubMed about this subject. We found increasing preclinical evidence from studies in animal models during the last 20 years pointing out a central role of RAAS in the development of MF in DDC. Local tissue RAAS acts directly mainly through its main fibrotic component angiotensin II (ANG2) and its transducer receptor (AT1R) and downstream TGF-b pathway. Additionally, it modulates the actions of most of the remaining pro-fibrotic factors involved in DDC. Despite limited clinical evidence, RAAS blockade constitutes the most studied, available and promising therapeutic strategy against MF and DDC. Conclusion: Based on the evidence reviewed, it would be recommendable to start RAAS blockade therapy through angiotensin converter enzyme inhibitors (ACEI) or AT1R blockers (ARBs) alone or in combination with mineralocorticoid receptor antagonists (MRa) at the youngest age after the diagnosis of dystrophinopathies, in order to delay the occurrence or slow the progression of MF, even before the detection of any cardiovascular alteration.
Collapse
Affiliation(s)
- Moises Rodriguez-Gonzalez
- Pediatric Cardiology Division of Puerta del Mar University Hospital, University of Cadiz, 11009 Cadiz, Spain
- Biomedical Research and Innovation Institute of Cadiz (INiBICA), Research Unit, Puerta del Mar University Hospital, University of Cadiz, 11009 Cadiz, Spain;
| | - Manuel Lubian-Gutierrez
- Pediatric Neurology Division of Puerta del Mar University Hospital, University of Cadiz, 11009 Cadiz, Spain;
- Pediatric Division of Doctor Cayetano Roldan Primary Care Center, 11100 San Fernando, Spain
| | | | | | - Ana Castellano-Martinez
- Biomedical Research and Innovation Institute of Cadiz (INiBICA), Research Unit, Puerta del Mar University Hospital, University of Cadiz, 11009 Cadiz, Spain;
- Pediatric Nephrology Division of Puerta del Mar University Hospital, University of Cadiz, 11009 Cadiz, Spain
| |
Collapse
|
38
|
Restoration of Sarcoplasmic Reticulum Ca 2+ ATPase (SERCA) Activity Prevents Age-Related Muscle Atrophy and Weakness in Mice. Int J Mol Sci 2020; 22:ijms22010037. [PMID: 33375170 PMCID: PMC7792969 DOI: 10.3390/ijms22010037] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/18/2020] [Accepted: 12/19/2020] [Indexed: 02/08/2023] Open
Abstract
Sarcopenia has a significant negative impact on healthspan in the elderly and effective pharmacologic interventions remain elusive. We have previously demonstrated that sarcopenia is associated with reduced activity of the sarcoplasmic reticulum Ca2+ ATPase (SERCA) pump. We asked whether restoring SERCA activity using pharmacologic activation in aging mice could mitigate the sarcopenia phenotype. We treated 16-month male C57BL/6J mice with vehicle or CDN1163, an allosteric SERCA activator, for 10 months. At 26 months, maximal SERCA activity was reduced 41% in gastrocnemius muscle in vehicle-treated mice but maintained in old CDN1163 treated mice. Reductions in gastrocnemius mass (9%) and in vitro specific force generation in extensor digitorum longus muscle (11%) in 26 versus 16-month-old wild-type mice were also reversed by CDN1163. CDN1163 administered by intra-peritoneal injection also prevented the increase in mitochondrial ROS production in gastrocnemius muscles of aged mice. Transcriptomic analysis revealed that these effects are at least in part mediated by enhanced cellular energetics by activation of PGC1-α, UCP1, HSF1, and APMK and increased regenerative capacity by suppression of MEF2C and p38 MAPK signaling. Together, these exciting findings are the first to support that pharmacological targeting of SERCA can be an effective therapy to counter age-related muscle dysfunction.
