1
|
Gallo-Soljancic P, De Stefano ME, Lucas-Ochoa AM, Sánchez-Rodrigo C, Cuemca-Bermejo L, González-Cuello AM, Fernández-Villalba E, Herrero MT. Age- and sex-related development of osteosarcopenia in the aging Octodon degus rodent model. FRONTIERS IN AGING 2025; 6:1486670. [PMID: 40018266 PMCID: PMC11865034 DOI: 10.3389/fragi.2025.1486670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 01/20/2025] [Indexed: 03/01/2025]
Abstract
The increase in life expectancy in recent years has resulted in a higher incidence of age-related diseases. Among these, osteoporosis and sarcopenia, collectively known as osteosarcopenia, have the most significant impact on the quality of life, general health and frailty in the elderly. As for other age-related diseases, pre-clinical studies on these conditions are primarily limited by the availability of experimental model systems. The Octodon degus (O. degus) is a long-lived diurnal rodent identified as a potential tool in ageing research. However, age-related osteosarcopenia changes have not yet been explored. In this study, male and female O. degus from juvenile to senile ages were used (6 months-7 years old). Changes in the volume of several forelimbs and hindlimbs muscles, e.g., biceps femoris, triceps brachii, femur, and humerus, were evaluated using computed tomography. Aged animals showed a significant decrease in muscle volume in both hindlimbs and forelimbs, along with a significant reduction in cortical bone volume. With ageing, sex differences were also observed, with female O. degus showing greater cortical bone volume in both hind and forelimbs, and greater muscle mass in the sole hindlimbs, compared to male. These findings enhance the characterization of O. degus as a model to study age-related pathologies, also considering sex differences, and lay down solid foundations for future studies that can address in more detail the molecular mechanisms underlying the initiation and progression of osteosarcopenia.
Collapse
Affiliation(s)
- Pablo Gallo-Soljancic
- Clinical & Experimental Neuroscience (NiCE), Department of Human Anatomy and Psychobiology, Institute for Aging Research, School of Medicine, European University for Well-Being (EUniWell), Campus Mare Nostrum, University of Murcia, Murcia, Spain
- Institute for Bio-Health Research of Murcia (IMIB-Pascual Parrilla), Campus of Health Sciences, El PalmarMurcia, Spain
| | - Maria Egle De Stefano
- Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, Rome, Italy
- Center for research in Neurobiology “Daniel Bovet”, Sapienza University of Rome, Rome, Italy
| | - Ana-María Lucas-Ochoa
- Clinical & Experimental Neuroscience (NiCE), Department of Human Anatomy and Psychobiology, Institute for Aging Research, School of Medicine, European University for Well-Being (EUniWell), Campus Mare Nostrum, University of Murcia, Murcia, Spain
- Institute for Bio-Health Research of Murcia (IMIB-Pascual Parrilla), Campus of Health Sciences, El PalmarMurcia, Spain
| | - Consuelo Sánchez-Rodrigo
- Clinical & Experimental Neuroscience (NiCE), Department of Human Anatomy and Psychobiology, Institute for Aging Research, School of Medicine, European University for Well-Being (EUniWell), Campus Mare Nostrum, University of Murcia, Murcia, Spain
| | - Lorena Cuemca-Bermejo
- Clinical & Experimental Neuroscience (NiCE), Department of Human Anatomy and Psychobiology, Institute for Aging Research, School of Medicine, European University for Well-Being (EUniWell), Campus Mare Nostrum, University of Murcia, Murcia, Spain
| | - Ana-María González-Cuello
- Clinical & Experimental Neuroscience (NiCE), Department of Human Anatomy and Psychobiology, Institute for Aging Research, School of Medicine, European University for Well-Being (EUniWell), Campus Mare Nostrum, University of Murcia, Murcia, Spain
- Institute for Bio-Health Research of Murcia (IMIB-Pascual Parrilla), Campus of Health Sciences, El PalmarMurcia, Spain
| | - Emiliano Fernández-Villalba
- Clinical & Experimental Neuroscience (NiCE), Department of Human Anatomy and Psychobiology, Institute for Aging Research, School of Medicine, European University for Well-Being (EUniWell), Campus Mare Nostrum, University of Murcia, Murcia, Spain
- Institute for Bio-Health Research of Murcia (IMIB-Pascual Parrilla), Campus of Health Sciences, El PalmarMurcia, Spain
| | - María-Trinidad Herrero
- Clinical & Experimental Neuroscience (NiCE), Department of Human Anatomy and Psychobiology, Institute for Aging Research, School of Medicine, European University for Well-Being (EUniWell), Campus Mare Nostrum, University of Murcia, Murcia, Spain
- Institute for Bio-Health Research of Murcia (IMIB-Pascual Parrilla), Campus of Health Sciences, El PalmarMurcia, Spain
| |
Collapse
|
2
|
Xue Q, Zhang D, Zou J, Wang H, Shi R, Dong L. Treatment advances of sepsis‑induced myopathy (Review). Biomed Rep 2025; 22:19. [PMID: 39651403 PMCID: PMC11621912 DOI: 10.3892/br.2024.1897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Accepted: 10/04/2024] [Indexed: 12/11/2024] Open
Abstract
Sepsis-induced myopathy (SIM) is a muscle disease caused by multiple pathological and physiological mechanisms associated with sepsis. The pathogenesis of SIM is extremely complex and still unclear, making treatment challenging. At present, clinical treatment includes early functional exercise, respiratory muscle strength training, regulation of nutritional structure and functional electrical stimulation. Drugs targeting the regulation of the ubiquitin-proteasome system, autophagy-lysosome system, calpain and caspase activation pathways, have provided potential therapeutic targets for the treatment of muscle atrophy. Stem cell transplantation therapy brings new hope for the treatment of SIM.
Collapse
Affiliation(s)
- Qiuli Xue
- Department of Intensive Care Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Deyou Zhang
- Department of Intensive Care Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jiarui Zou
- Department of Intensive Care Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Haitao Wang
- Department of Intensive Care Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Ruiyuan Shi
- Department of Intensive Care Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Lihua Dong
- Department of Intensive Care Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
3
|
Gonzalez MB, McPherson NO, Connaughton HS, Winstanley YE, Kennedy DT, Campugan CA, Febbraio MA, Barry M, Rose RD, Robker RL. Mitochondrial activator BGP-15 protects sperm quality against oxidative damage and improves embryo developmental competence. F&S SCIENCE 2025; 6:42-54. [PMID: 39675561 DOI: 10.1016/j.xfss.2024.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 12/10/2024] [Accepted: 12/10/2024] [Indexed: 12/17/2024]
Abstract
OBJECTIVE To study the efficacy of mitochondrial activator BGP-15 to preserve sperm quality and competence against cellular damage. DESIGN Spermatozoa from mice or humans were treated in vitro with BGP-15, and sperm quality markers were assessed. Spermatozoa from young (8-12 weeks old) or reproductively old (>14 months old) mice were treated with BGP-15 for 1 hour and assessed for sperm quality and preimplantation embryo development after in vitro fertilization. The safety of BGP-15 on offspring outcomes was assessed through embryo transfers. In parallel studies, spermatozoa from healthy (not infertile) men were incubated in hydrogen peroxide, to induce oxidative stress, plus increasing doses of BGP-15, and sperm quality was evaluated. Spermatozoa from patients undergoing assisted reproductive technology (ART) treatment were incubated in the optimized dose of BGP-15 for 30 minutes, and sperm quality was assessed. SUBJECTS C57BL/6 mice (N = 4-15 per group) for sperm quality and embryo development. CBAF1 mice (n = 6 per group) produced embryos for transfer. Human spermatozoa were from men with no infertility diagnosis (n = 14-20) or men undergoing ART (n = 33) at a local fertility clinic. EXPOSURE Mouse spermatozoa were treated with 10-μM BGP-15. Human spermatozoa were treated with BGP-15 at doses from 1 to 100 μM. MAIN OUTCOME MEASURES Sperm quality measures (mouse and human) included motility, mitochondrial membrane potential (JC-1 dye), deoxyribonucleic acid (DNA) fragmentation ("HALO" assay), and DNA oxidation (8-oxoguanine immunodetection). Mouse embryo and offspring measures included on-time development after in vitro fertilization, morphokinetic analysis, and blastocyst inner cell mass and trophectoderm cell number, and growth and development from birth to 21 days postnatally. RESULTS BGP-15 increased sperm motility and mitochondrial membrane potential and decreased DNA oxidation in old mice. BGP-15 improved on-time development of 2-cell and blastocyst embryos and increased the inner cell mass blastomere number. Embryos from BGP-15-treated mouse spermatozoa produced normal offspring. In human spermatozoa subjected to in vitro oxidative stress, BGP-15 increased motility by 45% and prevented DNA fragmentation (by 45%) and oxidative damage (by 60%). In spermatozoa from men attending a fertility clinic, BGP-15 increased motility by 12% and reduced both DNA oxidation and fragmentation by >20%. CONCLUSION BGP-15 protects sperm against cellular damage and has the potential to improve ART outcomes.
Collapse
Affiliation(s)
- Macarena B Gonzalez
- School of Biomedicine, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Nicole O McPherson
- School of Biomedicine, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia; Genea Fertility SA, Adelaide, South Australia, Australia; Freemasons Centre for Male Health and Wellbeing, University of Adelaide, Adelaide, South Australia, Australia
| | - Haley S Connaughton
- School of Biomedicine, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Yasmyn E Winstanley
- School of Biomedicine, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - David T Kennedy
- School of Biomedicine, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Carl A Campugan
- School of Biomedicine, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Mark A Febbraio
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Michael Barry
- School of Biomedicine, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia; Genea Fertility SA, Adelaide, South Australia, Australia
| | - Ryan D Rose
- School of Biomedicine, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia; Genea Fertility SA, Adelaide, South Australia, Australia
| | - Rebecca L Robker
- School of Biomedicine, Robinson Research Institute, University of Adelaide, Adelaide, South Australia, Australia.
| |
Collapse
|
4
|
Powers SK, Radak Z, Ji LL, Jackson M. Reactive oxygen species promote endurance exercise-induced adaptations in skeletal muscles. JOURNAL OF SPORT AND HEALTH SCIENCE 2024; 13:780-792. [PMID: 38719184 PMCID: PMC11336304 DOI: 10.1016/j.jshs.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 10/26/2023] [Accepted: 11/09/2023] [Indexed: 05/22/2024]
Abstract
The discovery that contracting skeletal muscle generates reactive oxygen species (ROS) was first reported over 40 years ago. The prevailing view in the 1980s was that exercise-induced ROS production promotes oxidation of proteins and lipids resulting in muscle damage. However, a paradigm shift occurred in the 1990s as growing research revealed that ROS are signaling molecules, capable of activating transcriptional activators/coactivators and promoting exercise-induced muscle adaptation. Growing evidence supports the notion that reduction-oxidation (redox) signaling pathways play an important role in the muscle remodeling that occurs in response to endurance exercise training. This review examines the specific role that redox signaling plays in this endurance exercise-induced skeletal muscle adaptation. We begin with a discussion of the primary sites of ROS production in contracting muscle fibers followed by a summary of the antioxidant enzymes involved in the regulation of ROS levels in the cell. We then discuss which redox-sensitive signaling pathways promote endurance exercise-induced muscle adaptation and debate the strength of the evidence supporting the notion that redox signaling plays an essential role in muscle adaptation to endurance exercise training. In hopes of stimulating future research, we highlight several important unanswered questions in this field.
Collapse
Affiliation(s)
- Scott K Powers
- Department of Applied Physiology, University of Florida, Gainesville, FL 32608, USA.
| | - Zsolt Radak
- Research Institute of Sport Science, Hungarian University of Sport Science, Budapest 1123, Hungary
| | - Li Li Ji
- Department of Kinesiology, University of Minnesota, St. Paul, MN 55455, USA
| | - Malcolm Jackson
- Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK
| |
Collapse
|
5
|
Makhnovskii PA, Kukushkina IV, Kurochkina NS, Popov DV. Knockout of Hsp70 genes significantly affects locomotion speed and gene expression in leg skeletal muscles of Drosophila melanogaster. Physiol Genomics 2024; 56:567-575. [PMID: 38881428 DOI: 10.1152/physiolgenomics.00143.2023] [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: 11/20/2023] [Revised: 05/31/2024] [Accepted: 06/11/2024] [Indexed: 06/18/2024] Open
Abstract
The functions of the heat shock protein 70 (Hsp70) genes were studied using a line of Drosophila melanogaster with a knockout of 6 of these genes out of 13. Namely, the effect of knockout of Hsp70 genes on negative geotaxis climbing (locomotor) speed and the ability to adapt to climbing training (0.5-1.5 h/day, 7 days/wk, 19 days) were examined. Seven- and 23-day-old Hsp70- flies demonstrated a comparable reduction (twofold) in locomotor speed and widespread changes in leg skeletal muscle transcriptome (RNA sequencing) compared with w1118 flies. To identify the functions of genes related to decreased locomotor speed, the overlapped differentially expressed genes at both time points were analyzed: the upregulated genes encoded extracellular proteins, regulators of drug metabolism, and the antioxidant response, whereas downregulated genes encoded regulators of carbohydrate metabolism and transmembrane proteins. In addition, in Hsp70- flies, activation of transcription factors related to disruption of the fibril structure and heat shock response (Hsf) was predicted, using the position weight matrix approach. In control flies, adaptation to chronic exercise training was associated mainly with gene response to a single exercise bout, whereas the predicted transcription factors were related to stress/immune (Hsf, NF-κB, etc.) and early gene response. In contrast, Hsp70- flies demonstrated no adaptation to training as well as a significantly impaired gene response to a single exercise bout. In conclusion, the knockout of Hsp70 genes not only reduced physical performance but also disrupted adaptation to chronic physical training, which is associated with changes in the leg skeletal muscle transcriptome and impaired gene response to a single exercise bout.NEW & NOTEWORTHY Knockout of six heat shock protein 70 (Hsp70) genes in Drosophila melanogaster reduced locomotion (climbing) speed that is associated with genotype-specific differences in leg skeletal muscle gene expression. Disrupted adaptation of Hsp70- flies to chronic exercise training is associated with impaired gene response to a single exercise bout.
Collapse
Affiliation(s)
- Pavel A Makhnovskii
- Institute of Biomedical Problems of the Russian Academy of Sciences, Moscow, Russia
| | - Inna V Kukushkina
- Institute of Biomedical Problems of the Russian Academy of Sciences, Moscow, Russia
- Lomonosov Moscow State University, Moscow, Russia
| | - Nadia S Kurochkina
- Institute of Biomedical Problems of the Russian Academy of Sciences, Moscow, Russia
| | - Daniil V Popov
- Institute of Biomedical Problems of the Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
6
|
Braun JL, Fajardo VA. Spaceflight increases sarcoplasmic reticulum Ca 2+ leak and this cannot be counteracted with BuOE treatment. NPJ Microgravity 2024; 10:78. [PMID: 39030182 PMCID: PMC11271499 DOI: 10.1038/s41526-024-00419-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 07/11/2024] [Indexed: 07/21/2024] Open
Abstract
Spending time in a microgravity environment is known to cause significant skeletal muscle atrophy and weakness via muscle unloading, which can be partly attributed to Ca2+ dysregulation. The sarco(endo)plasmic reticulum Ca2+ ATPase (SERCA) pump is responsible for bringing Ca2+ from the cytosol into its storage site, the sarcoplasmic reticulum (SR), at the expense of ATP. We have recently demonstrated that, in the soleus of space-flown mice, the Ca2+ uptake ability of the SERCA pump is severely impaired and this may be attributed to increases in reactive oxygen/nitrogen species (RONS), to which SERCA is highly susceptible. The purpose of this study was therefore to investigate whether treatment with the antioxidant, Manganese(III) meso-tetrakis(N-n-butoxyethylpyridinium-2-yl)porphyrin, MnTnBuOE-2-PyP5+ (BuOE), could attenuate muscle atrophy and SERCA dysfunction. We received soleus muscles from the rodent research 18 mission which had male mice housed on the international space station for 35 days and treated with either saline or BuOE. Spaceflight significantly reduced the soleus:body mass ratio and significantly increased SERCA's ionophore ratio, a measure of SR Ca2+ leak, and 4-HNE content (marker of RONS), none of which could be rescued by BuOE treatment. In conclusion, we find that spaceflight induces significant soleus muscle atrophy and SR Ca2+ leak that cannot be counteracted with BuOE treatment. Future work should investigate alternative therapeutics that are specifically aimed at increasing SERCA activation or reducing Ca2+ leak.
