1
|
Tammineni ER, Manno C, Oza G, Figueroa L. Skeletal muscle disorders as risk factors for type 2 diabetes. Mol Cell Endocrinol 2025; 599:112466. [PMID: 39848431 PMCID: PMC11886953 DOI: 10.1016/j.mce.2025.112466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/27/2024] [Accepted: 01/20/2025] [Indexed: 01/25/2025]
Abstract
The incidence and prevalence of muscular disorders and of type 2 diabetes (T2D) is increasing and both represent highly significant healthcare problems, both economically and compromising quality of life. Interestingly, skeletal muscle dysfunction and T2D share some commonalities including dysregulated glucose homeostasis, increased oxidative stress, dyslipidemia, and cytokine alterations. Several lines of evidence have hinted to a relationship between skeletal muscle dysfunction and T2D. For instance, T2D affects skeletal muscle morphology, functionality, and overall health through altered protein metabolism, impaired mitochondrial function, and ultimately cell viability. Conversely, humans suffering from myopathies and their experimental models demonstrated increased incidence of T2D through altered muscle glucose disposal function due to abnormal calcium homeostasis, compromised mitochondrial function, dyslipidemia, increased inflammatory cytokines and fiber size alterations and disproportions. Lifestyle modifications are essential for improving and maintaining mobility and metabolic health in individuals suffering from myopathies along with T2D. In this review, we updated current literature evidence on clinical incidence of T2D in inflammatory, mitochondrial, metabolic myopathies, and muscular dystrophies and further discussed the molecular basis of these skeletal muscle disorders leading to T2D.
Collapse
Affiliation(s)
| | - Carlo Manno
- Department of Physiology and Biophysics, Rush University, Chicago, USA
| | - Goldie Oza
- Centro de Investigación y Desarrollo Tecnológico en Electroquímica S. C., Queretaro, Mexico
| | - Lourdes Figueroa
- Department of Physiology and Biophysics, Rush University, Chicago, USA
| |
Collapse
|
2
|
Meizoso-Huesca A, Lamboley CR, Krycer JR, Hodson MP, Hudson JE, Launikonis BS. Muscle-specific Ryanodine receptor 1 properties underlie limb-girdle muscular dystrophy 2B/R2 progression. Nat Commun 2025; 16:3056. [PMID: 40155594 PMCID: PMC11953303 DOI: 10.1038/s41467-025-58393-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 03/17/2025] [Indexed: 04/01/2025] Open
Abstract
Ryanodine receptor 1 Ca2+ leak is a signal in skeletal muscle, but chronic leak can underlie pathology. Here we show that in healthy male mouse, limb-girdle muscle presents higher sympathetic input, elevated ryanodine receptor 1 basal phosphorylation, Ca2+ leak and mitochondrial Ca2+ content compared to distal leg muscles. These regional differences are consistent with heat generation in resting muscle to maintain core temperature. The dysferlin-null mouse develops severe pathology in the limb-girdle but not leg muscles. Absence of dysferlin disrupts dihydropyridine receptors' inhibitory control over ryanodine receptor 1 leak, synergistically increasing leak through the already phosphorylated channel of limb-girdle muscle. This alters Ca2+ handling and distribution leading to reactive oxygen species production prior to disease onset. With age, oxidation of Ca2+ -handling proteins in dysferlin-null limb-girdle muscle alters basal Ca2+ movements. Our results show that muscle-specific pathology in dysferlin-null mice is linked to increased ryanodine receptor 1 Ca2+ leak.