Collapse
|
39
|
Hyatt HW, Powers SK. Disturbances in Calcium Homeostasis Promotes Skeletal Muscle Atrophy: Lessons From Ventilator-Induced Diaphragm Wasting. Front Physiol 2020; 11:615351. [PMID: 33391032 PMCID: PMC7773636 DOI: 10.3389/fphys.2020.615351] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 11/19/2020] [Indexed: 12/23/2022] Open
Abstract
Mechanical ventilation (MV) is often a life-saving intervention for patients in respiratory failure. Unfortunately, a common and undesired consequence of prolonged MV is the development of diaphragmatic atrophy and contractile dysfunction. This MV-induced diaphragmatic weakness is commonly labeled “ventilator-induced diaphragm dysfunction” (VIDD). VIDD is an important clinical problem because diaphragmatic weakness is a major risk factor for the failure to wean patients from MV; this inability to remove patients from ventilator support results in prolonged hospitalization and increased morbidity and mortality. Although several processes contribute to the development of VIDD, it is clear that oxidative stress leading to the rapid activation of proteases is a primary contributor. While all major proteolytic systems likely contribute to VIDD, emerging evidence reveals that activation of the calcium-activated protease calpain plays a required role. This review highlights the signaling pathways leading to VIDD with a focus on the cellular events that promote increased cytosolic calcium levels and the subsequent activation of calpain within diaphragm muscle fibers. In particular, we discuss the emerging evidence that increased mitochondrial production of reactive oxygen species promotes oxidation of the ryanodine receptor/calcium release channel, resulting in calcium release from the sarcoplasmic reticulum, accelerated proteolysis, and VIDD. We conclude with a discussion of important and unanswered questions associated with disturbances in calcium homeostasis in diaphragm muscle fibers during prolonged MV.
Collapse
Affiliation(s)
- Hayden W Hyatt
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, United States
| | - Scott K Powers
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, FL, United States
| |
Collapse
|
40
|
Soo TCC, See SA, Bhassu S. Potential muscle activity disturbance in Penaeus monodon during Acute Hepatopancreatic Necrosis Disease (AHPND) infection: Inference through gene expression, calcium concentration, and MicroRNA. J Invertebr Pathol 2020; 177:107497. [PMID: 33130047 DOI: 10.1016/j.jip.2020.107497] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 01/21/2023]
Abstract
Global shrimp aquaculture farmers have suffered major economic losses due to disease outbreaks. A notable shrimp disease is Acute Hepatopancreatic Necrosis Disease (AHPND), which is caused by a new strain of Vibrio parahaemolyticus bacteria (VpAHPND) that mainly inhabits the shrimp gut and damages the hepatopancreas. Fewer studies have investigated whether this disease will affect shrimp muscle functioning or cause any muscle damage. We challenged Penaeus monodon shrimp with VpAHPND bacteria using an immersion method. Expression of Dystrophin gene, an important regulatory gene for maintenance of muscle integrity, was quantified from muscle samples using qRT-PCR. Additional verification was conducted by determining calcium concentration and bta-miR-4286 and dre-miR-107b miRNAs expression. P. monodon dystrophin gene demonstrated the highest expression level during AHPND infection when muscle calcium concentration was detected at its lowest level at 6 h post-infection (hpi). The highest muscle calcium concentration, determined at 36 hpi, was supported by higher bta-miR-4286 miRNA expression and lower dre-miR-107b miRNA expression in VpAHPND-infected samples compared to uninfected samples at the same time point. We deduced an interactive relationship between dystrophin gene expression, calcium concentration, and miRNA expression in P. monodon muscle tissues triggered by the invading VpAHPND bacterium.