Collapse
Affiliation(s)
- Jessica L Braun
- Department of Kinesiology, Brock University, St. Catharines, ON, Canada
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, Canada
| | - Val A Fajardo
- Department of Kinesiology, Brock University, St. Catharines, ON, Canada.
- Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, Canada.
| |
Collapse
|
7
|
Sato R, Vatic M, Peixoto da Fonseca GW, Anker SD, von Haehling S. Biological basis and treatment of frailty and sarcopenia. Cardiovasc Res 2024:cvae073. [PMID: 38828887 DOI: 10.1093/cvr/cvae073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 11/23/2022] [Accepted: 12/20/2022] [Indexed: 06/05/2024] Open
Abstract
In an ageing society, the importance of maintaining healthy life expectancy has been emphasized. As a result of age-related decline in functional reserve, frailty is a state of increased vulnerability and susceptibility to adverse health outcomes with a serious impact on healthy life expectancy. The decline in skeletal muscle mass and function, also known as sarcopenia, is key in the development of physical frailty. Both frailty and sarcopenia are highly prevalent in patients not only with advanced age but also in patients with illnesses that exacerbate their progression like heart failure (HF), cancer, or dementia, with the prevalence of frailty and sarcopenia in HF patients reaching up to 50-75% and 19.5-47.3%, respectively, resulting in 1.5-3 times higher 1-year mortality. The biological mechanisms of frailty and sarcopenia are multifactorial, complex, and not yet fully elucidated, ranging from DNA damage, proteostasis impairment, and epigenetic changes to mitochondrial dysfunction, cellular senescence, and environmental factors, many of which are further linked to cardiac disease. Currently, there is no gold standard for the treatment of frailty and sarcopenia, however, growing evidence supports that a combination of exercise training and nutritional supplement improves skeletal muscle function and frailty, with a variety of other therapies being devised based on the underlying pathophysiology. In this review, we address the involvement of frailty and sarcopenia in cardiac disease and describe the latest insights into their biological mechanisms as well as the potential for intervention through exercise, diet, and specific therapies.
Collapse
Affiliation(s)
- Ryosuke Sato
- Department of Cardiology and Pneumology, University of Göttingen Medical Center, Robert-Koch-Str. 40, 37075 Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany
| | - Mirela Vatic
- Department of Cardiology and Pneumology, University of Göttingen Medical Center, Robert-Koch-Str. 40, 37075 Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany
| | - Guilherme Wesley Peixoto da Fonseca
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, SP, Brazil
- School of Physical Education and Sport, University of São Paulo, São Paulo, Brazil
| | - Stefan D Anker
- Department of Cardiology (CVK) of German Heart Center Charité; German Centre for Cardiovascular Research (DZHK) partner site Berlin, Charité Universitätsmedizin, Berlin, Germany
- Institute of Heart Diseases, Wroclaw Medical University, Wroclaw, Poland
| | - Stephan von Haehling
- Department of Cardiology and Pneumology, University of Göttingen Medical Center, Robert-Koch-Str. 40, 37075 Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany
| |
Collapse
|
8
|
Membrez M, Migliavacca E, Christen S, Yaku K, Trieu J, Lee AK, Morandini F, Giner MP, Stiner J, Makarov MV, Garratt ES, Vasiloglou MF, Chanvillard L, Dalbram E, Ehrlich AM, Sanchez-Garcia JL, Canto C, Karagounis LG, Treebak JT, Migaud ME, Heshmat R, Razi F, Karnani N, Ostovar A, Farzadfar F, Tay SKH, Sanders MJ, Lillycrop KA, Godfrey KM, Nakagawa T, Moco S, Koopman R, Lynch GS, Sorrentino V, Feige JN. Trigonelline is an NAD + precursor that improves muscle function during ageing and is reduced in human sarcopenia. Nat Metab 2024; 6:433-447. [PMID: 38504132 DOI: 10.1038/s42255-024-00997-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 01/26/2024] [Indexed: 03/21/2024]
Abstract
Mitochondrial dysfunction and low nicotinamide adenine dinucleotide (NAD+) levels are hallmarks of skeletal muscle ageing and sarcopenia1-3, but it is unclear whether these defects result from local changes or can be mediated by systemic or dietary cues. Here we report a functional link between circulating levels of the natural alkaloid trigonelline, which is structurally related to nicotinic acid4, NAD+ levels and muscle health in multiple species. In humans, serum trigonelline levels are reduced with sarcopenia and correlate positively with muscle strength and mitochondrial oxidative phosphorylation in skeletal muscle. Using naturally occurring and isotopically labelled trigonelline, we demonstrate that trigonelline incorporates into the NAD+ pool and increases NAD+ levels in Caenorhabditis elegans, mice and primary myotubes from healthy individuals and individuals with sarcopenia. Mechanistically, trigonelline does not activate GPR109A but is metabolized via the nicotinate phosphoribosyltransferase/Preiss-Handler pathway5,6 across models. In C. elegans, trigonelline improves mitochondrial respiration and biogenesis, reduces age-related muscle wasting and increases lifespan and mobility through an NAD+-dependent mechanism requiring sirtuin. Dietary trigonelline supplementation in male mice enhances muscle strength and prevents fatigue during ageing. Collectively, we identify nutritional supplementation of trigonelline as an NAD+-boosting strategy with therapeutic potential for age-associated muscle decline.
Collapse
Affiliation(s)
- Mathieu Membrez
- Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland
| | | | - Stefan Christen
- Nestlé Institute of Food Safety and Analytical Sciences, Nestlé Research, Lausanne, Switzerland
| | - Keisuke Yaku
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Jennifer Trieu
- Centre for Muscle Research, Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Alaina K Lee
- Centre for Muscle Research, Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Francesco Morandini
- Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Maria Pilar Giner
- Nestlé Institute of Food Safety and Analytical Sciences, Nestlé Research, Lausanne, Switzerland
| | - Jade Stiner
- Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Mikhail V Makarov
- Mitchell Cancer Institute, Department of Pharmacology, F. P. Whiddon College of Medicine, University of South Alabama, Mobile, AL, USA
| | - Emma S Garratt
- Institute of Developmental Sciences, Human Developmental and Health, Faculty of Medicine, University of Southampton, Southampton, UK
- National Institute for Health and Care Research, Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
| | - Maria F Vasiloglou
- Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland
| | - Lucie Chanvillard
- Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Emilie Dalbram
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Amy M Ehrlich
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Carles Canto
- Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Leonidas G Karagounis
- Nestlé Health Science, Translation Research, Lausanne, Switzerland
- Mary MacKillop Institute for Health Research, Australian Catholic University, Melbourne, Victoria, Australia
- Institute of Social and Preventive Medicine, University of Bern, Bern, Switzerland
| | - Jonas T Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Marie E Migaud
- Mitchell Cancer Institute, Department of Pharmacology, F. P. Whiddon College of Medicine, University of South Alabama, Mobile, AL, USA
| | - Ramin Heshmat
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Farideh Razi
- Metabolomics and Genomics Research Center, Endocrinology and Metabolism Molecular-Cellular Science Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Neerja Karnani
- Singapore Institute for Clinical Sciences (A*STAR), Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Afshin Ostovar
- Osteoporosis Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Farshad Farzadfar
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Stacey K H Tay
- KTP-National University Children's Medical Institute, National University Hospital, Singapore, Singapore
| | - Matthew J Sanders
- Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland
| | - Karen A Lillycrop
- Institute of Developmental Sciences, Human Developmental and Health, Faculty of Medicine, University of Southampton, Southampton, UK
- National Institute for Health and Care Research, Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
- Biological Sciences, Faculty of Environmental and Life Sciences, University of Southampton, Southampton, UK
| | - Keith M Godfrey
- Institute of Developmental Sciences, Human Developmental and Health, Faculty of Medicine, University of Southampton, Southampton, UK
- National Institute for Health and Care Research, Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton, UK
- Medical Research Council Lifecourse Epidemiology Centre, University of Southampton, Southampton, UK
| | - Takashi Nakagawa
- Department of Molecular and Medical Pharmacology, Faculty of Medicine, University of Toyama, Toyama, Japan
| | - Sofia Moco
- Nestlé Institute of Food Safety and Analytical Sciences, Nestlé Research, Lausanne, Switzerland
- Division of Molecular and Computational Toxicology, Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute for Molecular and Life Sciences, Vrije Universiteit Amsterdam, Amsterdam, the Netherlands
| | - René Koopman
- Centre for Muscle Research, Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Gordon S Lynch
- Centre for Muscle Research, Department of Anatomy and Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Vincenzo Sorrentino
- Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland.
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Jerome N Feige
- Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland.
- School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.
| |
Collapse
|
9
|
Huang Y, Ji W, Zhang J, Huang Z, Ding A, Bai H, Peng B, Huang K, Du W, Zhao T, Li L. The involvement of the mitochondrial membrane in drug delivery. Acta Biomater 2024; 176:28-50. [PMID: 38280553 DOI: 10.1016/j.actbio.2024.01.027] [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: 10/10/2023] [Revised: 12/23/2023] [Accepted: 01/18/2024] [Indexed: 01/29/2024]
Abstract
Treatment effectiveness and biosafety are critical for disease therapy. Bio-membrane modification facilitates the homologous targeting of drugs in vivo by exploiting unique antibodies or antigens, thereby enhancing therapeutic efficacy while ensuring biosafety. To further enhance the precision of disease treatment, future research should shift focus from targeted cellular delivery to targeted subcellular delivery. As the cellular powerhouses, mitochondria play an indispensable role in cell growth and regulation and are closely involved in many diseases (e.g., cancer, cardiovascular, and neurodegenerative diseases). The double-layer membrane wrapped on the surface of mitochondria not only maintains the stability of their internal environment but also plays a crucial role in fundamental biological processes, such as energy generation, metabolite transport, and information communication. A growing body of evidence suggests that various diseases are tightly related to mitochondrial imbalance. Moreover, mitochondria-targeted strategies hold great potential to decrease therapeutic threshold dosage, minimize side effects, and promote the development of precision medicine. Herein, we introduce the structure and function of mitochondrial membranes, summarize and discuss the important role of mitochondrial membrane-targeting materials in disease diagnosis/treatment, and expound the advantages of mitochondrial membrane-assisted drug delivery for disease diagnosis, treatment, and biosafety. This review helps readers understand mitochondria-targeted therapies and promotes the application of mitochondrial membranes in drug delivery. STATEMENT OF SIGNIFICANCE: Bio-membrane modification facilitates the homologous targeting of drugs in vivo by exploiting unique antibodies or antigens, thereby enhancing therapeutic efficacy while ensuring biosafety. Compared to cell-targeted treatment, targeting of mitochondria for drug delivery offers higher efficiency and improved biosafety and will promote the development of precision medicine. As a natural material, the mitochondrial membrane exhibits excellent biocompatibility and can serve as a carrier for mitochondria-targeted delivery. This review provides an overview of the structure and function of mitochondrial membranes and explores the potential benefits of utilizing mitochondrial membrane-assisted drug delivery for disease treatment and biosafety. The aim of this review is to enhance readers' comprehension of mitochondrial targeted therapy and to advance the utilization of mitochondrial membrane in drug delivery.
Collapse
Affiliation(s)
- Yinghui Huang
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China
| | - Wenhui Ji
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China
| | - Jiaxin Zhang
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an 710072, China
| | - Ze Huang
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China; Future Display Institute in Xiamen, Xiamen 361005, China
| | - Aixiang Ding
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China
| | - Hua Bai
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an 710072, China
| | - Bo Peng
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an 710072, China
| | - Kai Huang
- Future Display Institute in Xiamen, Xiamen 361005, China
| | - Wei Du
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China.
| | - Tingting Zhao
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China.
| | - Lin Li
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China; Future Display Institute in Xiamen, Xiamen 361005, China.
| |
Collapse
|
10
|
Kordowitzki P, Graczyk S, Haghani A, Klutstein M. Oocyte Aging: A Multifactorial Phenomenon in A Unique Cell. Aging Dis 2024; 15:5-21. [PMID: 37307833 PMCID: PMC10796106 DOI: 10.14336/ad.2023.0527] [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: 05/06/2023] [Accepted: 05/27/2023] [Indexed: 06/14/2023] Open
Abstract
The oocyte is considered to be the largest cell in mammalian species. Women hoping to become pregnant face a ticking biological clock. This is becoming increasingly challenging as an increase in life expectancy is accompanied by the tendency to conceive at older ages. With advancing maternal age, the fertilized egg will exhibit lower quality and developmental competence, which contributes to increased chances of miscarriage due to several causes such as aneuploidy, oxidative stress, epigenetics, or metabolic disorders. In particular, heterochromatin in oocytes and with it, the DNA methylation landscape undergoes changes. Further, obesity is a well-known and ever-increasing global problem as it is associated with several metabolic disorders. More importantly, both obesity and aging negatively affect female reproduction. However, among women, there is immense variability in age-related decline of oocytes' quantity, developmental competence, or quality. Herein, the relevance of obesity and DNA-methylation will be discussed as these aspects have a tremendous effect on female fertility, and it is a topic of continuous and widespread interest that has yet to be fully addressed for the mammalian oocyte.
Collapse
Affiliation(s)
- Pawel Kordowitzki
- Department of Preclinical and Basic Sciences, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Torun, Poland.
| | - Szymon Graczyk
- Department of Preclinical and Basic Sciences, Faculty of Biological and Veterinary Sciences, Nicolaus Copernicus University, Torun, Poland.
| | - Amin Haghani
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- Altos Labs, San Diego, CA, USA.
| | - Michael Klutstein
- Institute of Biomedical and Oral Research, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
11
|
Wu Z, Feng C, Hu Y, Zhou Y, Li S, Zhang S, Hu Y, Chen Y, Chao H, Ni Q, Chen M. HALD, a human aging and longevity knowledge graph for precision gerontology and geroscience analyses. Sci Data 2023; 10:851. [PMID: 38040715 PMCID: PMC10692171 DOI: 10.1038/s41597-023-02781-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 11/23/2023] [Indexed: 12/03/2023] Open
Abstract
Human aging is a natural and inevitable biological process that leads to an increased risk of aging-related diseases. Developing anti-aging therapies for aging-related diseases requires a comprehensive understanding of the mechanisms and effects of aging and longevity from a multi-modal and multi-faceted perspective. However, most of the relevant knowledge is scattered in the biomedical literature, the volume of which reached 36 million in PubMed. Here, we presented HALD, a text mining-based human aging and longevity dataset of the biomedical knowledge graph from all published literature related to human aging and longevity in PubMed. HALD integrated multiple state-of-the-art natural language processing (NLP) techniques to improve the accuracy and coverage of the knowledge graph for precision gerontology and geroscience analyses. Up to September 2023, HALD had contained 12,227 entities in 10 types (gene, RNA, protein, carbohydrate, lipid, peptide, pharmaceutical preparations, toxin, mutation, and disease), 115,522 relations, 1,855 aging biomarkers, and 525 longevity biomarkers from 339,918 biomedical articles in PubMed. HALD is available at https://bis.zju.edu.cn/hald .
Collapse
Affiliation(s)
- Zexu Wu
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Cong Feng
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
- The First Affiliated Hospital, Zhejiang University School of Medicine; Institute of Hematology, Zhejiang University, Hangzhou, 310058, China
| | - Yanshi Hu
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yincong Zhou
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
- Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, 314400, China
| | - Sida Li
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Shilong Zhang
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yueming Hu
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Yuhao Chen
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Haoyu Chao
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Qingyang Ni
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Ming Chen
- Department of Bioinformatics, College of Life Sciences, Zhejiang University, Hangzhou, 310058, China.
- The First Affiliated Hospital, Zhejiang University School of Medicine; Institute of Hematology, Zhejiang University, Hangzhou, 310058, China.