Collapse
MESH Headings
- Animals
- Ryanodine Receptor Calcium Release Channel/metabolism
- Ryanodine Receptor Calcium Release Channel/genetics
- Muscular Dystrophies, Limb-Girdle/metabolism
- Muscular Dystrophies, Limb-Girdle/genetics
- Muscular Dystrophies, Limb-Girdle/pathology
- Male
- Mice
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/pathology
- Calcium/metabolism
- Mice, Knockout
- Dysferlin/metabolism
- Dysferlin/genetics
- Disease Progression
- Reactive Oxygen Species/metabolism
- Phosphorylation
- Mice, Inbred C57BL
- Calcium Channels, L-Type/metabolism
- Calcium Channels, L-Type/genetics
- Disease Models, Animal
Collapse
Affiliation(s)
- Aldo Meizoso-Huesca
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - Cedric R Lamboley
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
| | - James R Krycer
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Mark P Hodson
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
- School of Pharmacy, The University of Queensland, Brisbane, QLD, Australia
| | - James E Hudson
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Bradley S Launikonis
- School of Biomedical Sciences, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
3
|
Le H, Rai V, Agrawal DK. Inflammation and Fatty Infiltration Correlates with Rotator Cuff Muscle Atrophy in Hypercholesterolemic Yucatan Microswine. JOURNAL OF ORTHOPAEDICS AND SPORTS MEDICINE 2024; 6:198-213. [PMID: 39639857 PMCID: PMC11619632 DOI: 10.26502/josm.511500161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Rotator cuff injuries are the most common injuries among active and training astronauts. According to the CDC, 1 in 4 adults in the U.S. experience rotator cuff injuries, particularly affecting the supraspinatus muscle. Hypercholesterolemia, a condition characterized by high levels of LDL cholesterol, is prevalent in approximately 2 in 5 adults in the US and is a risk factor for worsened outcomes in shoulder inflammation and rotator cuff injury repairs. Chronic inflammation, a prolonged low-grade inflammatory state, can arise from conditions like hypercholesterolemia and contribute to muscle atrophy. Skeletal muscle atrophy can be caused by factors such as disuse, aging, malnutrition, and microgravity, and currently lacks approved drug therapies. Thus, gaining a comprehensive understanding of the associations between hypercholesterolemia, chronic inflammation, and skeletal muscle atrophy is imperative for developing effective strategies to manage this condition. We conducted an animal study in Yucatan miniswine to investigate the impact of a high-cholesterol diet on rotator cuff muscle. The results suggested the presence of chronic inflammation in rotator cuff muscle hypercholesterolemic swine, associated with elevated pro-inflammatory cytokines and intramuscular adipocytes, and skeletal muscle atrophy. The results also revealed upregulation of the FOXO3/TRIM63/Titin axis in a hyperlipidemic state. These findings open new perspectives for developing better treatment strategies by targeting the FOXO3/TRIM63/Titin axis to manage rotator cuff muscle atrophy in the context of hypercholesterolemia.
Collapse
Affiliation(s)
- Hoangvi Le
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California USA
| | - Vikrant Rai
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California USA
| | - Devendra K Agrawal
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California USA
| |
Collapse
|
4
|
Sun Z, White Z, Theret M, Bernatchez P. Apolipoprotein E knockout, but not cholesteryl ester transfer protein (CETP)-associated high-density lipoprotein cholesterol (HDL-C) lowering, exacerbates muscle wasting in dysferlin-null mice. Lipids Health Dis 2024; 23:247. [PMID: 39138561 PMCID: PMC11321019 DOI: 10.1186/s12944-024-02227-5] [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: 05/11/2024] [Accepted: 07/30/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Dysferlin-deficient limb-girdle muscular dystrophy type 2B (Dysf) mice are notorious for their mild phenotype. Raising plasma total cholesterol (CHOL) via apolipoprotein E (ApoE) knockout (KO) drastically exacerbates muscle wasting in Dysf mice. However, dysferlinopathic patients have abnormally reduced plasma high-density lipoprotein cholesterol (HDL-C) levels. The current study aimed to determine whether HDL-C lowering can exacerbate the mild phenotype of dysferlin-null mice. METHODS Human cholesteryl ester transfer protein (CETP), a plasma lipid transfer protein not found in mice that reduces HDL-C, and/or its optimal adapter protein human apolipoprotein B (ApoB), were overexpressed in Dysf mice. Mice received a 2% cholesterol diet from 2 months of age and characterized through ambulatory and hanging functional tests, plasma analyses, and muscle histology. RESULTS CETP/ApoB expression in Dysf mice caused reduced HDL-C (54.5%) and elevated ratio of CHOL/HDL-C (181.3%) compared to control Dysf mice in plasma, but without raising CHOL. Compared to the severe muscle pathology found in high CHOL Dysf/ApoE double knockout mice, Dysf/CETP/ApoB mice did not show significant changes in ambulation, hanging capacity, increases in damaged area, collagen deposition, or decreases in cross-sectional area and healthy myofibre coverage. CONCLUSIONS CETP/ApoB over-expression in Dysf mice decreases HDL-C without increasing CHOL or exacerbating muscle pathology. High CHOL or nonHDL-C caused by ApoE KO, rather than low HDL-C, likely lead to rodent muscular dystrophy phenotype humanization.