Collapse
Affiliation(s)
- Tze Chiew Christie Soo
- Animal Genetics and Genome Evolutionary Laboratory (AGAGEL), Department of Genetics and Molecular Biology, Institute of Biological Sciences, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia; Terra Aqua Laboratory, Centre for Research in Biotechnology for Agriculture (CEBAR), Research Management and Innovation Complex, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - SiouNing Aileen See
- Animal Genetics and Genome Evolutionary Laboratory (AGAGEL), Department of Genetics and Molecular Biology, Institute of Biological Sciences, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia; Terra Aqua Laboratory, Centre for Research in Biotechnology for Agriculture (CEBAR), Research Management and Innovation Complex, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Subha Bhassu
- Animal Genetics and Genome Evolutionary Laboratory (AGAGEL), Department of Genetics and Molecular Biology, Institute of Biological Sciences, Faculty of Science, University of Malaya, 50603 Kuala Lumpur, Malaysia; Terra Aqua Laboratory, Centre for Research in Biotechnology for Agriculture (CEBAR), Research Management and Innovation Complex, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| |
Collapse
|
41
|
|
42
|
"Betwixt Mine Eye and Heart a League Is Took": The Progress of Induced Pluripotent Stem-Cell-Based Models of Dystrophin-Associated Cardiomyopathy. Int J Mol Sci 2020; 21:ijms21196997. [PMID: 32977524 PMCID: PMC7582534 DOI: 10.3390/ijms21196997] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/17/2020] [Accepted: 09/21/2020] [Indexed: 12/19/2022] Open
Abstract
The ultimate goal of precision disease modeling is to artificially recreate the disease of affected people in a highly controllable and adaptable external environment. This field has rapidly advanced which is evident from the application of patient-specific pluripotent stem-cell-derived precision therapies in numerous clinical trials aimed at a diverse set of diseases such as macular degeneration, heart disease, spinal cord injury, graft-versus-host disease, and muscular dystrophy. Despite the existence of semi-adequate treatments for tempering skeletal muscle degeneration in dystrophic patients, nonischemic cardiomyopathy remains one of the primary causes of death. Therefore, cardiovascular cells derived from muscular dystrophy patients' induced pluripotent stem cells are well suited to mimic dystrophin-associated cardiomyopathy and hold great promise for the development of future fully effective therapies. The purpose of this article is to convey the realities of employing precision disease models of dystrophin-associated cardiomyopathy. This is achieved by discussing, as suggested in the title echoing William Shakespeare's words, the settlements (or "leagues") made by researchers to manage the constraints ("betwixt mine eye and heart") distancing them from achieving a perfect precision disease model.
Collapse
|
43
|
Zorlu Y, Brown C, Keil C, Ayhan MM, Haase H, Thompson RB, Lengyel I, Yücesan G. Fluorescent Arylphosphonic Acids: Synergic Interactions between Bone and the Fluorescent Core. Chemistry 2020; 26:11129-11134. [PMID: 32293767 PMCID: PMC7496659 DOI: 10.1002/chem.202001613] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Indexed: 12/23/2022]
Abstract
Herein, we report the third generation of fluorescent probes (arylphosphonic acids) to target calcifications, particularly hydroxyapatite (HAP). In this study, we use highly conjugated porphyrin-based arylphosphonic acids and their diesters, namely 5,10,15,20-tetrakis[m-(diethoxyphosphoryl)phenyl]porphyrin (m-H8 TPPA-OEt8 ) and 5,10,15,20-tetrakis [m-phenylphosphonic acid]porphyrin (m-H8 TPPA), in comparison with their positional isomers 5,10,15,20-tetrakis[p-(diisopropoxyphosphoryl)phenyl]porphyrin (p-H8 TPPA-iPr8 ) and 5,10,15,20-tetrakis [p-phenylphosphonic acid]porphyrin (p-H8 TPPA), which have phosphonic acid units bonded to sp2 carbon atoms of the fluorescent core. The conjugation of the fluorescent core is thus extended to the (HAP) through sp2 -bonded -PO3 H2 units, which generates increased fluorescence upon HAP binding. The resulting fluorescent probes are highly sensitive towards the HAP in rat bone sections. The designed probes are readily taken up by cells. Due to the lower reported toxicity of (p-H8 TPPA), these probes could find applications in monitoring bone resorption or adsorption, or imaging vascular or soft tissue calcifications for breast cancer diagnosis etc.