- Joint Research Centre for Engineering Biology, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Haining, 314400, China.
| |
Collapse
|
12
|
Dowling P, Swandulla D, Ohlendieck K. Cellular pathogenesis of Duchenne muscular dystrophy: progressive myofibre degeneration, chronic inflammation, reactive myofibrosis and satellite cell dysfunction. Eur J Transl Myol 2023; 33:11856. [PMID: 37846661 PMCID: PMC10811648 DOI: 10.4081/ejtm.2023.11856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 10/10/2023] [Indexed: 10/18/2023] Open
Abstract
Duchenne muscular dystrophy is a highly progressive muscle wasting disease of early childhood and characterized by complex pathophysiological and histopathological changes in the voluntary contractile system, including myonecrosis, chronic inflammation, fat substitution and reactive myofibrosis. The continued loss of functional myofibres and replacement with non-contractile cells, as well as extensive tissue scarring and decline in tissue elasticity, leads to severe skeletal muscle weakness. In addition, dystrophic muscles exhibit a greatly diminished regenerative capacity to counteract the ongoing process of fibre degeneration. In normal muscle tissues, an abundant stem cell pool consisting of satellite cells that are localized between the sarcolemma and basal lamina, provides a rich source for the production of activated myogenic progenitor cells that are involved in efficient myofibre repair and tissue regeneration. Interestingly, the self-renewal of satellite cells for maintaining an essential pool of stem cells in matured skeletal muscles is increased in dystrophin-deficient fibres. However, satellite cell hyperplasia does not result in efficient recovery of dystrophic muscles due to impaired asymmetric cell divisions. The lack of expression of the full-length dystrophin isoform Dp427-M, which is due to primary defects in the DMD gene, appears to affect key regulators of satellite cell polarity causing a reduced differentiation of myogenic progenitors, which are essential for myofibre regeneration. This review outlines the complexity of dystrophinopathy and describes the importance of the pathophysiological role of satellite cell dysfunction. A brief discussion of the bioanalytical usefulness of single cell proteomics for future studies of satellite cell biology is provided.
Collapse
Affiliation(s)
- Paul Dowling
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland; Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare.
| | - Dieter Swandulla
- Institute of Physiology, Medical Faculty, University of Bonn, Bonn.
| | - Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, Maynooth, Co. Kildare, Ireland; Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co. Kildare.
| |
Collapse
|
13
|
Boslem E, Reibe S, Carlessi R, Smeuninx B, Tegegne S, Egan CL, McLennan E, Terry LV, Nobis M, Mu A, Nowell C, Horadagoda N, Mellett NA, Timpson P, Jones M, Denisenko E, Forrest AR, Tirnitz-Parker JE, Meikle PJ, Rose-John S, Karin M, Febbraio MA. Therapeutic blockade of ER stress and inflammation prevents NASH and progression to HCC. SCIENCE ADVANCES 2023; 9:eadh0831. [PMID: 37703359 PMCID: PMC10499313 DOI: 10.1126/sciadv.adh0831] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 08/07/2023] [Indexed: 09/15/2023]
Abstract
The incidence of hepatocellular carcinoma (HCC) is rapidly rising largely because of increased obesity leading to nonalcoholic steatohepatitis (NASH), a known HCC risk factor. There are no approved treatments to treat NASH. Here, we first used single-nucleus RNA sequencing to characterize a mouse model that mimics human NASH-driven HCC, the MUP-uPA mouse fed a high-fat diet. Activation of endoplasmic reticulum (ER) stress and inflammation was observed in a subset of hepatocytes that was enriched in mice that progress to HCC. We next treated MUP-uPA mice with the ER stress inhibitor BGP-15 and soluble gp130Fc, a drug that blocks inflammation by preventing interleukin-6 trans-signaling. Both drugs have progressed to phase 2/3 human clinical trials for other indications. We show that this combined therapy reversed NASH and reduced NASH-driven HCC. Our data suggest that these drugs could provide a potential therapy for NASH progression to HCC.
Collapse
Affiliation(s)
- Ebru Boslem
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Saskia Reibe
- Garvan Institute of Medical Research, Sydney, Australia
| | - Rodrigo Carlessi
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Nedlands, WA 6009, Australia
- Curtin Medical School, Curtin Health Innovation Research Institute, Curtin University, Bentley, WA 6102, Australia
| | - Benoit Smeuninx
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Surafel Tegegne
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Casey L. Egan
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Emma McLennan
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Lauren V. Terry
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Max Nobis
- Garvan Institute of Medical Research, Sydney, Australia
| | - Andre Mu
- Wellcome Sanger Institute, Cambridge, UK
- EMBL's European Bioinformatics Institute, Cambridge UK
| | - Cameron Nowell
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | - Neil Horadagoda
- Faculty of Veterinary Science, University of Sydney, Camden, Australia
| | | | - Paul Timpson
- Garvan Institute of Medical Research, Sydney, Australia
| | - Matthew Jones
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Nedlands, WA 6009, Australia
| | - Elena Denisenko
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Nedlands, WA 6009, Australia
| | - Alistair R. R. Forrest
- Harry Perkins Institute of Medical Research, QEII Medical Centre and Centre for Medical Research, The University of Western Australia, Nedlands, WA 6009, Australia
| | - Janina E. E. Tirnitz-Parker
- Curtin Medical School, Curtin Health Innovation Research Institute, Curtin University, Bentley, WA 6102, Australia
| | - Peter J. Meikle
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Stefan Rose-John
- Department of Biochemistry, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| | - Michael Karin
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, USA
| | - Mark A. Febbraio
- Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Australia
| |
Collapse
|
14
|
Kishore P, Collinet ACT, Brundel BJJM. Prevention of Atrial Fibrillation: Putting Proteostasis Derailment Back on Track. J Clin Med 2023; 12:4352. [PMID: 37445387 DOI: 10.3390/jcm12134352] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/21/2023] [Accepted: 06/23/2023] [Indexed: 07/15/2023] Open
Abstract
Despite the many attempts to treat atrial fibrillation (AF), the most common cardiac tachyarrhythmia in the Western world, the treatment efficacy of AF is still suboptimal. A plausible reason for the suboptimal efficacy is that the current treatments are not directed at the underlying molecular mechanisms that drive AF. Recent discoveries revealed that the derailment of specific molecular proteostasis pathways drive electrical conduction disorders, contractile dysfunction and AF. The degree of this so-called 'electropathology' corresponds to the response to anti-AF treatment. Hence, to develop effective therapies to prevent AF, understanding the molecular mechanisms is of key importance. In this review, we highlight the key modulators of proteostasis derailment and describe the mechanisms that explain how they affect electrical and contractile function in atrial cardiomyocytes and AF. The key modulators of proteostasis derailment include (1) exhaustion of cardioprotective heat shock proteins (HSPs), (2) excessive endoplasmic reticulum (ER) stress and downstream autophagic protein degradation, (3) histone deacetylase 6 (HDAC6)-induced microtubule disruption, (4) activation of DNA damage-PARP1 activation and NAD+ axis and (5) mitochondrial dysfunction. Furthermore, we discuss druggable targets within these pathways that are involved in the prevention of proteostasis derailment, as well as the targets that aid in the recovery from AF. Finally, we will elaborate on the most favorable druggable targets for (future) testing in patients with AF, as well as drugs with potential benefits for AF recovery.
Collapse
Affiliation(s)
- Preetam Kishore
- Physiology, Amsterdam UMC, Vrije Universiteit, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, 1081 HZ Amsterdam, The Netherlands
| | - Amelie C T Collinet
- Physiology, Amsterdam UMC, Vrije Universiteit, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, 1081 HZ Amsterdam, The Netherlands
| | - Bianca J J M Brundel
- Physiology, Amsterdam UMC, Vrije Universiteit, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, 1081 HZ Amsterdam, The Netherlands
| |
Collapse
|
15
|
Mnuskina S, Bauer J, Wirth-Hücking A, Schneidereit D, Nübler S, Ritter P, Cacciani N, Li M, Larsson L, Friedrich O. Single fibre cytoarchitecture in ventilator-induced diaphragm dysfunction (VIDD) assessed by quantitative morphometry second harmonic generation imaging: Positive effects of BGP-15 chaperone co-inducer and VBP-15 dissociative corticosteroid treatment. Front Physiol 2023; 14:1207802. [PMID: 37440999 PMCID: PMC10333583 DOI: 10.3389/fphys.2023.1207802] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 06/01/2023] [Indexed: 07/15/2023] Open
Abstract
Ventilator-induced diaphragm dysfunction (VIDD) is a common sequela of intensive care unit (ICU) treatment requiring mechanical ventilation (MV) and neuromuscular blockade (NMBA). It is characterised by diaphragm weakness, prolonged respirator weaning and adverse outcomes. Dissociative glucocorticoids (e.g., vamorolone, VBP-15) and chaperone co-inducers (e.g., BGP-15) previously showed positive effects in an ICU-rat model. In limb muscle critical illness myopathy, preferential myosin loss prevails, while myofibrillar protein post-translational modifications are more dominant in VIDD. It is not known whether the marked decline in specific force (force normalised to cross-sectional area) is a pure consequence of altered contractility signaling or whether diaphragm weakness also has a structural correlate through sterical remodeling of myofibrillar cytoarchitecture, how quickly it develops, and to which extent VBP-15 or BGP-15 may specifically recover myofibrillar geometry. To address these questions, we performed label-free multiphoton Second Harmonic Generation (SHG) imaging followed by quantitative morphometry in single diaphragm muscle fibres from healthy rats subjected to five or 10 days of MV + NMBA to simulate ICU treatment without underlying confounding pathology (like sepsis). Rats received daily treatment of either Prednisolone, VBP-15, BGP-15 or none. Myosin-II SHG signal intensities, fibre diameters (FD) as well as the parameters of myofibrillar angular parallelism (cosine angle sum, CAS) and in-register of adjacent myofibrils (Vernier density, VD) were computed from SHG images. ICU treatment caused a decline in FD at day 10 as well as a significant decline in CAS and VD from day 5. Vamorolone effectively recovered FD at day 10, while BGP-15 was more effective at day 5. BGP-15 was more effective than VBP-15 in recovering CAS at day 10 although not to control levels. In-register VD levels were restored at day 10 by both compounds. Our study is the first to provide quantitative insights into VIDD-related myofibrillar remodeling unravelled by SHG imaging, suggesting that both VBP-15 and BGP-15 can effectively ameliorate the structure-related dysfunction in VIDD.
Collapse
Affiliation(s)
- Sofia Mnuskina
- Department of Chemical and Biological Engineering (CBI), Institute of Medical Biotechnology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Julian Bauer
- Department of Chemical and Biological Engineering (CBI), Institute of Medical Biotechnology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Anette Wirth-Hücking
- Department of Chemical and Biological Engineering (CBI), Institute of Medical Biotechnology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Dominik Schneidereit
- Department of Chemical and Biological Engineering (CBI), Institute of Medical Biotechnology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Stefanie Nübler
- Department of Chemical and Biological Engineering (CBI), Institute of Medical Biotechnology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Paul Ritter
- Department of Chemical and Biological Engineering (CBI), Institute of Medical Biotechnology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | - Nicola Cacciani
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Meishan Li
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Lars Larsson
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Neuroscience, Clinical Neurophysiology, Karolinska Institutet, Stockholm, Sweden
- Viron Molecular Medicine Institute, Boston, MA, United States
| | - Oliver Friedrich
- Department of Chemical and Biological Engineering (CBI), Institute of Medical Biotechnology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
- Muscle Research Center Erlangen (MURCE), Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
- School of Medical Sciences, University of New South Wales, Kensington Campus, Sydney, NSW, Australia
| |
Collapse
|
16
|
Liu Q, Wang X, Hu Y, Zhao JN, Huang CH, Li T, Zhang BG, He Y, Wu YQ, Zhang ZJ, Wang GP, Liu GP. Acetylated tau exacerbates learning and memory impairment by disturbing with mitochondrial homeostasis. Redox Biol 2023; 62:102697. [PMID: 37037158 PMCID: PMC10114242 DOI: 10.1016/j.redox.2023.102697] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/22/2023] [Accepted: 04/05/2023] [Indexed: 04/09/2023] Open
Abstract
Increased tau acetylation at K274 and K281 has been observed in the brains of Alzheimer's disease (AD) patients and animal models, and mitochondrial dysfunction are noticeable and early features of AD. However, the effect of acetylated tau on mitochondria has been unclear until now. Here, we constructed three type of tau forms, acetylated tau mutant by mutating its K274/K281 into Glutamine (TauKQ) to mimic disease-associated lysine acetylation, the non-acetylation tau mutant by mutating its K274/K281 into Arginine (TauKR) and the wild-type human full-length tau (TauWT). By overexpression of these tau forms in vivo and in vitro, we found that, TauKQ induced more severe cognitive deficits with neuronal loss, dendritic plasticity damage and mitochondrial dysfunctions than TauWT. Unlike TauWT induced mitochondria fusion, TauKQ not only induced mitochondria fission by decreasing mitofusion proteins, but also inhibited mitochondrial biogenesis via reduction of PGC-1a/Nrf1/Tfam levels. TauKR had no significant difference in the cognitive and mitochondrial abnormalities compared with TauWT. Treatment with BGP-15 rescued impaired learning and memory by attenuation of mitochondrial dysfunction, neuronal loss and dendritic complexity damage, which caused by TauKQ. Our data suggested that, acetylation at K274/281 was an important post translational modification site for tau neurotoxicity, and BGP-15 is a potential therapeutic drug for AD.
Collapse
Affiliation(s)
- Qian Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xin Wang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China
| | - Yu Hu
- Department of Pathology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Jun-Ning Zhao
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Chun-Hui Huang
- Guangdong Province Key Laboratory of Pharmacodynamic, Constituents of TCM and New Drugs Research, Institute of New Drug Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Ting Li
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Bing-Ge Zhang
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ye He
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yan-Qing Wu
- Department of Neurology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, China
| | - Zai-Jun Zhang
- Guangdong Province Key Laboratory of Pharmacodynamic, Constituents of TCM and New Drugs Research, Institute of New Drug Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China.
| | - Guo-Ping Wang
- Department of Pathology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Gong-Ping Liu
- Department of Pathophysiology, School of Basic Medicine, Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, China.
| |
Collapse
|
17
|
Younger DS. Childhood muscular dystrophies. HANDBOOK OF CLINICAL NEUROLOGY 2023; 195:461-496. [PMID: 37562882 DOI: 10.1016/b978-0-323-98818-6.00024-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
Infancy- and childhood-onset muscular dystrophies are associated with a characteristic distribution and progression of motor dysfunction. The underlying causes of progressive childhood muscular dystrophies are heterogeneous involving diverse genetic pathways and genes that encode proteins of the plasma membrane, extracellular matrix, sarcomere, and nuclear membrane components. The prototypical clinicopathological features in an affected child may be adequate to fully distinguish it from other likely diagnoses based on four common features: (1) weakness and wasting of pelvic-femoral and scapular muscles with involvement of heart muscle; (2) elevation of serum muscle enzymes in particular serum creatine kinase; (3) necrosis and regeneration of myofibers; and (4) molecular neurogenetic assessment particularly utilizing next-generation sequencing of the genome of the likeliest candidates genes in an index case or family proband. A number of different animal models of therapeutic strategies have been developed for gene transfer therapy, but so far these techniques have not yet entered clinical practice. Treatment remains for the most part symptomatic with the goal of ameliorating locomotor and cardiorespiratory manifestations of the disease.
Collapse
Affiliation(s)
- David S Younger
- Department of Clinical Medicine and Neuroscience, CUNY School of Medicine, New York, NY, United States; Department of Medicine, Section of Internal Medicine and Neurology, White Plains Hospital, White Plains, NY, United States.
| |
Collapse
|
18
|
Valentim M, Brahmbhatt A, Tupling A. Skeletal and cardiac muscle calcium transport regulation in health and disease. Biosci Rep 2022; 42:BSR20211997. [PMID: 36413081 PMCID: PMC9744722 DOI: 10.1042/bsr20211997] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 11/04/2022] [Accepted: 11/22/2022] [Indexed: 11/23/2022] Open
Abstract
In healthy muscle, the rapid release of calcium ions (Ca2+) with excitation-contraction (E-C) coupling, results in elevations in Ca2+ concentrations which can exceed 10-fold that of resting values. The sizable transient changes in Ca2+ concentrations are necessary for the activation of signaling pathways, which rely on Ca2+ as a second messenger, including those involved with force generation, fiber type distribution and hypertrophy. However, prolonged elevations in intracellular Ca2+ can result in the unwanted activation of Ca2+ signaling pathways that cause muscle damage, dysfunction, and disease. Muscle employs several calcium handling and calcium transport proteins that function to rapidly return Ca2+ concentrations back to resting levels following contraction. This review will detail our current understanding of calcium handling during the decay phase of intracellular calcium transients in healthy skeletal and cardiac muscle. We will also discuss how impairments in Ca2+ transport can occur and how mishandling of Ca2+ can lead to the pathogenesis and/or progression of skeletal muscle myopathies and cardiomyopathies.