Collapse
Affiliation(s)
- Zeren Sun
- Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia (UBC), 217-2176 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| | - Zoe White
- Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia (UBC), 217-2176 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| | - Marine Theret
- School of Biomedical Engineering, Department of Medical Genetics, University of British Columbia (UBC), Vancouver, BC, Canada
| | - Pascal Bernatchez
- Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia (UBC), 217-2176 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada.
- UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada.
| |
Collapse
|
5
|
Lloyd EM, Crew RC, Haynes VR, White RB, Mark PJ, Jackaman C, Papadimitriou JM, Pinniger GJ, Murphy RM, Watt MJ, Grounds MD. Pilot investigations into the mechanistic basis for adverse effects of glucocorticoids in dysferlinopathy. Skelet Muscle 2024; 14:19. [PMID: 39123261 PMCID: PMC11312411 DOI: 10.1186/s13395-024-00350-6] [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: 04/21/2024] [Accepted: 07/19/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Dysferlinopathies are a clinically heterogeneous group of muscular dystrophies caused by gene mutations resulting in deficiency of the membrane-associated protein dysferlin. They manifest post-growth and are characterised by muscle wasting (primarily in the limb and limb-gridle muscles), inflammation, and replacement of myofibres with adipose tissue. The precise pathomechanism for dysferlinopathy is currently unclear; as such there are no treatments currently available. Glucocorticoids (GCs) are widely used to reduce inflammation and treat muscular dystrophies, but when administered to patients with dysferlinopathy, they have unexpected adverse effects, with accelerated loss of muscle strength. METHODS To investigate the mechanistic basis for the adverse effects of GCs in dysferlinopathy, the potent GC dexamethasone (Dex) was administered for 4-5 weeks (0.5-0.75 µg/mL in drinking water) to dysferlin-deficient BLA/J and normal wild-type (WT) male mice, sampled at 5 (Study 1) or 10 months (Study 2) of age. A wide range of analyses were conducted. Metabolism- and immune-related gene expression was assessed in psoas muscles at both ages and in quadriceps at 10 months of age. For the 10-month-old mice, quadriceps and psoas muscle histology was assessed. Additionally, we investigated the impact of Dex on the predominantly slow and fast-twitch soleus and extensor digitorum longus (EDL) muscles (respectively) in terms of contractile function, myofibre-type composition, and levels of proteins related to contractile function and metabolism, plus glycogen. RESULTS At both ages, many complement-related genes were highly expressed in BLA/J muscles, and WT mice were generally more responsive to Dex than BLA/J. The effects of Dex on BLA/J mice included (i) increased expression of inflammasome-related genes in muscles (at 5 months) and (ii) exacerbated histopathology of quadriceps and psoas muscles at 10 months. A novel observation was pronounced staining for glycogen in many myofibres of the damaged quadriceps muscles, with large pale vacuolated myofibres, suggesting possible myofibre death by oncosis. CONCLUSION These pilot studies provide a new focus for further investigation into the adverse effects of GCs on dysferlinopathic muscles.