Collapse
Affiliation(s)
- Yunus Zorlu
- Department of ChemistryFaculty of ScienceGebze Technical University41400Gebze-KocaeliTurkey
| | - Connor Brown
- Wellcome-Wolfson Institute for Experimental MedicineSchool of Medicine, Dentistry and Biomedical ScienceQueen's University BelfastBelfastBT9 7BLUK
| | - Claudia Keil
- Technische Universität BerlinChair of Food Chemistry and ToxicologyStraße des 17. Juni 13510623BerlinGermany
| | - M. Menaf Ayhan
- Department of ChemistryFaculty of ScienceGebze Technical University41400Gebze-KocaeliTurkey
| | - Hajo Haase
- Technische Universität BerlinChair of Food Chemistry and ToxicologyStraße des 17. Juni 13510623BerlinGermany
| | - Richard B. Thompson
- Department of Biochemistry and Molecular BiologyUniversity of Maryland School of MedicineBaltimoreMaryland21201USA
| | - Imre Lengyel
- Wellcome-Wolfson Institute for Experimental MedicineSchool of Medicine, Dentistry and Biomedical ScienceQueen's University BelfastBelfastBT9 7BLUK
| | - Gündoğ Yücesan
- Technische Universität BerlinChair of Food Chemistry and ToxicologyStraße des 17. Juni 13510623BerlinGermany
| |
Collapse
|
44
|
Truong KM, Cherednichenko G, Pessah IN. Interactions of Dichlorodiphenyltrichloroethane (DDT) and Dichlorodiphenyldichloroethylene (DDE) With Skeletal Muscle Ryanodine Receptor Type 1. Toxicol Sci 2020; 170:509-524. [PMID: 31127943 DOI: 10.1093/toxsci/kfz120] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Dichlorodiphenyltrichloroethane (DDT) and its metabolite dichlorodiphenyldichloroethylene (DDE) are ubiquitous in the environment and detected in tissues of living organisms. Although DDT owes its insecticidal activity to impeding closure of voltage-gated sodium channels, it mediates toxicity in mammals by acting as an endocrine disruptor (ED). Numerous studies demonstrate DDT/DDE to be EDs, but studies examining muscle-specific effects mediated by nonhormonal receptors in mammals are lacking. Therefore, we investigated whether o,p'-DDT, p,p'-DDT, o,p'-DDE, and p,p'-DDE (DDx, collectively) alter the function of ryanodine receptor type 1 (RyR1), a protein critical for skeletal muscle excitation-contraction coupling and muscle health. DDx (0.01-10 µM) elicited concentration-dependent increases in [3H]ryanodine ([3H]Ry) binding to RyR1 with o,p'-DDE showing highest potency and efficacy. DDx also showed sex differences in [3H]Ry-binding efficacy toward RyR1, where [3H]Ry-binding in female muscle preparations was greater than male counterparts. Measurements of Ca2+ transport across sarcoplasmic reticulum (SR) membrane vesicles further confirmed DDx can selectively engage with RyR1 to cause Ca2+ efflux from SR stores. DDx also disrupts RyR1-signaling in HEK293T cells stably expressing RyR1 (HEK-RyR1). Pretreatment with DDx (0.1-10 µM) for 100 s, 12 h, or 24 h significantly sensitized Ca2+-efflux triggered by RyR agonist caffeine in a concentration-dependent manner. o,p'-DDE (24 h; 1 µM) significantly increased Ca2+-transient amplitude from electrically stimulated mouse myotubes compared with control and displayed abnormal fatigability. In conclusion, our study demonstrates DDx can directly interact and modulate RyR1 conformation, thereby altering SR Ca2+-dynamics and sensitize RyR1-expressing cells to RyR1 activators, which may ultimately contribute to long-term impairments in muscle health.
Collapse
Affiliation(s)
- Kim M Truong
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, California 95616-5270
| | - Gennady Cherednichenko
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, California 95616-5270
| | - Isaac N Pessah
- Department of Molecular Biosciences, School of Veterinary Medicine, University of California, Davis, Davis, California 95616-5270
| |
Collapse
|
45
|
Bittel AJ, Sreetama SC, Bittel DC, Horn A, Novak JS, Yokota T, Zhang A, Maruyama R, Rowel Q. Lim K, Jaiswal JK, Chen YW. Membrane Repair Deficit in Facioscapulohumeral Muscular Dystrophy. Int J Mol Sci 2020; 21:E5575. [PMID: 32759720 PMCID: PMC7432481 DOI: 10.3390/ijms21155575] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 07/28/2020] [Accepted: 07/30/2020] [Indexed: 12/14/2022] Open
Abstract
Deficits in plasma membrane repair have been identified in dysferlinopathy and Duchenne Muscular Dystrophy, and contribute to progressive myopathy. Although Facioscapulohumeral Muscular Dystrophy (FSHD) shares clinicopathological features with these muscular dystrophies, it is unknown if FSHD is characterized by plasma membrane repair deficits. Therefore, we exposed immortalized human FSHD myoblasts, immortalized myoblasts from unaffected siblings, and myofibers from a murine model of FSHD (FLExDUX4) to focal, pulsed laser ablation of the sarcolemma. Repair kinetics and success were determined from the accumulation of intracellular FM1-43 dye post-injury. We subsequently treated FSHD myoblasts with a DUX4-targeting antisense oligonucleotide (AON) to reduce DUX4 expression, and with the antioxidant Trolox to determine the role of DUX4 expression and oxidative stress in membrane repair. Compared to unaffected myoblasts, FSHD myoblasts demonstrate poor repair and a greater percentage of cells that failed to repair, which was mitigated by AON and Trolox treatments. Similar repair deficits were identified in FLExDUX4 myofibers. This is the first study to identify plasma membrane repair deficits in myoblasts from individuals with FSHD, and in myofibers from a murine model of FSHD. Our results suggest that DUX4 expression and oxidative stress may be important targets for future membrane-repair therapies.