Collapse
Affiliation(s)
- Mark A. Valentim
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, Ontario, N2L 3G1, Canada
| | - Aditya N. Brahmbhatt
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, Ontario, N2L 3G1, Canada
| | - A. Russell Tupling
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, Ontario, N2L 3G1, Canada
| |
Collapse
|
19
|
Al-Azab M, Safi M, Idiiatullina E, Al-Shaebi F, Zaky MY. Aging of mesenchymal stem cell: machinery, markers, and strategies of fighting. Cell Mol Biol Lett 2022; 27:69. [PMID: 35986247 PMCID: PMC9388978 DOI: 10.1186/s11658-022-00366-0] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 07/18/2022] [Indexed: 02/08/2023] Open
Abstract
Human mesenchymal stem cells (MSCs) are primary multipotent cells capable of differentiating into osteocytes, chondrocytes, and adipocytes when stimulated under appropriate conditions. The role of MSCs in tissue homeostasis, aging-related diseases, and cellular therapy is clinically suggested. As aging is a universal problem that has large socioeconomic effects, an improved understanding of the concepts of aging can direct public policies that reduce its adverse impacts on the healthcare system and humanity. Several studies of aging have been carried out over several years to understand the phenomenon and different factors affecting human aging. A reduced ability of adult stem cell populations to reproduce and regenerate is one of the main contributors to the human aging process. In this context, MSCs senescence is a major challenge in front of cellular therapy advancement. Many factors, ranging from genetic and metabolic pathways to extrinsic factors through various cellular signaling pathways, are involved in regulating the mechanism of MSC senescence. To better understand and reverse cellular senescence, this review highlights the underlying mechanisms and signs of MSC cellular senescence, and discusses the strategies to combat aging and cellular senescence.
Collapse
|
20
|
Choi MC, Jo J, Lee M, Park J, Yao TP, Park Y. Cathelicidin-related antimicrobial peptide mediates skeletal muscle degeneration caused by injury and Duchenne muscular dystrophy in mice. J Cachexia Sarcopenia Muscle 2022; 13:3091-3105. [PMID: 36059045 PMCID: PMC9745559 DOI: 10.1002/jcsm.13065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 07/06/2022] [Accepted: 07/20/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Cathelicidin, an antimicrobial peptide, plays a key role in regulating bacterial killing and innate immunity; however, its role in skeletal muscle function is unknown. We investigated the potential role of cathelicidin in skeletal muscle pathology resulting from acute injury and Duchenne muscular dystrophy (DMD) in mice. METHODS Expression changes and muscular localization of mouse cathelicidin-related antimicrobial peptide (Cramp) were examined in the skeletal muscle of normal mice treated with chemicals (cardiotoxin and BaCl2 ) or in dystrophic muscle of DMD mouse models (mdx, mdx/Utrn+/- and mdx/Utrn-/- ). Cramp penetration into myofibres and effects on muscle damage were studied by treating synthetic peptides to mouse skeletal muscles or C2C12 myotubes. Cramp knockout (KO) mice and mdx/Utrn/Cramp KO lines were used to determine whether Cramp mediates muscle degeneration. Muscle pathophysiology was assessed by histological methods, serum analysis, grip strength and lifespan. Molecular factors targeted by Cramp were identified by the pull-down assay and proteomic analysis. RESULTS In response to acute muscle injury, Cramp was activated in muscle-infiltrating neutrophils and internalized into myofibres. Cramp treatments of mouse skeletal muscles or C2C12 myotubes resulted in muscle degeneration and myotube damage, respectively. Genetic ablation of Cramp reduced neutrophil infiltration and ameliorated muscle pathology, such as fibre size (P < 0.001; n = 6) and fibrofatty infiltration (P < 0.05). Genetic reduction of Cramp in mdx/Utrn+/- mice not only attenuated muscle damage (35%, P < 0.05; n = 9-10), myonecrosis (53%, P < 0.05), inflammation (37-65%, P < 0.01) and fibrosis (14%, P < 0.05) but also restored muscle fibre size (14%, P < 0.05) and muscle force (18%, P < 0.05). Reducing Cramp levels led to a 63% (male, P < 0.05; n = 10-14) and a 124% (female, P < 0.001; n = 20) increase in the lifespan of mdx/Utrn-/- mice. Proteomic and mechanistic studies revealed that Cramp cross-talks with Ca2+ signalling in skeletal muscle through sarcoplasmic/endoplasmic reticulum Ca2+ -ATPase1 (SERCA1). Cramp binds and inactivates SERCA1, leading to the activation of Ca2+ -dependent calpain proteases that exacerbate DMD progression. CONCLUSIONS These findings identify Cramp as an immune cell-derived regulator of skeletal muscle degeneration and provide a potential therapeutic target for DMD.
Collapse
Affiliation(s)
- Moon-Chang Choi
- Department of Biomedical Science, Chosun University, Gwangju, South Korea
| | - Jiwon Jo
- Department of Biomedical Science, Chosun University, Gwangju, South Korea
| | - Myeongjin Lee
- Department of Biomedical Science, Chosun University, Gwangju, South Korea
| | - Jonggwan Park
- Department of Bioinformatics, Kongju National University, Kongju, South Korea
| | - Tso-Pang Yao
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Yoonkyung Park
- Department of Biomedical Science, Chosun University, Gwangju, South Korea
| |
Collapse
|
21
|
Morera C, Kim J, Paredes-Redondo A, Nobles M, Rybin D, Moccia R, Kowala A, Meng J, Garren S, Liu P, Morgan JE, Muntoni F, Christoforou N, Owens J, Tinker A, Lin YY. CRISPR-mediated correction of skeletal muscle Ca 2+ handling in a novel DMD patient-derived pluripotent stem cell model. Neuromuscul Disord 2022; 32:908-922. [PMID: 36418198 DOI: 10.1016/j.nmd.2022.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 10/28/2022] [Accepted: 10/30/2022] [Indexed: 11/05/2022]
Abstract
Mutations in the dystrophin gene cause the most common and currently incurable Duchenne muscular dystrophy (DMD) characterized by progressive muscle wasting. Although abnormal Ca2+ handling is a pathological feature of DMD, mechanisms underlying defective Ca2+ homeostasis remain unclear. Here we generate a novel DMD patient-derived pluripotent stem cell (PSC) model of skeletal muscle with an isogenic control using clustered regularly interspaced short palindromic repeat (CRISPR)-mediated precise gene correction. Transcriptome analysis identifies dysregulated gene sets in the absence of dystrophin, including genes involved in Ca2+ handling, excitation-contraction coupling and muscle contraction. Specifically, analysis of intracellular Ca2+ transients and mathematical modeling of Ca2+ dynamics reveal significantly reduced cytosolic Ca2+ clearance rates in DMD-PSC derived myotubes. Pharmacological assays demonstrate Ca2+ flux in myotubes is determined by both intracellular and extracellular sources. DMD-PSC derived myotubes display significantly reduced velocity of contractility. Compared with a non-isogenic wildtype PSC line, these pathophysiological defects could be rescued by CRISPR-mediated precise gene correction. Our study provides new insights into abnormal Ca2+ homeostasis in DMD and suggests that Ca2+ signaling pathways amenable to pharmacological modulation are potential therapeutic targets. Importantly, we have established a human physiology-relevant in vitro model enabling rapid pre-clinical testing of potential therapies for DMD.
Collapse
Affiliation(s)
- Cristina Morera
- Centre for Genomics and Child Health, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London E1 2AT, United Kingdom; Stem Cell Laboratory, National Bowel Research Centre, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, 2 Newark Street, London E1 2AT, United Kingdom
| | - Jihee Kim
- Centre for Genomics and Child Health, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London E1 2AT, United Kingdom; Stem Cell Laboratory, National Bowel Research Centre, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, 2 Newark Street, London E1 2AT, United Kingdom
| | - Amaia Paredes-Redondo
- Centre for Genomics and Child Health, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London E1 2AT, United Kingdom; Stem Cell Laboratory, National Bowel Research Centre, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, 2 Newark Street, London E1 2AT, United Kingdom; Centre for Predictive in vitro Model, Queen Mary University of London, Mile End Road, London E1 4NS, United Kingdom
| | - Muriel Nobles
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom
| | - Denis Rybin
- Rare Disease Research Unit, Pfizer, 610 Main Street, Cambridge, MA 02139, USA
| | - Robert Moccia
- Rare Disease Research Unit, Pfizer, 610 Main Street, Cambridge, MA 02139, USA
| | - Anna Kowala
- Centre for Genomics and Child Health, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London E1 2AT, United Kingdom; Stem Cell Laboratory, National Bowel Research Centre, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, 2 Newark Street, London E1 2AT, United Kingdom; Centre for Predictive in vitro Model, Queen Mary University of London, Mile End Road, London E1 4NS, United Kingdom
| | - Jinhong Meng
- UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, United Kingdom
| | - Seth Garren
- Rare Disease Research Unit, Pfizer, 610 Main Street, Cambridge, MA 02139, USA
| | - Pentao Liu
- School of Biomedical Sciences, Stem Cell and Regenerative Medicine Consortium, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong, China
| | - Jennifer E Morgan
- UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, United Kingdom; NIHR Biomedical Research Centre at Great Ormond Street Hospital, Great Ormond Street, London, United Kingdom
| | - Francesco Muntoni
- UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, United Kingdom; NIHR Biomedical Research Centre at Great Ormond Street Hospital, Great Ormond Street, London, United Kingdom
| | | | - Jane Owens
- Rare Disease Research Unit, Pfizer, 610 Main Street, Cambridge, MA 02139, USA
| | - Andrew Tinker
- William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom
| | - Yung-Yao Lin
- Centre for Genomics and Child Health, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London E1 2AT, United Kingdom; Stem Cell Laboratory, National Bowel Research Centre, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, 2 Newark Street, London E1 2AT, United Kingdom; Centre for Predictive in vitro Model, Queen Mary University of London, Mile End Road, London E1 4NS, United Kingdom.
| |
Collapse
|
22
|
Gonçalves RSDSA, Maciel ÁCC, Rolland Y, Vellas B, de Souto Barreto P. Frailty biomarkers under the perspective of geroscience: A narrative review. Ageing Res Rev 2022; 81:101737. [PMID: 36162706 DOI: 10.1016/j.arr.2022.101737] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 01/31/2023]
Abstract
Cellular and molecular aging biomarkers might contribute to identify at-risk individuals for frailty before overt clinical manifestations appear. Although studies on the associations of aging biomarkers and frailty exist, no investigation has gathered this information using a structured framework for identifying aging biomarkers; as a result, the evidence on frailty and aging biomarkers is diffuse and incomplete. Therefore, this narrative review aimed to gather information on the associations of the hallmarks of aging and frailty under the perspective of geroscience. The literature on human studies on this topic is sparse and mainly composed of cross-sectional investigations performed in small study samples. The main putative aging biomarkers associated to frailty were: mitochondrial DNA copy number (genomic instability and mitochondrial dysfunction), telomere length (telomere attrition), global DNA methylation (epigenetic alterations), Hsp70 and Hsp72 (loss of proteostasis), IGF-1 and SIRT1 (deregulated nutrient-sensing), GDF-15 (mitochondrial dysfunction, cellular senescence and altered intercellular communication), CD4 + and CD8 + cell percentages (cellular senescence), circulating osteogenic progenitor (COP) cells (stem cell exhaustion), and IL-6, CRP and TNF-alpha (altered intercellular communication). IGF-1, SIRT1, GDF-15, IL-6, CRP and TNF-alpha presented more evidence among these biomarkers, highlighting the importance of inflammation and nutrient sensing on frailty. Further longitudinal studies investigating biomarkers across the hallmarks of aging would provide valuable information on this topic.
Collapse
Affiliation(s)
| | | | - Yves Rolland
- Gerontopole of Toulouse, Institute of Aging, Toulouse University Hospital (CHU Toulouse), Toulouse, France; CERPOP, Inserm 1295, Université de Toulouse, UPS, Toulouse, France.
| | - Bruno Vellas
- Gerontopole of Toulouse, Institute of Aging, Toulouse University Hospital (CHU Toulouse), Toulouse, France; CERPOP, Inserm 1295, Université de Toulouse, UPS, Toulouse, France.
| | - Philipe de Souto Barreto
- Gerontopole of Toulouse, Institute of Aging, Toulouse University Hospital (CHU Toulouse), Toulouse, France; CERPOP, Inserm 1295, Université de Toulouse, UPS, Toulouse, France.
| |
Collapse
|
23
|
Cleverdon RE, Braun JL, Geromella MS, Whitley KC, Marko DM, Hamstra SI, Roy BD, MacPherson RE, Fajardo VA. Sarco(endo)plasmic reticulum Ca2+-ATPase function is impaired in skeletal and cardiac muscles from young DBA/2J mdx mice. iScience 2022; 25:104972. [PMID: 36093052 PMCID: PMC9459692 DOI: 10.1016/j.isci.2022.104972] [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: 11/23/2021] [Revised: 06/16/2022] [Accepted: 08/15/2022] [Indexed: 11/18/2022] Open
Abstract
The DBA/2J (D2) mdx mouse is a more severe model of Duchenne muscular dystrophy when compared to the traditional C57BL/10 (C57) mdx mouse. Here, we questioned whether sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA) function would differ in muscles from young D2 and C57 mdx mice. Both D2 and C57 mdx mice exhibited signs of impaired Ca2+ uptake in the gastrocnemius, diaphragm, and left ventricle; however, the level of impairment was more severe in D2 mdx mice. Reductions in maximal SERCA activity were also more prominent in the D2 mdx gastrocnemius and diaphragm when compared to those from C57 mdx mice; however, there were no differences detected in the left ventricle. Across all muscles, D2 mdx mice had the highest levels of oxidative stress as indicated by protein nitrosylation and/or nitration. In conclusion, our study shows that SERCA function is more impaired in young D2 mdx mice compared with age-matched C57 mdx mice. Ca2+ uptake is severely impaired in muscles from young DBA/2J (D2) mdx mice Maximal SERCA activity is lowered to a greater degree in muscles from D2 mdx mice Muscles from young D2 mdx mice have higher levels of oxidative/nitrosative stress Worsened SERCA function may contribute to worsened muscle pathology in D2 mdx mice
Collapse
|
24
|
García-Castañeda M, Michelucci A, Zhao N, Malik S, Dirksen RT. Postdevelopmental knockout of Orai1 improves muscle pathology in a mouse model of Duchenne muscular dystrophy. J Gen Physiol 2022; 154:213383. [PMID: 35939054 PMCID: PMC9365874 DOI: 10.1085/jgp.202213081] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 07/07/2022] [Indexed: 11/20/2022] Open
Abstract
Duchenne muscular dystrophy (DMD), an X-linked disorder caused by loss-of-function mutations in the dystrophin gene, is characterized by progressive muscle degeneration and weakness. Enhanced store-operated Ca2+ entry (SOCE), a Ca2+ influx mechanism coordinated by STIM1 sensors of luminal Ca2+ within the sarcoplasmic reticulum (SR) and Ca2+-permeable Orai1 channels in the sarcolemma, is proposed to contribute to Ca2+-mediated muscle damage in DMD. To directly determine the impact of Orai1-dependent SOCE on the dystrophic phenotype, we crossed mdx mice with tamoxifen-inducible, muscle-specific Orai1 knockout mice (mdx-Orai1 KO mice). Both constitutive and SOCE were significantly increased in flexor digitorum brevis fibers from mdx mice, while SOCE was absent in fibers from both Orai1 KO and mdx-Orai1 KO mice. Compared with WT mice, fibers from mdx mice exhibited (1) increased resting myoplasmic Ca2+ levels, (2) reduced total releasable Ca2+ store content, and (3) a prolonged rate of electrically evoked Ca2+ transient decay. These effects were partially normalized in fibers from mdx-Orai1 KO mice. Intact extensor digitorum longus muscles from mdx mice exhibited a significant reduction of maximal specific force, which was rescued in muscles from mdx-Orai1 KO mice. Finally, during exposure to consecutive eccentric contractions, muscles from mdx mice displayed a more pronounced decline in specific force compared with that of WT mice, which was also significantly attenuated by Orai1 ablation. Together, these results indicate that enhanced Orai1-dependent SOCE exacerbates the dystrophic phenotype and that Orai1 deficiency improves muscle pathology by both normalizing Ca2+ homeostasis and promoting sarcolemmal integrity/stability.