Collapse
Affiliation(s)
- Erin M Lloyd
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, WA, Australia
- Curtin Health Innovation Research Institute, Curtin Medical School, Curtin University, Bentley, WA, Australia
| | - Rachael C Crew
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, WA, Australia
- Department of Obstetrics and Gynaecology, University of Cambridge, Cambridge, UK
| | - Vanessa R Haynes
- Department of Anatomy and Physiology, Faculty of Medicine Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Robert B White
- MD Education Unit, UWA Medical School, The University of Western Australia, Perth, WA, Australia
| | - Peter J Mark
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, WA, Australia
| | - Connie Jackaman
- Curtin Health Innovation Research Institute, Curtin Medical School, Curtin University, Bentley, WA, Australia
| | - John M Papadimitriou
- Department of Pathology and Laboratory Medicine, UWA Medical School, The University of Western Australia, Perth, WA, Australia
| | - Gavin J Pinniger
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, WA, Australia
| | - Robyn M Murphy
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC, Australia
| | - Matthew J Watt
- Department of Anatomy and Physiology, Faculty of Medicine Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Miranda D Grounds
- Department of Anatomy, Physiology and Human Biology, School of Human Sciences, The University of Western Australia, Perth, WA, Australia.
| |
Collapse
|
6
|
Khodabukus A, Prabhu NK, Roberts T, Buldo M, Detwiler A, Fralish ZD, Kondash ME, Truskey GA, Koves TR, Bursac N. Bioengineered Model of Human LGMD2B Skeletal Muscle Reveals Roles of Intracellular Calcium Overload in Contractile and Metabolic Dysfunction in Dysferlinopathy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400188. [PMID: 38887849 PMCID: PMC11336985 DOI: 10.1002/advs.202400188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/19/2024] [Indexed: 06/20/2024]
Abstract
Dysferlin is a multi-functional protein that regulates membrane resealing, calcium homeostasis, and lipid metabolism in skeletal muscle. Genetic loss of dysferlin results in limb girdle muscular dystrophy 2B/2R (LGMD2B/2R) and other dysferlinopathies - rare untreatable muscle diseases that lead to permanent loss of ambulation in humans. The mild disease severity in dysferlin-deficient mice and diverse genotype-phenotype relationships in LGMD2B patients have prompted the development of new in vitro models for personalized studies of dysferlinopathy. Here the first 3-D tissue-engineered hiPSC-derived skeletal muscle ("myobundle") model of LGMD2B is described that exhibits compromised contractile function, calcium-handling, and membrane repair, and transcriptomic changes indicative of impaired oxidative metabolism and mitochondrial dysfunction. In response to the fatty acid (FA) challenge, LGMD2B myobundles display mitochondrial deficits and intracellular lipid droplet (LD) accumulation. Treatment with the ryanodine receptor (RyR) inhibitor dantrolene or the dissociative glucocorticoid vamorolone restores LGMD2B contractility, improves membrane repair, and reduces LD accumulation. Lastly, it is demonstrated that chemically induced chronic RyR leak in healthy myobundles phenocopies LGMD2B contractile and metabolic deficit, but not the loss of membrane repair capacity. Together, these results implicate intramyocellular Ca2+ leak as a critical driver of dysferlinopathic phenotype and validate the myobundle system as a platform to study LGMD2B pathogenesis.