Collapse
Affiliation(s)
- Adam J. Bittel
- Research Center for Genetic Medicine, Children’s National Hospital, 111 Michigan Ave NW, Washington, DC 20010, USA; (A.J.B.); (S.C.S.); (D.C.B.); (A.H.); (J.S.N.); (A.Z.)
| | - Sen Chandra Sreetama
- Research Center for Genetic Medicine, Children’s National Hospital, 111 Michigan Ave NW, Washington, DC 20010, USA; (A.J.B.); (S.C.S.); (D.C.B.); (A.H.); (J.S.N.); (A.Z.)
| | - Daniel C. Bittel
- Research Center for Genetic Medicine, Children’s National Hospital, 111 Michigan Ave NW, Washington, DC 20010, USA; (A.J.B.); (S.C.S.); (D.C.B.); (A.H.); (J.S.N.); (A.Z.)
| | - Adam Horn
- Research Center for Genetic Medicine, Children’s National Hospital, 111 Michigan Ave NW, Washington, DC 20010, USA; (A.J.B.); (S.C.S.); (D.C.B.); (A.H.); (J.S.N.); (A.Z.)
| | - James S. Novak
- Research Center for Genetic Medicine, Children’s National Hospital, 111 Michigan Ave NW, Washington, DC 20010, USA; (A.J.B.); (S.C.S.); (D.C.B.); (A.H.); (J.S.N.); (A.Z.)
- Department of Genomics and Precision Medicine, The George Washington University School of Medicine and Health Science, 111 Michigan Ave NW, Washington, DC 20010, USA
| | - Toshifumi Yokota
- Department of Medical Genetics, University of Alberta, 116 St. & 85 Ave., Edmonton, AB T6G 2R3, Canada; (T.Y.); (R.M.); (K.R.Q.L.)
| | - Aiping Zhang
- Research Center for Genetic Medicine, Children’s National Hospital, 111 Michigan Ave NW, Washington, DC 20010, USA; (A.J.B.); (S.C.S.); (D.C.B.); (A.H.); (J.S.N.); (A.Z.)
| | - Rika Maruyama
- Department of Medical Genetics, University of Alberta, 116 St. & 85 Ave., Edmonton, AB T6G 2R3, Canada; (T.Y.); (R.M.); (K.R.Q.L.)
| | - Kenji Rowel Q. Lim
- Department of Medical Genetics, University of Alberta, 116 St. & 85 Ave., Edmonton, AB T6G 2R3, Canada; (T.Y.); (R.M.); (K.R.Q.L.)
| | - Jyoti K. Jaiswal
- Research Center for Genetic Medicine, Children’s National Hospital, 111 Michigan Ave NW, Washington, DC 20010, USA; (A.J.B.); (S.C.S.); (D.C.B.); (A.H.); (J.S.N.); (A.Z.)
- Department of Integrative Systems Biology, Institute for Biomedical Sciences, The George Washington University, 2121 I St. NW, Washington, DC 20052, USA
| | - Yi-Wen Chen
- Research Center for Genetic Medicine, Children’s National Hospital, 111 Michigan Ave NW, Washington, DC 20010, USA; (A.J.B.); (S.C.S.); (D.C.B.); (A.H.); (J.S.N.); (A.Z.)