Collapse
Affiliation(s)
- Maricela García-Castañeda
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Antonio Michelucci
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY,Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Nan Zhao
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Sundeep Malik
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY
| | - Robert T. Dirksen
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY
| |
Collapse
|
25
|
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
|
26
|
Yadav R, Devi SS, Oswalia J, Ramalingam S, Arya R. Role of HSP70 chaperone in protein aggregate phenomenon of GNE mutant cells: Therapeutic lead for GNE Myopathy. Int J Biochem Cell Biol 2022; 149:106258. [PMID: 35777599 DOI: 10.1016/j.biocel.2022.106258] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 06/04/2022] [Accepted: 06/25/2022] [Indexed: 10/17/2022]
Abstract
Limited treatment options and research in understanding the pathomechanisms of rare diseases has raised concerns about their therapeutic development. One such poorly understood ultra-rare neuromuscular disorder is GNE Myopathy (GNEM) which is caused due to mutation in key sialic acid biosynthetic enzyme, GNE. Treatment with sialic acid or its derivatives/precursors slows the disease progression, but curative strategies need to be explored further. Pathologically, muscle biopsy samples of GNEM patients reveal rimmed vacuole formation due to aggregation of β-amyloid, Tau, presenilin proteins with unknown mechanism. The present study aims to understand the mechanism of protein aggregate formation in GNE mutant cells to decipher role of chaperones in disease phenotype. The pathologically relevant GNE mutations expressed as recombinant proteins in HEK cells was used as a model system for GNEM to estimate extent of protein aggregation. We identified HSP70, a chaperone, as binding partner of GNE. Downregulation of HSP70 with altered BAG3, JNK, BAX expression levels was observed in GNE mutant cells. The cell apoptosis was observed in GNE mutation specific manner. An activator of HSP70 chaperone, BGP-15, rescued the phenotypic defects due to GNE mutation, thereby, reducing protein aggregation significantly. The results were further validated in rat skeletal muscle cell lines carrying single Gne allele. Our study suggests that HSP70 activators can be a promising therapeutic target in the treatment of ultra-rare GNE Myopathy disease.
Collapse
Affiliation(s)
- Rashmi Yadav
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India.
| | | | - Jyoti Oswalia
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India.
| | | | - Ranjana Arya
- School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India; Special Center for Systems Medicine, Jawaharlal Nehru University, New Delhi 110067, India.
| |
Collapse
|
27
|
Schiemann R, Buhr A, Cordes E, Walter S, Heinisch JJ, Ferrero P, Milting H, Paululat A, Meyer H. Neprilysins regulate muscle contraction and heart function via cleavage of SERCA-inhibitory micropeptides. Nat Commun 2022; 13:4420. [PMID: 35906206 PMCID: PMC9338278 DOI: 10.1038/s41467-022-31974-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 07/06/2022] [Indexed: 12/26/2022] Open
Abstract
Muscle contraction depends on strictly controlled Ca2+ transients within myocytes. A major player maintaining these transients is the sarcoplasmic/endoplasmic reticulum Ca2+ ATPase, SERCA. Activity of SERCA is regulated by binding of micropeptides and impaired expression or function of these peptides results in cardiomyopathy. To date, it is not known how homeostasis or turnover of the micropeptides is regulated. Herein, we find that the Drosophila endopeptidase Neprilysin 4 hydrolyzes SERCA-inhibitory Sarcolamban peptides in membranes of the sarcoplasmic reticulum, thereby ensuring proper regulation of SERCA. Cleavage is necessary and sufficient to maintain homeostasis and function of the micropeptides. Analyses on human Neprilysin, sarcolipin, and ventricular cardiomyocytes indicates that the regulatory mechanism is evolutionarily conserved. By identifying a neprilysin as essential regulator of SERCA activity and Ca2+ homeostasis in cardiomyocytes, these data contribute to a more comprehensive understanding of the complex mechanisms that control muscle contraction and heart function in health and disease.
Collapse
Affiliation(s)
- Ronja Schiemann
- Department of Zoology & Developmental Biology, Osnabrück University, 49076, Osnabrück, Germany
| | - Annika Buhr
- Department of Zoology & Developmental Biology, Osnabrück University, 49076, Osnabrück, Germany
| | - Eva Cordes
- Department of Zoology & Developmental Biology, Osnabrück University, 49076, Osnabrück, Germany
| | - Stefan Walter
- Center of Cellular Nanoanalytics Osnabrück - CellNanOs, 49076, Osnabrück, Germany
| | - Jürgen J Heinisch
- Center of Cellular Nanoanalytics Osnabrück - CellNanOs, 49076, Osnabrück, Germany.,Department of Genetics, Osnabrück University, 49076, Osnabrück, Germany
| | - Paola Ferrero
- Center for Cardiovascular Research - CONICET/National University of La Plata, 1900, La Plata, Argentina
| | - Hendrik Milting
- Heart & Diabetes Center NRW, University of Bochum, Erich & Hanna Klessmann-Institute for Cardiovascular Research and Development, 32545, Bad Oeynhausen, Germany
| | - Achim Paululat
- Department of Zoology & Developmental Biology, Osnabrück University, 49076, Osnabrück, Germany.,Center of Cellular Nanoanalytics Osnabrück - CellNanOs, 49076, Osnabrück, Germany
| | - Heiko Meyer
- Department of Zoology & Developmental Biology, Osnabrück University, 49076, Osnabrück, Germany. .,Center of Cellular Nanoanalytics Osnabrück - CellNanOs, 49076, Osnabrück, Germany.
| |
Collapse
|
28
|
Umehara T, Winstanley YE, Andreas E, Morimoto A, Williams EJ, Smith KM, Carroll J, Febbraio MA, Shimada M, Russell DL, Robker RL. Female reproductive life span is extended by targeted removal of fibrotic collagen from the mouse ovary. SCIENCE ADVANCES 2022; 8:eabn4564. [PMID: 35714185 PMCID: PMC9205599 DOI: 10.1126/sciadv.abn4564] [Citation(s) in RCA: 106] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
The female ovary contains a finite number of oocytes, and their release at ovulation becomes sporadic and disordered with aging and with obesity, leading to loss of fertility. Understanding the molecular defects underpinning this pathology is essential as age of childbearing and obesity rates increase globally. We identify that fibrosis within the ovarian stromal compartment is an underlying mechanism responsible for impaired oocyte release, which is initiated by mitochondrial dysfunction leading to diminished bioenergetics, oxidative damage, inflammation, and collagen deposition. Furthermore, antifibrosis drugs (pirfenidone and BGP-15) eliminate fibrotic collagen and restore ovulation in reproductively old and obese mice, in association with dampened M2 macrophage polarization and up-regulated MMP13 protease. This is the first evidence that ovarian fibrosis is reversible and indicates that drugs targeting mitochondrial metabolism may be a viable therapeutic strategy for women with metabolic disorders or advancing age to maintain ovarian function and extend fertility.
Collapse
Affiliation(s)
- Takashi Umehara
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
| | - Yasmyn E. Winstanley
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - Eryk Andreas
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - Atsushi Morimoto
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - Elisha J. Williams
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - Kirsten M. Smith
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - John Carroll
- Development and Stem Cells Program and Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Mark A. Febbraio
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Masayuki Shimada
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
| | - Darryl L. Russell
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
| | - Rebecca L. Robker
- Robinson Research Institute, School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia
- Development and Stem Cells Program and Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Corresponding author.
| |
Collapse
|
29
|
Zhang XZ, Xie WQ, Chen L, Xu GD, Wu L, Li YS, Wu YX. Blood Flow Restriction Training for the Intervention of Sarcopenia: Current Stage and Future Perspective. Front Med (Lausanne) 2022; 9:894996. [PMID: 35770017 PMCID: PMC9234289 DOI: 10.3389/fmed.2022.894996] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 05/20/2022] [Indexed: 11/23/2022] Open
Abstract
Sarcopenia is a geriatric syndrome that is characterized by a progressive and generalized skeletal muscle disorder and can be associated with many comorbidities, including obesity, diabetes, and fracture. Its definitions, given by the AWGS and EWGSOP, are widely used. Sarcopenia is measured by muscle strength, muscle quantity or mass and physical performance. Currently, the importance and urgency of sarcopenia have grown. The application of blood flow restriction (BFR) training has received increased attention in managing sarcopenia. BFR is accomplished using a pneumatic cuff on the proximal aspect of the exercising limb. Two main methods of exercise, aerobic exercise and resistance exercise, have been applied with BFR in treating sarcopenia. Both methods can increase muscle mass and muscle strength to a certain extent. Intricate mechanisms are involved during BFRT. Currently, the presented mechanisms mainly include responses in the blood vessels and related hormones, such as growth factors, tissue hypoxia-related factors and recruitment of muscle fiber as well as muscle satellite cells. These mechanisms contribute to the positive balance of skeletal muscle synthesis, which in turn mitigates sarcopenia. As a more suited and more effective way of treating sarcopenia and its comorbidities, BFRT can serve as an alternative to traditional exercise for people who have marked physical limitations or even show superior outcomes under low loads. However, the possibility of causing stress or muscle damage must be considered. Cuff size, pressure, training load and other variables can affect the outcome of sarcopenia, which must also be considered. Thoroughly studying these factors can help to better determine an ideal BFRT scheme and better manage sarcopenia and its associated comorbidities. As a well-tolerated and novel form of exercise, BFRT offers more potential in treating sarcopenia and involves deeper insights into the function and regulation of skeletal muscle.
Collapse
Affiliation(s)
- Xu-zhi Zhang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Wen-qing Xie
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Lin Chen
- Department of Health and Kinesiology, School of Physical Education, Jianghan University, Wuhan, China
| | - Guo-dong Xu
- Department of Health and Kinesiology, School of Physical Education, Jianghan University, Wuhan, China
| | - Li Wu
- Department of Health and Kinesiology, School of Physical Education, Jianghan University, Wuhan, China
| | - Yu-sheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Yu-sheng Li
| | - Yu-xiang Wu
- Department of Health and Kinesiology, School of Physical Education, Jianghan University, Wuhan, China
- Yu-xiang Wu
| |
Collapse
|
30
|
Alves FM, Kysenius K, Caldow MK, Hardee JP, Chung JD, Trieu J, Hare DJ, Crouch PJ, Ayton S, Bush AI, Lynch GS, Koopman R. Iron overload and impaired iron handling contribute to the dystrophic pathology in models of Duchenne muscular dystrophy. J Cachexia Sarcopenia Muscle 2022; 13:1541-1553. [PMID: 35249268 PMCID: PMC9178167 DOI: 10.1002/jcsm.12950] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 01/14/2022] [Accepted: 01/23/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Oxidative stress is implicated in the pathophysiology of Duchenne muscular dystrophy (DMD, caused by mutations in the dystrophin gene), which is the most common and severe of the muscular dystrophies. To our knowledge, the distribution of iron, an important modulator of oxidative stress, has not been assessed in DMD. We tested the hypotheses that iron accumulation occurs in mouse models of DMD and that modulation of iron through the diet or chelation could modify disease severity. METHODS We assessed iron distribution and total elemental iron using LA-ICP-MS on skeletal muscle cross-sections of 8-week-old Bl10 control mice and dystrophic mdx mice (with moderate dystrophy) and dystrophin/utrophin-null mice (dko, with severe dystrophy). In addition, mdx mice (4 weeks) were treated with either an iron chelator (deferiprone 150 mg/kg/day) or iron-enriched feed (containing 1% added iron as carbonyl iron). Immunoblotting was used to determine the abundance of iron- and mitochondria-related proteins. (Immuno)histochemical and mRNA assessments of fibrosis and inflammation were also performed. RESULTS We observed a significant increase in total elemental iron in hindlimb muscles of dko mice (+50%, P < 0.05) and in the diaphragm of mdx mice (+80%, P < 0.05), with both tissues exhibiting severe pathology. Iron dyshomeostasis was further evidenced by an increase in the storage protein ferritin (dko: +39%, P < 0.05) and ferroportin compared with Bl10 control mice (mdx: +152% and dko: +175%, P < 0.05). Despite having features of iron overload, dystrophic muscles had lower protein expression of ALAS-1, the rate-limiting enzyme for haem synthesis (dko -44%, P < 0.05), and the haem-containing protein myoglobin (dko -54%, P < 0.05). Deferiprone treatment tended to decrease muscle iron levels in mdx mice (-30%, P < 0.1), which was associated with lower oxidative stress and fibrosis, but suppressed haem-containing proteins and mitochondrial content. Increasing iron via dietary intervention elevated total muscle iron (+25%, P < 0.05) but did not aggravate the pathology. CONCLUSIONS Muscles from dystrophic mice have increased iron levels and dysregulated iron-related proteins that are associated with dystrophic pathology. Muscle iron levels were manipulated by iron chelation and iron enriched feed. Iron chelation reduced fibrosis and reactive oxygen species (ROS) but also suppressed haem-containing proteins and mitochondrial activity. Conversely, iron supplementation increased ferritin and haem-containing proteins but did not alter ROS, fibrosis, or mitochondrial activity. Further studies are required to investigate the contribution of impaired ferritin breakdown in the dysregulation of iron homeostasis in DMD.
Collapse
Affiliation(s)
- Francesca M Alves
- Centre for Muscle Research, Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia.,Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Kai Kysenius
- Department of Biochemistry and Pharmacology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Marissa K Caldow
- Centre for Muscle Research, Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Justin P Hardee
- Centre for Muscle Research, Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Jin D Chung
- Centre for Muscle Research, Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Jennifer Trieu
- Centre for Muscle Research, Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Dominic J Hare
- Monash eResearch Centre, Monash University, Clayton, Victoria, Australia
| | - Peter J Crouch
- Department of Biochemistry and Pharmacology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Scott Ayton
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Ashley I Bush
- Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, Victoria, Australia
| | - Gordon S Lynch
- Centre for Muscle Research, Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| | - René Koopman
- Centre for Muscle Research, Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
31
|
Fennel ZJ, Amorim FT, Deyhle MR, Hafen PS, Mermier CM. The Heat Shock Connection: Skeletal Muscle Hypertrophy and Atrophy. Am J Physiol Regul Integr Comp Physiol 2022; 323:R133-R148. [PMID: 35536704 DOI: 10.1152/ajpregu.00048.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Skeletal muscle is an integral tissue system that plays a crucial role in the physical function of all vertebrates and is a key target for maintaining or improving health and performance across the lifespan. Based largely on cellular and animal models, there is some evidence that various forms of heat stress with or without resistance exercise may enhance skeletal muscle growth or reduce its loss. It is not clear whether these stimuli are similarly effective in humans or meaningful in comparison to exercise alone across various heating methodologies. Furthermore, the magnitude by which heat stress may influence whole body thermoregulatory responses and the connection to skeletal muscle adaptation remains ambiguous. Finally, the underlying mechanisms, which may include interaction between relevant heat shock proteins and intracellular hypertrophy and atrophy related factors, remain unclear. In this narrative mini-review we examine the relevant literature regarding heat stress alone or in combination with resistance exercise emphasizing skeletal muscle hypertrophy and atrophy across cellular and animal models, as well as human investigations. Additionally, we present working mechanistic theories for heat shock protein mediated signaling effects regarding hypertrophy and atrophy related signaling processes. Importantly, continued research is necessary to determine the practical effects and mechanisms of heat stress with and without resistance exercise on skeletal muscle function via growth and maintenance.