Collapse
Affiliation(s)
| | - Neel K. Prabhu
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Taylor Roberts
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Meghan Buldo
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Amber Detwiler
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | | | - Megan E. Kondash
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | | | - Timothy R. Koves
- Duke Molecular Physiology InstituteDuke UniversityDurhamNC27708USA
| | - Nenad Bursac
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| |
Collapse
|
7
|
Sun Z, Wang X, White Z, Dormuth C, Morales F, Bernatchez P. Dyslipidemia in Muscular Dystrophy: A Systematic Review and Meta-Analysis. J Neuromuscul Dis 2023:JND230064. [PMID: 37182897 DOI: 10.3233/jnd-230064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
BACKGROUND Muscular dystrophies (MDs) are characterized by chronic muscle wasting but also poorly understood metabolic co-morbidities. We have recently shown that Duchenne MD (DMD) patients, dogs and asymptomatic carriers are affected by a new form of dyslipidemia that may exacerbate muscle damage. OBJECTIVE We aimed to perform a systematic review and meta-analysis for evidence that other types of MDs are associated with dyslipidemia compared to healthy controls. METHODS Search was conducted using MEDLINE, EMBASE, Cochrane Central Register of Controlled Trials for reports that compare plasma/serum lipids from MD patients and controls, and meta-analysis of cross-sectional studies quantifying total cholesterol, high-density lipoprotein, low density lipoprotein and triglycerides was performed. RESULTS Out of 749 studies, 17 met our inclusion criteria for meta-analysis. 14 of the 17 studies (82% ) included investigated myotonic dystrophy (DM); other studies were on pseudohypertrophic MD (PMD) or DMD. As a whole, MD individuals had significantly higher levels of circulating total cholesterol (Hedges' g with 95% confidence interval [CI], 0.80 [0.03 - 1.56]; p = 0.04) and triglycerides (Hedges' g with 95% confidence interval [CI], 2.28[0.63 - 3.92]; p = 0.01) compared to controls. Meta-regression analysis showed the percentage of male gender was significantly associated with the difference in total cholesterol (beta = 0.05; 95% CI, - 0.02 to 0.11; p = 0.043) and high-density lipoprotein (beta = - 9.38; 95% CI, - 16.26 to - 2.50; p = 0.028). CONCLUSIONS MD is associated with significantly higher circulating levels of total cholesterol and triglycerides. However, caution on the interpretation of these findings is warranted and future longitudinal research is required to better understand this relationship.
Collapse
Affiliation(s)
- Zeren Sun
- University of British Columbia (UBC) Department of Anesthesiology, Pharmacology & Therapeutics, Vancouver, Canada
- UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, Canada
| | - Xindi Wang
- University of British Columbia (UBC) Department of Anesthesiology, Pharmacology & Therapeutics, Vancouver, Canada
- UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, Canada
| | - Zoe White
- University of British Columbia (UBC) Department of Anesthesiology, Pharmacology & Therapeutics, Vancouver, Canada
- UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, Canada
| | - Colin Dormuth
- University of British Columbia (UBC) Department of Anesthesiology, Pharmacology & Therapeutics, Vancouver, Canada
| | - Fernando Morales
- Instituto de Investigaciones en Salud (INISA), Universidad de Costa Rica, SanJosé, Costa Rica
| | - Pascal Bernatchez
- University of British Columbia (UBC) Department of Anesthesiology, Pharmacology & Therapeutics, Vancouver, Canada
- UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, Canada
| |
Collapse
|
8
|
Le H, Rai V, Agrawal DK. Cholesterol: An Important Determinant of Muscle Atrophy in Astronauts. JOURNAL OF BIOTECHNOLOGY AND BIOMEDICINE 2023; 6:67-79. [PMID: 37006714 PMCID: PMC10062007 DOI: 10.26502/jbb.2642-91280072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
Since cholesterol is not routinely measured in astronauts before and after their return from space, there is no data on the role of blood cholesterol level in muscle atrophy and microgravity. Since the first moon landing, aerospace medicine became outdated and has not pushed boundaries like its rocket engineering counterpart. Since the 2019 astronaut twin study, there has yet to be another scientific breakthrough for aerospace medicine. Microgravity-induced muscle atrophy is the most known consequence of spaceflight. Yet, so far, there is no therapeutic solution to prevent it or any real efforts in understanding it on a cellular or molecular level. The most obvious reason to this unprecedented level of research is due to the small cohort of astronauts. With the establishment of private space industries and exponential recruitment of astronauts, there is more reason to push forward spaceflight-related health guidelines and ensure the safety of the brave humans who risk their lives for the progression of mankind. Spaceflight is considered the most challenging job and the failure to prevent injury or harm should be considered reckless negligence by the institutions that actively prevented sophistication of aerospace medicine. In this critical review, role of cholesterol is analyzed across the NASA-established parameters of microgravity-induced muscle atrophy with a focus on potential therapeutic targets for research.