- Department of Integrative Systems Biology, Institute for Biomedical Sciences, The George Washington University, 2121 I St. NW, Washington, DC 20052, USA
| |
Collapse
|
46
|
Aguayo-Ortiz R, Espinoza-Fonseca LM. Linking Biochemical and Structural States of SERCA: Achievements, Challenges, and New Opportunities. Int J Mol Sci 2020; 21:ijms21114146. [PMID: 32532023 PMCID: PMC7313052 DOI: 10.3390/ijms21114146] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/05/2020] [Accepted: 06/08/2020] [Indexed: 02/07/2023] Open
Abstract
Sarcoendoplasmic reticulum calcium ATPase (SERCA), a member of the P-type ATPase family of ion and lipid pumps, is responsible for the active transport of Ca2+ from the cytoplasm into the sarcoplasmic reticulum lumen of muscle cells, into the endoplasmic reticulum (ER) of non-muscle cells. X-ray crystallography has proven to be an invaluable tool in understanding the structural changes of SERCA, and more than 70 SERCA crystal structures representing major biochemical states (defined by bound ligand) have been deposited in the Protein Data Bank. Consequently, SERCA is one of the best characterized components of the calcium transport machinery in the cell. Emerging approaches in the field, including spectroscopy and molecular simulation, now help integrate and interpret this rich structural information to understand the conformational transitions of SERCA that occur during activation, inhibition, and regulation. In this review, we provide an overview of the crystal structures of SERCA, focusing on identifying metrics that facilitate structure-based categorization of major steps along the catalytic cycle. We examine the integration of crystallographic data with different biophysical approaches and computational methods to link biochemical and structural states of SERCA that are populated in the cell. Finally, we discuss the challenges and new opportunities in the field, including structural elucidation of functionally important and novel regulatory complexes of SERCA, understanding the structural basis of functional divergence among homologous SERCA regulators, and bridging the gap between basic and translational research directed toward therapeutic modulation of SERCA.
Collapse
|
47
|
Liu JC, Syder NC, Ghorashi NS, Willingham TB, Parks RJ, Sun J, Fergusson MM, Liu J, Holmström KM, Menazza S, Springer DA, Liu C, Glancy B, Finkel T, Murphy E. EMRE is essential for mitochondrial calcium uniporter activity in a mouse model. JCI Insight 2020; 5:134063. [PMID: 32017711 PMCID: PMC7101141 DOI: 10.1172/jci.insight.134063] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 01/29/2020] [Indexed: 11/17/2022] Open
Abstract
The mitochondrial calcium uniporter is widely accepted as the primary route of rapid calcium entry into mitochondria, where increases in matrix calcium contribute to bioenergetics but also mitochondrial permeability and cell death. Hence, regulation of uniporter activity is critical to mitochondrial homeostasis. The uniporter subunit EMRE is known to be an essential regulator of the channel-forming protein MCU in cell culture, but EMRE's impact on organismal physiology is less understood. Here we characterize a mouse model of EMRE deletion and show that EMRE is indeed required for mitochondrial calcium uniporter function in vivo. EMRE-/- mice are born less frequently; however, the mice that are born are viable, healthy, and do not manifest overt metabolic impairment, at rest or with exercise. Finally, to investigate the role of EMRE in disease processes, we examine the effects of EMRE deletion in a muscular dystrophy model associated with mitochondrial calcium overload.