Collapse
Affiliation(s)
| | | | | | - Paul Samuel Hafen
- Department of Health, Exercise, and Sport Sciences, University of New Mexico, Albuquerque, NM, United States.,Indiana University School of Medicine Department of Anatomy, Cell Biology, and Physiology; Indiana Center for Musculoskeletal Health, Indianapolis, IN, United States
| | | |
Collapse
|
32
|
Braun JL, Messner HN, Cleverdon REG, Baranowski RW, Hamstra SI, Geromella MS, Stuart JA, Fajardo VA. Heterozygous SOD2 deletion selectively impairs SERCA function in the soleus of female mice. Physiol Rep 2022; 10:e15285. [PMID: 35581738 PMCID: PMC9114654 DOI: 10.14814/phy2.15285] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/05/2022] [Accepted: 04/08/2022] [Indexed: 06/15/2023] Open
Abstract
The sarco(endo)plasmic reticulum Ca2+ ATPase (SERCA) restores intracellular Ca2+ ([Ca2+ ]i ) to resting levels after muscle contraction, ultimately eliciting relaxation. SERCA pumps are highly susceptible to tyrosine (T)-nitration, impairing their ability to take up Ca2+ resulting in reduced muscle function and increased [Ca2+ ]i and cellular damage. The mitochondrial antioxidant enzyme, superoxide dismutase 2 (SOD2), converts superoxide radicals into less reactive H2 O2 . Heterozygous deletion of SOD2 (Sod2+/- ) in mice increases mitochondrial oxidative stress; however, the consequences of reduced SOD2 expression in skeletal and cardiac muscle, specifically the effect on SERCA pumps, has yet to be investigated. We obtained soleus, extensor digitorum longus (EDL), and left ventricle (LV) muscles from 6 to 7 month-old wild-type (WT) and Sod2+/- female C57BL/6J mice. Ca2+ -dependent SERCA activity assays were performed to assess SERCA function. Western blotting was conducted to examine the protein content of SERCA, phospholamban, and sarcolipin; and immunoprecipitation experiments were done to assess SERCA2a- and SERCA1a-specific T-nitration. Heterozygous SOD2 deletion did not alter SERCA1a or SERCA2a expression in the soleus or LV but reduced SERCA2a in the EDL compared with WT, though this was not statistically significant. Soleus muscles from Sod2+/- mice showed a significant reduction in SERCA's apparent affinity for Ca2+ when compared to WT, corresponding with significantly elevated SERCA2a T-nitration in the soleus. No effect was seen in the EDL or the LV. This is the first study to investigate the effects of SOD2 deficiency on muscle SERCA function and shows that it selectively impairs SERCA function in the soleus.
Collapse
Affiliation(s)
- Jessica L. Braun
- Department of KinesiologyBrock UniversitySt. CatharinesOntarioCanada
- Centre for Bone and Muscle HealthBrock UniversitySt. CatharinesOntarioCanada
- Centre for NeuroscienceBrock UniversitySt. CatharinesOntarioCanada
| | - Holt N. Messner
- Department of KinesiologyBrock UniversitySt. CatharinesOntarioCanada
- Centre for Bone and Muscle HealthBrock UniversitySt. CatharinesOntarioCanada
- Department of Biological SciencesBrock UniversitySt. CatharinesOntarioCanada
| | - Riley E. G. Cleverdon
- Department of KinesiologyBrock UniversitySt. CatharinesOntarioCanada
- Centre for Bone and Muscle HealthBrock UniversitySt. CatharinesOntarioCanada
| | - Ryan W. Baranowski
- Department of KinesiologyBrock UniversitySt. CatharinesOntarioCanada
- Centre for Bone and Muscle HealthBrock UniversitySt. CatharinesOntarioCanada
| | - Sophie I. Hamstra
- Department of KinesiologyBrock UniversitySt. CatharinesOntarioCanada
- Centre for Bone and Muscle HealthBrock UniversitySt. CatharinesOntarioCanada
| | - Mia S. Geromella
- Department of KinesiologyBrock UniversitySt. CatharinesOntarioCanada
- Centre for Bone and Muscle HealthBrock UniversitySt. CatharinesOntarioCanada
| | - Jeffrey A. Stuart
- Department of Biological SciencesBrock UniversitySt. CatharinesOntarioCanada
| | - Val A. Fajardo
- Department of KinesiologyBrock UniversitySt. CatharinesOntarioCanada
- Centre for Bone and Muscle HealthBrock UniversitySt. CatharinesOntarioCanada
- Centre for NeuroscienceBrock UniversitySt. CatharinesOntarioCanada
| |
Collapse
|
33
|
Central and Peripheral NPY Age-Related Regulation: A Comparative Analysis in Fish Translational Models. Int J Mol Sci 2022; 23:ijms23073839. [PMID: 35409198 PMCID: PMC8998975 DOI: 10.3390/ijms23073839] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/28/2022] [Accepted: 03/28/2022] [Indexed: 02/04/2023] Open
Abstract
NPY is among the most abundant neuropeptides in vertebrate brain and is primarily involved in the regulation of food intake. The NPY system is also associated with the aging process showing beneficial effects on neuronal survival via autophagy modulation. Here, we explore the age-related regulation of NPY in the brain and foregut of the shortest- and longest-lived fish species, Nothobranchius furzeri and Danio rerio, respectively. These two research models, despite some similarities, display profound biological differences making them attractive vertebrates to elucidate the mechanisms underlying the regulation of neuropeptide synthesis and function. It is noteworthy that in both fish species only Npya has been identified, while in the other teleosts two classes of NPY (Npya and Npyb) have been annotated. Our findings document that in both species: (i) NPY is centrally regulated; (ii) NPY levels increase in the brain during aging; (iii) NPY is localized in the enteroendocrine cells as well as in the myenteric plexus and drastically decreases in old animals. According to our data, the age-related regulation in the gut resembles that described in other vertebrate species while the increased levels in the brain offer the unique possibility to explore the role of NPY in model organisms to develop future experimental and translatable approaches.
Collapse
|
34
|
Ca 2+ leak through ryanodine receptor 1 regulates thermogenesis in resting skeletal muscle. Proc Natl Acad Sci U S A 2022; 119:2119203119. [PMID: 35046046 PMCID: PMC8794839 DOI: 10.1073/pnas.2119203119] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2021] [Indexed: 11/26/2022] Open
Abstract
The evolution of mammals to use skeletal muscle as a source of heat allowed them to spread to all parts of the globe. The generation of heat requires increased adenosine triphosphate (ATP) hydrolysis in the resting muscle in a regulated manner, but how this mechanism works is unknown. The results suggest that mammals increase their RyR1 Ca2+ leak rate to amplify a basal ATP turnover rate at the sarcoplasmic reticulum Ca2+ pump that is higher than that of lower vertebrates. Muscle-based thermogenesis allows regulation of body temperature that is essential for life in mammals and provides a potential pathway for manipulating body weight or temperature by altering metabolic rate. Mammals rely on nonshivering thermogenesis (NST) from skeletal muscle so that cold temperatures can be tolerated. NST results from activity of the sarcoplasmic reticulum (SR) Ca2+ pump in skeletal muscle, but the mechanisms that regulate this activity are unknown. Here, we develop a single-fiber assay to investigate the role of Ca2+ leak through ryanodine receptor 1 (RyR1) to generate heat at the SR Ca2+ pump in resting muscle. By inhibiting a subpopulation of RyR1s in a single-fiber preparation via targeted delivery of ryanodine through transverse tubules, we achieve in-preparation isolation of RyR1 Ca2+ leak. This maneuver provided a critical increase in signal-to-noise of the SR-temperature-sensitive dye ER thermoyellow fluorescence signal from the fiber to allow detection of SR temperature changes as either RyR1 or SR Ca2+ pump activity was altered. We found that RyR1 Ca2+ leak raises cytosolic [Ca2+] in the local vicinity of the SR Ca2+ pump to amplify thermogenesis. Furthermore, gene-dose-dependent increases in RyR1 leak in RYR1 mutant mice result in progressive rises in leak-dependent heat, consistent with raised local [Ca2+] at the SR Ca2+ pump via RyR1 Ca2+ leak. We also show that basal RyR Ca2+ leak and the heat generated by the SR Ca2+ pump in the absence of RyR Ca2+ leak is greater in fibers from mice than from toads. The distinct function of RyRs and SR Ca2+ pump in endothermic mammals compared to ectothermic amphibians provides insights into the mechanisms by which mammalian skeletal muscle achieves thermogenesis at rest.
Collapse
|
35
|
Chambers PJ, Juracic ES, Fajardo VA, Tupling AR. The role of SERCA and sarcolipin in adaptive muscle remodeling. Am J Physiol Cell Physiol 2022; 322:C382-C394. [PMID: 35044855 DOI: 10.1152/ajpcell.00198.2021] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Sarcolipin (SLN) is a small integral membrane protein that regulates the sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA) pump. When bound to SERCA, SLN reduces the apparent Ca2+ affinity of SERCA and uncouples SERCA Ca2+ transport from its ATP consumption. As such, SLN plays a direct role in altering skeletal muscle relaxation and energy expenditure. Interestingly, the expression of SLN is dynamic during times of muscle adaptation, where large increases in SLN content are found in response to development, atrophy, overload and disease. Several groups have suggested that increases in SLN, especially in dystrophic muscle, are deleterious to muscle function and exacerbate already abhorrent intracellular Ca2+ levels. However, there is also significant evidence to show that increased SLN content is a beneficial adaptive mechanism which protects the SERCA pump and activates Ca2+ signaling and adaptive remodeling during times of cell stress. In this review, we first discuss the role for SLN in healthy muscle during both development and overload, where SLN has been shown to activate Ca2+ signaling to promote mitochondrial biogenesis, fibre type shifts and muscle hypertrophy. Then, with respect to muscle disease, we summarize the discrepancies in the literature as to whether SLN upregulation is adaptive or maladaptive in nature. This review is the first to offer the concept of SLN hormesis in muscle disease, wherein both too much and too little SLN are detrimental to muscle health. Finally, the underlying mechanisms which activate SLN upregulation are discussed, specifically acknowledging a potential positive feedback loop between SLN and Ca2+ signaling molecules.
Collapse
Affiliation(s)
- Paige J Chambers
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, Ontario, Canada
| | - Emma S Juracic
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, Ontario, Canada
| | - Val A Fajardo
- Department Department of Kinesiology, Brock University, St. Catharines, Ontario, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, Ontario, Canada
| | - A Russell Tupling
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, Ontario, Canada
| |
Collapse
|
36
|
Angelini G, Mura G, Messina G. Therapeutic approaches to preserve the musculature in Duchenne Muscular Dystrophy: The importance of the secondary therapies. Exp Cell Res 2022; 410:112968. [PMID: 34883113 DOI: 10.1016/j.yexcr.2021.112968] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 11/15/2021] [Accepted: 12/04/2021] [Indexed: 02/07/2023]
Abstract
Muscular dystrophies (MDs) are heterogeneous diseases, characterized by primary wasting of skeletal muscle, which in severe cases, such as Duchenne Muscular Dystrophy (DMD), leads to wheelchair dependency, respiratory failure, and premature death. Research is ongoing to develop efficacious therapies, particularly for DMD. Most of the efforts, currently focusing on correcting or restoring the primary defect of MDs, are based on gene-addition, exon-skipping, stop codon read-through, and genome-editing. Although promising, most of them revealed several practical limitations. Shared knowledge in the field is that, in order to be really successful, any therapeutic approach has to rely on spared functional muscle tissue, restricting the number of patients eligible for clinical trials to the youngest and less compromised individuals. In line with this, many therapeutic strategies aim to preserve muscle tissue and function. This Review outlines the most interesting and recent studies addressing the secondary outcomes of DMD and how to better deliver the therapeutic agents. In the future, the effective treatment of DMD will likely require combinations of therapies addressing both the primary genetic defect and its consequences.
Collapse
Affiliation(s)
- Giuseppe Angelini
- Department of Biosciences, University of Milan, Via Celoria 26, 20133, Milan, Italy
| | - Giada Mura
- Department of Biosciences, University of Milan, Via Celoria 26, 20133, Milan, Italy
| | - Graziella Messina
- Department of Biosciences, University of Milan, Via Celoria 26, 20133, Milan, Italy.
| |
Collapse
|
37
|
Priksz D, Lampe N, Kovacs A, Herwig M, Bombicz M, Varga B, Wilisicz T, Szilvassy J, Posa A, Kiss R, Gesztelyi R, Raduly A, Szekeres R, Sieme M, Papp Z, Toth A, Hamdani N, Szilvassy Z, Juhasz B. Nicotinic-acid derivative BGP-15 improves diastolic function in a rabbit model of atherosclerotic cardiomyopathy. Br J Pharmacol 2021; 179:2240-2258. [PMID: 34811751 DOI: 10.1111/bph.15749] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 10/25/2021] [Accepted: 11/05/2021] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Small molecule BGP-15 has been reported to alleviate signs of heart failure and improve muscle function in murine models. Here, we investigated the acute and chronic effects of BGP-15 in a rabbit model of atherosclerotic cardiomyopathy. EXPERIMENTAL APPROACH Rabbits were maintained on standard chow (Control) or atherogenic diet (HC) for 16 weeks. BGP-15 was administered intravenously (once) or orally (for 16 weeks), to assess acute and chronic effects. Cardiac function was evaluated by echocardiography, endothelium-dependent vasorelaxation was assessed, and key molecules of the protein kinase G (PKG) axis were examined by ELISA and Western blot. Passive force generation was investigated in skinned cardiomyocytes. KEY RESULTS Both acute and chronic BGP-15 treatment improved the diastolic performance of the diseased heart, however, vasorelaxation and serum lipid markers were unaffected. Myocardial cGMP levels were elevated in the BGP-15-treated group, along with preserved PKG activity and increased phospholamban Ser16-phosphorylation. PDE5 expression decreased in the BGP-15-treated group, and the substance inhibited PDE1 enzyme. Cardiomyocyte passive tension reduced in BGP-15-treated rabbits, the ratio of titin N2BA/N2B isoforms increased, and PKG-dependent N2B-titin phosphorylation elevated in the BGP-15-treated group. CONCLUSIONS AND IMPLICATIONS Here we report that BGP-15-treatment improves diastolic function, reduces cardiomyocyte stiffness, and restores titin compliance in a rabbit model of atherosclerotic cardiomyopathy by increasing the activity of the cGMP-PKG axis. As BGP-15 is proven to be safe, it may have clinical value in the treatment of diastolic dysfunction.
Collapse
Affiliation(s)
- Daniel Priksz
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, Debrecen, Hungary
| | - Nora Lampe
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, Debrecen, Hungary
| | - Arpad Kovacs
- Department of Molecular and Experimental Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany.,Institute of Physiology Ruhr University Bochum, Bochum, Germany
| | - Melissa Herwig
- Department of Molecular and Experimental Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany.,Institute of Physiology Ruhr University Bochum, Bochum, Germany
| | - Mariann Bombicz
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, Debrecen, Hungary
| | - Balazs Varga
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, Debrecen, Hungary
| | - Tician Wilisicz
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, Debrecen, Hungary
| | - Judit Szilvassy
- Department of Otorhinolaryngology and Head-Neck Surgery, University of Debrecen, Debrecen, Hungary
| | - Aniko Posa
- Department of Physiology, Anatomy and Neuroscience, Interdisciplinary Excellence Centre, University of Szeged, Szeged, Hungary
| | - Rita Kiss
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, Debrecen, Hungary
| | - Rudolf Gesztelyi
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, Debrecen, Hungary
| | - Arnold Raduly
- Division of Clinical Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Reka Szekeres
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, Debrecen, Hungary
| | - Marcel Sieme
- Department of Molecular and Experimental Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany.,Institute of Physiology Ruhr University Bochum, Bochum, Germany
| | - Zoltan Papp
- Division of Clinical Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Attila Toth
- Division of Clinical Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Nazha Hamdani
- Department of Molecular and Experimental Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany.,Department of Cardiology, St. Josef-Hospital, Ruhr University Bochum, Bochum, Germany.,Institute of Physiology Ruhr University Bochum, Bochum, Germany.,Department of Clinical Pharmacology, Ruhr University Bochum, Bochum, Germany
| | - Zoltan Szilvassy
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, Debrecen, Hungary
| | - Bela Juhasz
- Department of Pharmacology and Pharmacotherapy, Faculty of General Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
38
|
Xu H, Van Remmen H. The SarcoEndoplasmic Reticulum Calcium ATPase (SERCA) pump: a potential target for intervention in aging and skeletal muscle pathologies. Skelet Muscle 2021; 11:25. [PMID: 34772465 PMCID: PMC8588740 DOI: 10.1186/s13395-021-00280-7] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 10/26/2021] [Indexed: 01/13/2023] Open
Abstract
As a key regulator of cellular calcium homeostasis, the Sarcoendoplasmic Reticulum Calcium ATPase (SERCA) pump acts to transport calcium ions from the cytosol back to the sarcoplasmic reticulum (SR) following muscle contraction. SERCA function is closely associated with muscle health and function, and SERCA activity is susceptible to muscle pathogenesis. For example, it has been well reported that pathological conditions associated with aging, neurodegeneration, and muscular dystrophy (MD) significantly depress SERCA function with the potential to impair intracellular calcium homeostasis and further contribute to muscle atrophy and weakness. As a result, targeting SERCA activity has attracted attention as a therapeutical method for the treatment of muscle pathologies. The interventions include activation of SERCA activity and genetic overexpression of SERCA. This review will focus on SERCA function and regulation mechanisms and describe how those mechanisms are affected under muscle pathological conditions including elevated oxidative stress induced by aging, muscle disease, or neuromuscular disorders. We also discuss the current progress and therapeutic approaches to targeting SERCA in vivo.