Collapse
Affiliation(s)
- Hoangvi Le
- Department of Translational Research, Western University of Health Sciences, Pomona, California 91766, USA
| | - Vikrant Rai
- Department of Translational Research, Western University of Health Sciences, Pomona, California 91766, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, Pomona, California 91766, USA
| |
Collapse
|
9
|
Donen G, Milad N, Bernatchez P. Humanization of the mdx Mouse Phenotype for Duchenne Muscular Dystrophy Modeling: A Metabolic Perspective. J Neuromuscul Dis 2023; 10:1003-1012. [PMID: 37574742 PMCID: PMC10657711 DOI: 10.3233/jnd-230126] [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] [Accepted: 07/25/2023] [Indexed: 08/15/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a severe form of muscular dystrophy (MD) that is characterized by early muscle wasting and lethal cardiorespiratory failure. While the mdx mouse is the most common model of DMD, it fails to replicate the severe loss of muscle mass and other complications observed in patients, in part due to the multiple rescue pathways found in mice. This led to several attempts at improving DMD animal models by interfering with these rescue pathways through double transgenic approaches, resulting in more severe phenotypes with mixed relevance to the human pathology. As a growing body of literature depicts DMD as a multi-system metabolic disease, improvements in mdx-based modeling of DMD may be achieved by modulating whole-body metabolism instead of muscle homeostasis. This review provides an overview of the established dual-transgenic approaches that exacerbate the mild mdx phenotype by primarily interfering with muscle homeostasis and highlights how advances in DMD modeling coincide with inducing whole-body metabolic changes. We focus on the DBA2/J strain-based D2.mdx mouse with heightened transforming growth factor (TGF)-β signaling and the dyslipidemic mdx/apolipoprotein E (mdx/ApoE) knock-out (KO) mouse, and summarize how these novel models emulate the metabolic changes observed in DMD.
Collapse
Affiliation(s)
| | | | - Pascal Bernatchez
- Correspondence to: Dr. Pascal Bernatchez, Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia, 2176 Health Sciences mall, room 217, Vancouver BC, V6T 1Z3, Canada. Tel.: +1 604 806 8346 /Ext.66060; E-mail:
| |
Collapse
|
10
|
White Z, Sun Z, Sauge E, Cox D, Donen G, Pechkovsky D, Straub V, Francis GA, Bernatchez P. Limb-girdle muscular dystrophy type 2B causes HDL-C abnormalities in patients and statin-resistant muscle wasting in dysferlin-deficient mice. Skelet Muscle 2022; 12:25. [PMID: 36447272 PMCID: PMC9706908 DOI: 10.1186/s13395-022-00308-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 11/11/2022] [Indexed: 12/03/2022] Open
Abstract
Limb-girdle muscular dystrophy (MD) type 2B (LGMD2B) and Duchenne MD (DMD) are caused by mutations to the Dysferlin and Dystrophin genes, respectively. We have recently demonstrated in typically mild dysferlin- and dystrophin-deficient mouse models that increased plasma cholesterol levels severely exacerbate muscle wasting, and that DMD patients display primary dyslipidemia characterized by elevated plasma cholesterol and triglycerides. Herein, we investigate lipoprotein abnormalities in LGMD2B and if statin therapy protects dysferlin-deficient mice (Dysf) from muscle damage. Herein, lipoproteins and liver enzymes from LGMD2B patients and dysferlin-null (Dysf) mice were analyzed. Simvastatin, which exhibits anti-muscle wasting effects in mouse models of DMD and corrects aberrant expression of key markers of lipid metabolism and endogenous cholesterol synthesis, was tested in Dysf mice. Muscle damage and fibrosis were assessed by immunohistochemistry and cholesterol signalling pathways via Western blot. LGMD2B patients show reduced serum high-density lipoprotein cholesterol (HDL-C) levels compared to healthy controls and exhibit a greater prevalence of abnormal total cholesterol (CHOL)/HDL-C ratios despite an absence of liver dysfunction. While Dysf mice presented with reduced CHOL and associated HDL-C and LDL-C-associated fractions, simvastatin treatment did not prevent muscle wasting in quadriceps and triceps muscle groups or correct aberrant low-density lipoprotein receptor (LDLR) and 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGCR) protein expression. LGMD2B patients present with reduced serum concentrations of HDL-C, a major metabolic comorbidity, and as a result, statin therapy is unlikely to prevent muscle wasting in this population. We propose that like DMD, LGMD2B should be considered as a new type of genetic dyslipidemia.