Collapse
Affiliation(s)
- Julia C. Liu
- Cardiovascular Branch and
- Center for Molecular Medicine, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
- Postdoctoral Research Associate Program, National Institute of General Medical Sciences, NIH, Bethesda, Maryland, USA
| | | | | | - Thomas B. Willingham
- Systems Biology Center, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | | | | | - Maria M. Fergusson
- Cardiovascular Branch and
- Center for Molecular Medicine, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Jie Liu
- Center for Molecular Medicine, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
- Aging Institute, University of Pittsburgh and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Kira M. Holmström
- Center for Molecular Medicine, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | | | | | - Chengyu Liu
- Transgenic Core, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
| | - Brian Glancy
- Systems Biology Center, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda, Maryland, USA
| | - Toren Finkel
- Center for Molecular Medicine, National Heart, Lung, and Blood Institute, NIH, Bethesda, Maryland, USA
- Aging Institute, University of Pittsburgh and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
48
|
Human muscle pathology is associated with altered phosphoprotein profile of mitochondrial proteins in the skeletal muscle. J Proteomics 2020; 211:103556. [PMID: 31655151 DOI: 10.1016/j.jprot.2019.103556] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 10/08/2019] [Accepted: 10/17/2019] [Indexed: 12/29/2022]
Abstract
Analysis of human muscle diseases highlights the role of mitochondrial dysfunction in the skeletal muscle. Our previous work revealed that diverse upstream events correlated with altered mitochondrial proteome in human muscle biopsies. However, several proteins showed relatively unchanged expression suggesting that post-translational modifications, mainly protein phosphorylation could influence their activity and regulate mitochondrial processes. We conducted mitochondrial phosphoprotein profiling, by proteomics approach, of healthy human skeletal muscle (n = 10) and three muscle diseases (n = 10 each): Dysferlinopathy, Polymyositis and Distal Myopathy with Rimmed Vacuoles. Healthy human muscle mitochondrial proteins displayed 253 phosphorylation sites (phosphosites), which contributed to metabolic and redox processes and mitochondrial organization etc. Electron transport chain complexes accounted for 84 phosphosites. Muscle pathologies displayed 33 hyperphosphorylated and 14 hypophorphorylated sites with only 5 common proteins, indicating varied phosphorylation profile across muscle pathologies. Molecular modelling revealed altered local structure in the phosphorylated sites of Voltage-Dependent Anion Channel 1 and complex V subunit ATP5B1. Molecular dynamics simulations in complex I subunits NDUFV1, NDUFS1 and NDUFV2 revealed that phosphorylation induced structural alterations thereby influencing electron transfer and potentially altering enzyme activity. We propose that altered phosphorylation at specific sites could regulate mitochondrial protein function in the skeletal muscle during physiological and pathological processes.
Collapse
|
49
|
Wasala NB, Yue Y, Lostal W, Wasala LP, Niranjan N, Hajjar RJ, Babu GJ, Duan D. Single SERCA2a Therapy Ameliorated Dilated Cardiomyopathy for 18 Months in a Mouse Model of Duchenne Muscular Dystrophy. Mol Ther 2020; 28:845-854. [PMID: 31981493 DOI: 10.1016/j.ymthe.2019.12.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 12/20/2019] [Accepted: 12/28/2019] [Indexed: 01/16/2023] Open
Abstract
Loss of dystrophin leads to Duchenne muscular dystrophy (DMD). A pathogenic feature of DMD is the significant elevation of cytosolic calcium. Supraphysiological calcium triggers protein degradation, membrane damage, and eventually muscle death and dysfunction. Sarcoplasmic/endoplasmic reticulum (SR) calcium ATPase (SERCA) is a calcium pump that transports cytosolic calcium to the SR during excitation-contraction coupling. We hypothesize that a single systemic delivery of SERCA2a with adeno-associated virus (AAV) may improve calcium recycling and provide long-lasting benefits in DMD. To test this, we injected an AAV9 human SERCA2a vector (6 × 1012 viral genome particles/mouse) intravenously to 3-month-old mdx mice, the most commonly used DMD model. Immunostaining and western blot showed robust human SERCA2a expression in the heart and skeletal muscle for 18 months. Concomitantly, SR calcium uptake was significantly improved in these tissues. SERCA2a therapy significantly enhanced grip force and treadmill performance, completely prevented myocardial fibrosis, and normalized electrocardiograms (ECGs). Cardiac catheterization showed normalization of multiple systolic and diastolic hemodynamic parameters in treated mice. Importantly, chamber dilation was completely prevented, and ejection fraction was restored to the wild-type level. Our results suggest that a single systemic AAV9 SERCA2a therapy has the potential to provide long-lasting benefits for DMD.