Collapse
Affiliation(s)
- Hongyang Xu
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Holly Van Remmen
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA. .,Oklahoma City VA Medical Center, Oklahoma City, OK, USA. .,Department of Physiology, OUHSC, Oklahoma City, OK, USA.
| |
Collapse
|
39
|
Oldfield C, Moffatt TL, Dolinsky VW, Duhamel TA. Sirtuin 3 overexpression preserves maximal sarco(endo)plasmic reticulum calcium ATPase activity in the skeletal muscle of mice subjected to high fat-high sucrose-feeding. Can J Physiol Pharmacol 2021; 100:361-370. [PMID: 34695364 DOI: 10.1139/cjpp-2021-0587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Sarco(endo)plasmic reticulum calcium (Ca2+) ATPase (SERCA) transports Ca2+ in muscle. Impaired SERCA activity contributes to diabetic myopathy. Sirtuin (SIRT) 3 regulates muscle metabolism and function. However, it is unknown if SIRT3 regulates muscle SERCA activity. We determined if SIRT3 overexpression enhances SERCA activity in mouse gastrocnemius muscle and if SIRT3 overexpression preserves gastrocnemius SERCA activity in a model of type 2 diabetes, induced by high fat-high sucrose (HFHS)-feeding. We also determined if the acetylation status of SERCA proteins in mouse gastrocnemius is altered by SIRT3 overexpression or HFHS-feeding. Wild-type (WT) mice and SIRT3 transgenic (SIRT3TG) mice, overexpressing SIRT3 in skeletal muscle, were fed a standard- or HFHS-diet for 4-months. SIRT3TG and WT mice developed obesity and glucose intolerance after 4-months of HFHS-feeding. SERCA Vmax was higher in gastrocnemius of SIRT3TG mice, compared to WT mice. HFHS-fed mice had lower SERCA1a protein levels and lower SERCA Vmax in their gastrocnemius than control-fed mice. The decrease in SERCA Vmax in gastrocnemius muscle due to HFHS-feeding was attenuated by SIRT3 overexpression in HFHS-fed SIRT3TG mice. SERCA1a and SERCA2a acetylation in mouse gastrocnemius was not altered by genotype or diet. These findings suggest SIRT3 overexpression improves SERCA function in diabetic mouse skeletal muscle.
Collapse
Affiliation(s)
- Christopher Oldfield
- University of Manitoba Faculty of Kinesiology and Recreation Management, 175106, Winnipeg, Canada.,St. Boniface Hospital Albrechtsen Research Centre, Institute of Cardiovascular Sciences, Manitoba, Canada;
| | - Teri L Moffatt
- St Boniface General Hospital Research Centre, 120927, Winnipeg, Manitoba, Canada;
| | - Vernon W Dolinsky
- University of Manitoba, Pharmacology and Therapeutics, 601 J. Buhler Research Centre, 715 McDermot Avenue, Winnipeg, Manitoba, Canada, R3E 3P4;
| | - Todd A Duhamel
- St. Boniface General Hospital Research Center, 351 Tach� Avenue, Winnipeg, Manitoba, Canada, R2H 2A6;
| |
Collapse
|
40
|
Li S, Yang P. Relationship between HSPA1A-regulated gene expression and alternative splicing in mouse cardiomyocytes and cardiac hypertrophy. J Thorac Dis 2021; 13:5517-5533. [PMID: 34659818 PMCID: PMC8482330 DOI: 10.21037/jtd-21-1222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 08/30/2021] [Indexed: 11/30/2022]
Abstract
Background Cardiac hypertrophy may be classified as either physiological or pathological. Pathological hypertrophy has a complex etiology and is genetically regulated. In this study, we used a mouse model of cardiac hypertrophy to explore the mechanisms of gene regulation, in particular, modulation of the expression of target genes through transcription factor activity, regulation of immune and inflammation-associated genes and regulation of the alternative splicing of transcription factors. Methods Mouse models of pathological cardiac hypertrophy were established by transverse aortic constriction (TAC). We overexpressed HSPA1A in mouse cardiac HL-1 cells. GO and KEGG pathway annotation database was used to analyze all DEGs. Results The expression of HSPA1A differed significantly between TAC + dantrolene vs. sham + dantrolene (Sham was the non-TAC group, and DMSO was the contrast agent), and TAC + DMSO vs. sham + DMSO. The RNA-binding protein Zfp36 was found to be differentially expressed between both TAC + dantrolene vs. sham + dantrolene and TAC + DMSO vs. sham + DMSO. The expression of mki67 and gm5619 was significantly different between TAC + dantrolene and TAC + DMSO. HSPA1A was found to selectively regulate the expression of non-coding RNAs related to cardiac hypertrophy, including Rn7sk and RMRP. The downregulated genes were mainly related to inflammation and the immune response. HSPA1A negatively regulated alternative splicing of Asxl2 and positively regulated alternative splicing of Runx1. Conclusions HSPA1A was closely related to cardiac hypertrophy. Zfp36 was also related to cardiac hypertrophy. Dantrolene may delay cardiac hypertrophy and ventricular remodeling by regulating the expression of the RNA-binding protein genes mki67 and gm5619. HSPA1A positively regulated the expression of the non-coding RNAs RN7SK and RMRP while negatively regulating the expression of inflammation- and immune response-related genes. HSPA1A can play a role in cardiac hypertrophy by regulating the alternative splicing of asxl2 and runx1.
Collapse
Affiliation(s)
- Shuai Li
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Ping Yang
- Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, Department of Cardiology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
41
|
Ohlendieck K, Swandulla D. Complexity of skeletal muscle degeneration: multi-systems pathophysiology and organ crosstalk in dystrophinopathy. Pflugers Arch 2021; 473:1813-1839. [PMID: 34553265 PMCID: PMC8599371 DOI: 10.1007/s00424-021-02623-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/07/2021] [Accepted: 09/08/2021] [Indexed: 02/07/2023]
Abstract
Duchenne muscular dystrophy is a highly progressive muscle wasting disorder due to primary abnormalities in one of the largest genes in the human genome, the DMD gene, which encodes various tissue-specific isoforms of the protein dystrophin. Although dystrophinopathies are classified as primary neuromuscular disorders, the body-wide abnormalities that are associated with this disorder and the occurrence of organ crosstalk suggest that a multi-systems pathophysiological view should be taken for a better overall understanding of the complex aetiology of X-linked muscular dystrophy. This article reviews the molecular and cellular effects of deficiency in dystrophin isoforms in relation to voluntary striated muscles, the cardio-respiratory system, the kidney, the liver, the gastrointestinal tract, the nervous system and the immune system. Based on the establishment of comprehensive biomarker signatures of X-linked muscular dystrophy using large-scale screening of both patient specimens and genetic animal models, this article also discusses the potential usefulness of novel disease markers for more inclusive approaches to differential diagnosis, prognosis and therapy monitoring that also take into account multi-systems aspects of dystrophinopathy. Current therapeutic approaches to combat muscular dystrophy are summarised.
Collapse
Affiliation(s)
- Kay Ohlendieck
- Department of Biology, Maynooth University, National University of Ireland, Co. Kildare, Maynooth, W23F2H6, Ireland.
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Co. Kildare, Maynooth, W23F2H6, Ireland.
| | - Dieter Swandulla
- Institute of Physiology, University of Bonn, 53115, Bonn, Germany.
| |
Collapse
|
42
|
Zhang J, Ji Y, Jiang S, Shi M, Cai W, Miron RJ, Zhang Y. Calcium-Collagen Coupling is Vital for Biomineralization Schedule. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2100363. [PMID: 34047068 PMCID: PMC8336496 DOI: 10.1002/advs.202100363] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/17/2021] [Indexed: 05/08/2023]
Abstract
Biomineralization is a chemical reaction that occurs in organisms in which collagen initiates and guides the growth and crystallization of matched apatite minerals. However, there is little known about the demand pattern for calcium salts and collagen needed by biomineralization. In this study, natural bone biomineralization is analyzed, and a novel interplay between calcium concentration and collagen production is observed. Any quantitative change in one of the entities causes a corresponding change in the other. Translocation-associated membrane protein 2 (TRAM2) is identified as an intermediate factor whose silencing disrupts this relationship and causes poor mineralization. TRAM2 directly interacts with the sarcoplasmic/endoplasmic reticulum calcium ATPase 2b (SERCA2b) and modulates SERCA2b activity to couple calcium enrichment with collagen biosynthesis. Collectively, these findings indicate that osteoblasts can independently and directly regulate the process of biomineralization via this coupling. This knowledge has significant implications for the developmentally inspired design of biomaterials for bone regenerative applications.
Collapse
Affiliation(s)
- Jinglun Zhang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) and Key Laboratory of Oral BiomedicineMinistry of EducationSchool and Hospital of StomatologyWuhan UniversityWuhan430079China
| | - Yaoting Ji
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) and Key Laboratory of Oral BiomedicineMinistry of EducationSchool and Hospital of StomatologyWuhan UniversityWuhan430079China
| | - Shuting Jiang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) and Key Laboratory of Oral BiomedicineMinistry of EducationSchool and Hospital of StomatologyWuhan UniversityWuhan430079China
| | - Miusi Shi
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) and Key Laboratory of Oral BiomedicineMinistry of EducationSchool and Hospital of StomatologyWuhan UniversityWuhan430079China
| | - Wenjin Cai
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) and Key Laboratory of Oral BiomedicineMinistry of EducationSchool and Hospital of StomatologyWuhan UniversityWuhan430079China
| | - Richard J. Miron
- Centre for Collaborative ResearchNova Southeastern UniversityCell Therapy InstituteFort LauderdaleFL33314‐7796USA
- Department of PeriodontologyCollege of Dental MedicineNova Southeastern UniversityFort LauderdaleFL33314‐7796USA
- Department of Periodontics and Oral SurgeryUniversity of Ann ArborAnn ArborMI48109USA
| | - Yufeng Zhang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei‐MOST) and Key Laboratory of Oral BiomedicineMinistry of EducationSchool and Hospital of StomatologyWuhan UniversityWuhan430079China
| |
Collapse
|
43
|
Swiderski K, Lynch GS. Murine models of Duchenne muscular dystrophy: is there a best model? Am J Physiol Cell Physiol 2021; 321:C409-C412. [PMID: 34260298 DOI: 10.1152/ajpcell.00212.2021] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 07/07/2021] [Indexed: 11/22/2022]
Affiliation(s)
- Kristy Swiderski
- Centre for Muscle Research, Department of Anatomy and Physiology, School of Biomedical Sciences, Faculty of Medicine, Dentistry, and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia
| | - Gordon S Lynch
- Centre for Muscle Research, Department of Anatomy and Physiology, School of Biomedical Sciences, Faculty of Medicine, Dentistry, and Health Sciences, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
44
|
Chemotherapy-Induced Myopathy: The Dark Side of the Cachexia Sphere. Cancers (Basel) 2021; 13:cancers13143615. [PMID: 34298829 PMCID: PMC8304349 DOI: 10.3390/cancers13143615] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 07/11/2021] [Accepted: 07/14/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary In addition to cancer-related factors, anti-cancer chemotherapy treatment can drive life-threatening body wasting in a syndrome known as cachexia. Emerging evidence has described the impact of several key chemotherapeutic agents on skeletal muscle in particular, and the mechanisms are gradually being unravelled. Despite this evidence, there remains very little research regarding therapeutic strategies to protect muscle during anti-cancer treatment and current global grand challenges focused on deciphering the cachexia conundrum fail to consider this aspect—chemotherapy-induced myopathy remains very much on the dark side of the cachexia sphere. This review explores the impact and mechanisms of, and current investigative strategies to protect against, chemotherapy-induced myopathy to illuminate this serious issue. Abstract Cancer cachexia is a debilitating multi-factorial wasting syndrome characterised by severe skeletal muscle wasting and dysfunction (i.e., myopathy). In the oncology setting, cachexia arises from synergistic insults from both cancer–host interactions and chemotherapy-related toxicity. The majority of studies have surrounded the cancer–host interaction side of cancer cachexia, often overlooking the capability of chemotherapy to induce cachectic myopathy. Accumulating evidence in experimental models of cachexia suggests that some chemotherapeutic agents rapidly induce cachectic myopathy, although the underlying mechanisms responsible vary between agents. Importantly, we highlight the capacity of specific chemotherapeutic agents to induce cachectic myopathy, as not all chemotherapies have been evaluated for cachexia-inducing properties—alone or in clinically compatible regimens. Furthermore, we discuss the experimental evidence surrounding therapeutic strategies that have been evaluated in chemotherapy-induced cachexia models, with particular focus on exercise interventions and adjuvant therapeutic candidates targeted at the mitochondria.
Collapse
|
45
|
Al-Zubaidi U, Adhikari D, Cinar O, Zhang QH, Yuen WS, Murphy MP, Rombauts L, Robker RL, Carroll J. Mitochondria-targeted therapeutics, MitoQ and BGP-15, reverse aging-associated meiotic spindle defects in mouse and human oocytes. Hum Reprod 2021; 36:771-784. [PMID: 33367783 DOI: 10.1093/humrep/deaa300] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/28/2020] [Indexed: 12/22/2022] Open
Abstract
STUDY QUESTION Do mitochondria-targeted therapies reverse ageing- and oxidative stress-induced spindle defects in oocytes from mice and humans? SUMMARY ANSWER Exposure to MitoQ or BGP-15 during IVM protected against spindle and chromosomal defects in mouse oocytes exposed to oxidative stress or derived from reproductively aged mice whilst MitoQ promoted nuclear maturation and protected against chromosomal misalignments in human oocytes. WHAT IS KNOWN ALREADY Spindle and chromosomal abnormalities in oocytes are more prevalent with maternal aging, increasing the risk of aneuploidy, miscarriage and genetic disorders such as Down's syndrome. The origin of compromised oocyte function may be founded in mitochondrial dysfunction and increased reactive oxygen species (ROS). STUDY DESIGN, SIZE, DURATION Oocytes from young and old mice were treated with MitoQ and/or BGP-15 during IVM. To directly induce mitochondrial dysfunction, oocytes were treated with H2O2, and then treated the MitoQ and/or BGP-15. Immature human oocytes were cultured with or without MitoQ. Each experiment was repeated at least three times, and data were analyzed by unpaired-sample t-test or chi-square test. PARTICIPANTS/MATERIALS, SETTING, METHODS Immature germinal vesicle (GV) stage oocytes from 1-, 12- and 18-month-old mice were obtained from preovulatory ovarian follicles. Oocytes were treated with MitoQ and/or BGP-15 during IVM. GV-stage human oocytes were cultured with or without MitoQ. Mitochondrial membrane potential and mitochondrial ROS were measured by live-cell imaging. Meiotic spindle and chromosome alignments were visualized by immunofluorescent labeling of fixed oocytes and the 3-dimensional images were analyzed by Imaris. MAIN RESULTS AND THE ROLE OF CHANCE MitoQ or BGP-15 during IVM protects against spindle and chromosomal defects in oocytes exposed to oxidative stress and in oocytes from aged mice (P < 0.001). In human oocytes, the presence of MitoQ during IVM promoted nuclear maturation and had a similar positive effect in protecting against chromosomal misalignments (P < 0.001). LIMITATIONS, REASONS FOR CAUTION Our study identifies two excellent candidates that may help to improve fertility in older women. However, these potential therapies must be tested for efficacy in clinical IVM systems, and undergo thorough examination of resultant offspring in preclinical models before utilization. WIDER IMPLICATIONS OF THE FINDINGS Our results using in-vitro systems for oocyte maturation in both mouse and human provide proof of principle that mitochondrially targeted molecules such as MitoQ and BGP-15 may represent a novel therapeutic approach against maternal aging-related spindle and chromosomal abnormalities. STUDY FUNDING/COMPETING INTEREST(S) The project was financially supported by the National Health and Medical Research Council and Australian Research Council, Australia. U.A.-Z. was supported by the Iraqi Higher Education and Scientific Research Ministry PhD scholarship and O.C. was supported by TUBITAK-1059B191601275. M.P.M. consults for MitoQ Inc. and holds patents in mitochondria-targeted therapies. R.L.R. is an inventor on patents relating to the use of BGP-15 to improve gamete quality. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Usama Al-Zubaidi
- Development and Stem Cell Program and Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia.,Applied Embryology Department, High Institute for Infertility Diagnosis and Assisted Reproductive Technologies, AL-Nahrain University, Baghdad, Iraq
| | - Deepak Adhikari
- Development and Stem Cell Program and Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia
| | - Ozgur Cinar
- Development and Stem Cell Program and Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia.,Department of Histology and Embryology, Ankara University School of Medicine, Ankara, Turkey
| | - Qing-Hua Zhang
- Development and Stem Cell Program and Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia
| | - Wai Shan Yuen
- Development and Stem Cell Program and Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia
| | - Michael P Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge, Hills Road, Cambridge CB2 0XY, UK
| | - Luk Rombauts
- Monash IVF, Melbourne, VIC, Australia.,Department of Obstetrics and Gynaecology, Monash University, Melbourne, VIC, Australia
| | - Rebecca L Robker
- Development and Stem Cell Program and Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia.,School of Medicine, Robinson Research Institute, The University of Adelaide, Adelaide, SA 5005, Australia
| | - John Carroll
- Development and Stem Cell Program and Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800, Australia
| |
Collapse
|
46
|
Cui H, Su W, Cao Y, Ma L, Xu G, Mou W, Zhang H, Yu J, Ma C, Zhang X, Huang Y. Lack of Spinal Neuropeptide Y Is Involved in Mechanical Itch in Aged Mice. Front Aging Neurosci 2021; 13:654761. [PMID: 34122040 PMCID: PMC8192807 DOI: 10.3389/fnagi.2021.654761] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 04/07/2021] [Indexed: 12/12/2022] Open
Abstract
Neuropeptide Y (NPY) signaling plays an essential role in gating the pruritic afferent information in the spinal cord. Recent studies revealed that the aging process down-regulated the expression of NPY in the central nervous system. We propose that the lack of spinal NPY may be involved in certain types of pruritus in the elderly population. This study was designed to investigate the role of NPY in aging-induced itch using the senile mouse model. The expression of NPY in the spinal dorsal horn was compared between young (2 months old) and aged (24 months old) mice. Western blotting and immunohistochemistry showed that the expression of NPY was significantly reduced in the spinal dorsal horn in aged mice. In addition, a neuronal maker of apoptosis, TUNEL, was detected in the NPY positive neurons only in the aged spinal cord. Behavioral assay indicated that light mechanical stimulus evoked significantly more scratching in the aged than in the young mice, whereas chemical-evoked itch and pain-related behaviors were not altered. Intrathecal injection of either NPY or LP-NPY, a NPY receptor 1 (NPY1R) agonist, significantly alleviated the mechanically evoked itch in aged mice without altering the responses to chemical pruritogens. Our study suggested that downregulation of spinal NPY in the aged mice might play a role in the higher incidence of the mechanically evoked itch than that in the young mice. Therapies targeting the NPY system might serve as a potential strategy for alleviating the pruritic symptoms among the elderly population.