Collapse
Affiliation(s)
- Zoe White
- grid.17091.3e0000 0001 2288 9830Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia (UBC), 217-2176 Health Sciences Mall, Vancouver, BC V6T 1Z3 Canada ,grid.416553.00000 0000 8589 2327UBC Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, Canada
| | - Zeren Sun
- grid.17091.3e0000 0001 2288 9830Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia (UBC), 217-2176 Health Sciences Mall, Vancouver, BC V6T 1Z3 Canada ,grid.416553.00000 0000 8589 2327UBC Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, Canada
| | - Elodie Sauge
- grid.17091.3e0000 0001 2288 9830Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia (UBC), 217-2176 Health Sciences Mall, Vancouver, BC V6T 1Z3 Canada ,grid.416553.00000 0000 8589 2327UBC Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, Canada
| | - Dan Cox
- grid.1006.70000 0001 0462 7212Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
| | - Graham Donen
- grid.17091.3e0000 0001 2288 9830Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia (UBC), 217-2176 Health Sciences Mall, Vancouver, BC V6T 1Z3 Canada ,grid.416553.00000 0000 8589 2327UBC Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, Canada
| | - Dmitri Pechkovsky
- grid.17091.3e0000 0001 2288 9830Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia (UBC), 217-2176 Health Sciences Mall, Vancouver, BC V6T 1Z3 Canada ,grid.416553.00000 0000 8589 2327UBC Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, Canada
| | - Volker Straub
- grid.1006.70000 0001 0462 7212Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
| | - Gordon A. Francis
- grid.416553.00000 0000 8589 2327UBC Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, Canada ,grid.17091.3e0000 0001 2288 9830Department of Medicine, UBC, Vancouver, Canada
| | - Pascal Bernatchez
- grid.17091.3e0000 0001 2288 9830Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia (UBC), 217-2176 Health Sciences Mall, Vancouver, BC V6T 1Z3 Canada ,grid.416553.00000 0000 8589 2327UBC Centre for Heart Lung Innovation, St. Paul’s Hospital, Vancouver, Canada
| |
Collapse
|
11
|
Pereira CC, Pagotto V, de Oliveira C, Silveira EA. Sarcopenia and mortality risk in community-dwelling Brazilian older adults. Sci Rep 2022; 12:17531. [PMID: 36266412 PMCID: PMC9585028 DOI: 10.1038/s41598-022-22153-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 10/10/2022] [Indexed: 01/13/2023] Open
Abstract
We estimated the impact of sarcopenia parameters on mortality risk and assessed its prevalence and associated factors in the older adults according to the European Working Group on Sarcopenia in Older People's 2010 (EWGSOP1) and 2018 (EWGSOP2) criteria. This was a 10-year follow-up cohort study. Low muscle mass (MM) was defined as low skeletal muscle mass index (SMI) using dual-energy X-ray absorptiometry (DXA), and low calf circumference (CC). Cox regression and the Kaplan-Meier method were performed. The prevalence of sarcopenia and associated factors were influenced by the MM measurement method and diagnostic criteria used [6.8% (SMI and EWGSOP2), 12.8% (CC and EWGSOP2; and SMI and EWGSOP1) and 17.4% (CC and EWGSOP1)]. While a low BMI was associated with sarcopenia regardless of the sarcopenia definitions, diabetes, and high TGs were associated with sarcopenia only when using the EWGSOP1 criteria. Low SMI increased mortality risk (EWGSOP1: HR = 2.01, 95% CI 1.03-3.92; EWGSOP2: HR = 2.07, 95% CI 1.05-4.06). The prevalence of sarcopenia was higher according to EWGSOP1 than EWGSOP2. A low BMI, diabetes, and high TGs were associated with sarcopenia. A low SMI doubled the risk of mortality in community-dwelling older adults.