Collapse
Affiliation(s)
- Nalinda B Wasala
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Yongping Yue
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - William Lostal
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Lakmini P Wasala
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| | - Nandita Niranjan
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | | | - Gopal J Babu
- Department of Cell Biology and Molecular Medicine, New Jersey Medical School, Rutgers University, Newark, NJ 07103, USA
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA; Department of Neurology, School of Medicine, University of Missouri, Columbia, MO 65212, USA; Department of Biomedical, Biological & Chemical Engineering, College of Engineering, University of Missouri, Columbia, MO 65212, USA; Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO 65212, USA.
| |
Collapse
|
50
|
Shu C, Kaxon-Rupp AN, Collado JR, Damoiseaux R, Crosbie RH. Development of a high-throughput screen to identify small molecule enhancers of sarcospan for the treatment of Duchenne muscular dystrophy. Skelet Muscle 2019; 9:32. [PMID: 31831063 PMCID: PMC6907331 DOI: 10.1186/s13395-019-0218-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 12/04/2019] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is caused by loss of sarcolemma connection to the extracellular matrix. Transgenic overexpression of the transmembrane protein sarcospan (SSPN) in the DMD mdx mouse model significantly reduces disease pathology by restoring membrane adhesion. Identifying SSPN-based therapies has the potential to benefit patients with DMD and other forms of muscular dystrophies caused by deficits in muscle cell adhesion. METHODS Standard cloning methods were used to generate C2C12 myoblasts stably transfected with a fluorescence reporter for human SSPN promoter activity. Assay development and screening were performed in a core facility using liquid handlers and imaging systems specialized for use with a 384-well microplate format. Drug-treated cells were analyzed for target gene expression using quantitative PCR and target protein expression using immunoblotting. RESULTS We investigated the gene expression profiles of SSPN and its associated proteins during myoblast differentiation into myotubes, revealing an increase in expression after 3 days of differentiation. We created C2C12 muscle cells expressing an EGFP reporter for SSPN promoter activity and observed a comparable increase in reporter levels during differentiation. Assay conditions for high-throughput screening were optimized for a 384-well microplate format and a high-content imager for the visualization of reporter levels. We conducted a screen of 3200 compounds and identified seven hits, which include an overrepresentation of L-type calcium channel antagonists, suggesting that SSPN gene activity is sensitive to calcium. Further validation of a select hit revealed that the calcium channel inhibitor felodipine increased SSPN transcript and protein levels in both wild-type and dystrophin-deficient myotubes, without increasing differentiation. CONCLUSIONS We developed a stable muscle cell line containing the promoter region of the human SSPN protein fused to a fluorescent reporter. Using the reporter cells, we created and validated a scalable, cell-based assay that is able to identify compounds that increase SSPN promoter reporter, transcript, and protein levels in wild-type and dystrophin-deficient muscle cells.
Collapse
Affiliation(s)
- Cynthia Shu
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, USA.,Department of Integrative Biology and Physiology, University of California Los Angeles, 610 Charles E. Young Drive East, Terasaki Life Sciences Building, Los Angeles, CA, 90095, USA.,Center for Duchenne Muscular Dystrophy, University of California Los Angeles, Los Angeles, USA
| | - Ariana N Kaxon-Rupp
- Department of Integrative Biology and Physiology, University of California Los Angeles, 610 Charles E. Young Drive East, Terasaki Life Sciences Building, Los Angeles, CA, 90095, USA
| | - Judd R Collado
- Department of Integrative Biology and Physiology, University of California Los Angeles, 610 Charles E. Young Drive East, Terasaki Life Sciences Building, Los Angeles, CA, 90095, USA
| | - Robert Damoiseaux
- Department of Molecular and Medicinal Pharmacology, University of California Los Angeles, Los Angeles, USA.,California NanoSystems Institute, University of California Los Angeles, Los Angeles, CA, USA
| | - Rachelle H Crosbie
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, USA. .,Department of Integrative Biology and Physiology, University of California Los Angeles, 610 Charles E. Young Drive East, Terasaki Life Sciences Building, Los Angeles, CA, 90095, USA. .,Center for Duchenne Muscular Dystrophy, University of California Los Angeles, Los Angeles, USA. .,Department of Neurology David Geffen School of Medicine, University of California Los Angeles, 610 Charles E. Young Drive East, Terasaki Life Sciences Building, Los Angeles, CA, 90095, USA.
| |
Collapse
|