Collapse
Affiliation(s)
- Huan Cui
- Department of Human Anatomy, Histology and Embryology, Neuroscience Center, Institute of Basic Medical Sciences, School of Basic Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Wenliang Su
- Department of Anesthesiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yan Cao
- Department of Human Anatomy, Histology and Embryology, Neuroscience Center, Institute of Basic Medical Sciences, School of Basic Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Lulu Ma
- Department of Anesthesiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Guangyan Xu
- Department of Anesthesiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Wanying Mou
- Department of Anesthesiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Hanlin Zhang
- Department of Human Anatomy, Histology and Embryology, Neuroscience Center, Institute of Basic Medical Sciences, School of Basic Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Jiawen Yu
- Department of Anesthesiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Chao Ma
- Department of Human Anatomy, Histology and Embryology, Neuroscience Center, Institute of Basic Medical Sciences, School of Basic Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China.,Chinese Institute for Brain Research, Beijing, China
| | - Xiuhua Zhang
- Department of Anesthesiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yuguang Huang
- Department of Anesthesiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| |
Collapse
|
47
|
Sadler KJ, Gatta PAD, Naim T, Wallace MA, Lee A, Zaw T, Lindsay A, Chung RS, Bello L, Pegoraro E, Lamon S, Lynch GS, Russell AP. Striated muscle activator of Rho signalling (STARS) overexpression in the mdx mouse enhances muscle functional capacity and regulates the actin cytoskeleton and oxidative phosphorylation pathways. Exp Physiol 2021; 106:1597-1611. [PMID: 33963617 DOI: 10.1113/ep089253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 05/04/2021] [Indexed: 11/08/2022]
Abstract
NEW FINDINGS What is the central question of this study? Striated muscle activator of rho signalling (STARS) is an actin-binding protein that regulates transcriptional pathways controlling muscle function, growth and myogenesis, processes that are impaired in dystrophic muscle: what is the regulation of the STARS pathway in Duchenne muscular dystrophy (DMD)? What is the main finding and its importance? Members of the STARS signalling pathway are reduced in the quadriceps of patients with DMD and in mouse models of muscular dystrophy. Overexpression of STARS in the dystrophic deficient mdx mouse model increased maximal isometric specific force and upregulated members of the actin cytoskeleton and oxidative phosphorylation pathways. Regulating STARS may be a therapeutic approach to enhance muscle health. ABSTRACT Duchenne muscular dystrophy (DMD) is characterised by impaired cytoskeleton organisation, cytosolic calcium handling, oxidative stress and mitochondrial dysfunction. This results in progressive muscle damage, wasting and weakness and premature death. The striated muscle activator of rho signalling (STARS) is an actin-binding protein that activates the myocardin-related transcription factor-A (MRTFA)/serum response factor (SRF) transcriptional pathway, a pathway regulating cytoskeletal structure and muscle function, growth and repair. We investigated the regulation of the STARS pathway in the quadriceps muscle from patients with DMD and in the tibialis anterior (TA) muscle from the dystrophin-deficient mdx and dko (utrophin and dystrophin null) mice. Protein levels of STARS, SRF and RHOA were reduced in patients with DMD. STARS, SRF and MRTFA mRNA levels were also decreased in DMD muscle, while Stars mRNA levels were decreased in the mdx mice and Srf and Mrtfa mRNAs decreased in the dko mice. Overexpressing human STARS (hSTARS) in the TA muscles of mdx mice increased maximal isometric specific force by 13% (P < 0.05). This was not associated with changes in muscle mass, fibre cross-sectional area, fibre type, centralised nuclei or collagen deposition. Proteomics screening followed by pathway enrichment analysis identified that hSTARS overexpression resulted in 31 upregulated and 22 downregulated proteins belonging to the actin cytoskeleton and oxidative phosphorylation pathways. These pathways are impaired in dystrophic muscle and regulate processes that are vital for muscle function. Increasing the STARS protein in dystrophic muscle improves muscle force production, potentially via synergistic regulation of cytoskeletal structure and energy production.
Collapse
Affiliation(s)
- Kate J Sadler
- Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Geelong, Victoria, Australia
| | - Paul A Della Gatta
- Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Geelong, Victoria, Australia
| | - Timur Naim
- Department of Physiology, Centre for Muscle Research, University of Melbourne, Parkville, Victoria, Australia
| | - Marita A Wallace
- Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Geelong, Victoria, Australia
| | - Albert Lee
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Centre for Motor Neuron Disease Research, Macquarie University, Sydney, New South Wales, Australia
| | - Thiri Zaw
- Australian Proteome Analysis Facility, Macquarie University, Sydney, New South Wales, Australia
| | - Angus Lindsay
- Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Geelong, Victoria, Australia
| | - Roger S Chung
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Centre for Motor Neuron Disease Research, Macquarie University, Sydney, New South Wales, Australia
| | - Luca Bello
- Department of Neurosciences, ERN Neuromuscular Center, University of Padua, Padua, Italy
| | - Elena Pegoraro
- Department of Neurosciences, ERN Neuromuscular Center, University of Padua, Padua, Italy
| | - Séverine Lamon
- Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Geelong, Victoria, Australia
| | - Gordon S Lynch
- Department of Physiology, Centre for Muscle Research, University of Melbourne, Parkville, Victoria, Australia
| | - Aaron P Russell
- Institute for Physical Activity and Nutrition (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Geelong, Victoria, Australia
| |
Collapse
|
48
|
Gherardi G, De Mario A, Mammucari C. The mitochondrial calcium homeostasis orchestra plays its symphony: Skeletal muscle is the guest of honor. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 362:209-259. [PMID: 34253296 DOI: 10.1016/bs.ircmb.2021.03.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
Abstract
Skeletal muscle mitochondria are placed in close proximity of the sarcoplasmic reticulum (SR), the main intracellular Ca2+ store. During muscle activity, excitation of sarcolemma and of T-tubule triggers the release of Ca2+ from the SR initiating myofiber contraction. The rise in cytosolic Ca2+ determines the opening of the mitochondrial calcium uniporter (MCU), the highly selective channel of the inner mitochondrial membrane (IMM), causing a robust increase in mitochondrial Ca2+ uptake. The Ca2+-dependent activation of TCA cycle enzymes increases the synthesis of ATP required for SERCA activity. Thus, Ca2+ is transported back into the SR and cytosolic [Ca2+] returns to resting levels eventually leading to muscle relaxation. In recent years, thanks to the molecular identification of MCU complex components, the role of mitochondrial Ca2+ uptake in the pathophysiology of skeletal muscle has been uncovered. In this chapter, we will introduce the reader to a general overview of mitochondrial Ca2+ accumulation. We will tackle the key molecular players and the cellular and pathophysiological consequences of mitochondrial Ca2+ dyshomeostasis. In the second part of the chapter, we will discuss novel findings on the physiological role of mitochondrial Ca2+ uptake in skeletal muscle. Finally, we will examine the involvement of mitochondrial Ca2+ signaling in muscle diseases.
Collapse
Affiliation(s)
- Gaia Gherardi
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Agnese De Mario
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | | |
Collapse
|
49
|
Solana-Manrique C, Muñoz-Soriano V, Sanz FJ, Paricio N. Oxidative modification impairs SERCA activity in Drosophila and human cell models of Parkinson's disease. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166152. [PMID: 33892078 DOI: 10.1016/j.bbadis.2021.166152] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 03/10/2021] [Accepted: 04/13/2021] [Indexed: 01/25/2023]
Abstract
DJ-1 is a causative gene for familial Parkinson's disease (PD) with different functions, standing out its role against oxidative stress (OS). Accordingly, PD model flies harboring a mutation in the DJ-1β gene (the Drosophila ortholog of human DJ-1) show high levels of OS markers like protein carbonylation, a common post-translational modification that may alter protein function. To increase our understanding of PD pathogenesis as well as to discover potential therapeutic targets for pharmacological intervention, we performed a redox proteomic assay in DJ-1β mutant flies. Among the proteins that showed increased carbonylation levels in PD model flies, we found SERCA, an endoplasmic reticulum Ca2+ channel that plays an important role in Ca2+ homeostasis. Interestingly, several studies have supported the involvement of Ca2+ dyshomeostasis in PD. Thus, we decided to study the relation between SERCA activity and PD physiopathology. Our results showed that SERCA enzymatic activity is significantly reduced in DJ-1β mutant flies, probably as a consequence of OS-induced carbonylation, as well as in a human cell PD model based on DJ-1-deficiency. Indeed, higher carbonylation levels of SERCA were also observed in DJ-1-deficient cells compared to controls. In addition, the specific activator of SERCA, CDN1163, was also able to restore PD-related phenotypes in both familial PD models by increasing SERCA activity. Taken together, our results indicate that impaired SERCA activity due to oxidative modification may play a role in PD physiopathology. Furthermore, we demonstrate that therapeutic strategies addressing SERCA activation could be beneficial to treat this disease as shown for CDN1163.
Collapse
Affiliation(s)
- Cristina Solana-Manrique
- Departamento de Genética, Facultad CC Biológicas, Universidad de Valencia, 46100 Burjassot, Spain; Instituto Universitario de Biotecnologia y Biomedicina (BIOTECMED), Universidad de Valencia, 46100 Burjassot, Spain
| | - Verónica Muñoz-Soriano
- Departamento de Genética, Facultad CC Biológicas, Universidad de Valencia, 46100 Burjassot, Spain; Instituto Universitario de Biotecnologia y Biomedicina (BIOTECMED), Universidad de Valencia, 46100 Burjassot, Spain
| | - Francisco José Sanz
- Departamento de Genética, Facultad CC Biológicas, Universidad de Valencia, 46100 Burjassot, Spain; Instituto Universitario de Biotecnologia y Biomedicina (BIOTECMED), Universidad de Valencia, 46100 Burjassot, Spain
| | - Nuria Paricio
- Departamento de Genética, Facultad CC Biológicas, Universidad de Valencia, 46100 Burjassot, Spain; Instituto Universitario de Biotecnologia y Biomedicina (BIOTECMED), Universidad de Valencia, 46100 Burjassot, Spain.
| |
Collapse
|
50
|
Wachal Z, Szilágyi A, Takács B, Szabó AM, Priksz D, Bombicz M, Szilvássy J, Juhász B, Szilvássy Z, Varga B. Improved Survival and Retinal Function of Aging ZDF Rats in Long-Term, Uncontrolled Diabetes by BGP-15 Treatment. Front Pharmacol 2021; 12:650207. [PMID: 33935754 PMCID: PMC8085539 DOI: 10.3389/fphar.2021.650207] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 03/02/2021] [Indexed: 11/13/2022] Open
Abstract
Retinal complications of diabetes often lead to deterioration or even loss of vision. This hastens discovery of pharmacological agents able to counterbalance diabetic retinopathy. BGP-15, an emerging small molecule agent, was formerly proven by our workgroup to be retinoprotective on nonobese diabetic animals, Goto-Kakizaki rats. In the present study, we aimed to examine its long-term tolerability or incidental side effects on obese-prone Zucker diabetic fatty (ZDF) rats to further increase the rationale for a future human translation. To make terminal visual status comparable with our other investigations, we also carried out electroretinography (ERG) at the end of the experiment. Our study was started on 16-week-old ZDF rats and lasted for 52 weeks, while BGP was administered daily by gavage. During the 12 months of treatment, 100% of BGP-treated animals survived compared to the non-treated ZDF group, where 60% of the animals died, which was a statistically significant difference. Based on ERG results, BGP-15 was able to counterbalance visual deterioration of ZDF rats caused by long-term diabetes. Some moderate but significant changes were seen in OGTT results and some relationship to oxidative stress by the western blot method: BGP-15 was able to increase expression of HSP70 and decrease that of NFkB in eyes of rats. These were in concert with our previous observations of SIRT1 increment and MMP9 decrement in diabetic eyes by BGP. In summary, not only is BGP-15 not harmful in the long run but it is even able to reduce the related mortality and the serious consequences of diabetes. BGP-15 is an excellent candidate for future drug development against diabetic retinopathy.
Collapse
Affiliation(s)
- Zita Wachal
- Department of Pharmacology and Pharmacotherapy, University of Debrecen, Debrecen, Hungary
| | - Anna Szilágyi
- Department of Pharmacology and Pharmacotherapy, University of Debrecen, Debrecen, Hungary
| | - Barbara Takács
- Department of Pharmacology and Pharmacotherapy, University of Debrecen, Debrecen, Hungary
| | - Adrienn Mónika Szabó
- Department of Pharmacology and Pharmacotherapy, University of Debrecen, Debrecen, Hungary
| | - Dániel Priksz
- Department of Pharmacology and Pharmacotherapy, University of Debrecen, Debrecen, Hungary
| | - Mariann Bombicz
- Department of Pharmacology and Pharmacotherapy, University of Debrecen, Debrecen, Hungary
| | - Judit Szilvássy
- Department of Oto-Rhino-Laryngology and Head and Neck Surgery, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Béla Juhász
- Department of Pharmacology and Pharmacotherapy, University of Debrecen, Debrecen, Hungary
| | - Zoltán Szilvássy
- Department of Pharmacology and Pharmacotherapy, University of Debrecen, Debrecen, Hungary
| | - Balázs Varga
- Department of Pharmacology and Pharmacotherapy, University of Debrecen, Debrecen, Hungary
| |
Collapse
|