Collapse
Affiliation(s)
- Cristina Camargo Pereira
- Postgraduate Program in Health Sciences, Medical School, Federal University of Goiás (UFG), Goiania, Brazil
| | - Valéria Pagotto
- Postgraduate Program in Nursing, Faculty of Nursing, Federal University of Goiás (UFG), Goiania, Brazil
| | - Cesar de Oliveira
- Department of Epidemiology and Public Health, University College London, London, UK
| | - Erika Aparecida Silveira
- Postgraduate Program in Health Sciences, Medical School, Federal University of Goiás (UFG), Goiania, Brazil.
- Department of Epidemiology and Public Health, University College London, London, UK.
| |
Collapse
|
12
|
Yazdani AN, Rai V, Agrawal DK. Rotator Cuff Health, Pathology, and Repair in the Perspective of Hyperlipidemia. JOURNAL OF ORTHOPAEDICS AND SPORTS MEDICINE 2022; 4:263-275. [PMID: 36381991 PMCID: PMC9648405 DOI: 10.26502/josm.511500063] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Rotator Cuff Injuries (RCI) are prevalent cause of shoulder pain affecting over 20% of the population in the USA. Surgical repair of the torn rotator cuff helps in relieving the pressure on the rotator cuff tendon and from symptoms, however tendon-to-bone healing after rotator cuff surgery still has a high failure rate. Hyperlipidemia has been strongly associated with RCI although the cellular and molecular mechanisms are largely unknown. The focus of this critical review is to further explore the role of hyperlipidemia in RCI and rotator cuff tissue repair to determine its implication as a risk factor for tears, repair, and retears. A literature review was conducted to elucidate the role of hyperlipidemia as an inflammatory mediator and catalyst for structural instability within the shoulder. The results from various studies were critically reviewed to summarize the relationship between hyperlipidemia and rotator cuff pathology. Hyperlipidemia induces LDL-particle entrapment within the dense regular collagen of rotator cuff tendons resulting in foam cell aggregation and macrophage recruitment. Subsequent inflammatory pathways including the JAK2/STAT3 pathway and NLRP3 inflammasome pathway led to persistent inflammation and Extracellular Matrix (ECM) degradation within the rotator cuff. While arthroscopic repair remains the most common treatment modality, nonsurgical treatment including statins, vitamin D, and targeting miRNA are also of therapeutic benefit. Hyperlipidemia interferes with arthroscopic repairs by inducing inflammation and stiffness within tendons and increases the risk of retears. Most notably, targeting underlying mechanisms influencing inflammation has large therapeutic value as a novel treatment strategy for the management of rotator cuff pathology.
Collapse
Affiliation(s)
- Armand N Yazdani
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, 91766, USA
| | - Vikrant Rai
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, 91766, USA
| | - Devendra K Agrawal
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California, 91766, USA
| |
Collapse
|
13
|
Donen GS, White Z, Sauge E, Ritso M, Theret M, Boyd J, Devlin AM, Rossi FMV, Bernatchez P. Thermoneutral Housing and a Western Diet Combination Exacerbates Dysferlin-Deficient Muscular Dystrophy. Muscle Nerve 2022; 66:513-522. [PMID: 35859452 DOI: 10.1002/mus.27680] [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: 12/20/2021] [Revised: 07/10/2022] [Accepted: 07/17/2022] [Indexed: 11/11/2022]
Abstract
INTRODUCTION/AIMS Most mouse models of muscular dystrophy (MD) show mild phenotypes, which limits the translatability of experimental therapies to patients. A growing body of evidence suggests that MD is accompanied by metabolic abnormalities that could potentially exacerbate the primary muscle wasting process. Since thermoneutral (TN) housing of mice (~30°C) has been shown to affect many metabolic parameters, particularly when combined with a Western diet (WD), our aim was to determine whether the combination of TN and WD exacerbates muscle wasting in dysferlin-deficient BLAJ mice, a common model of limb-girdle MD type 2b (LGMD2b). METHODS Two-month-old wild-type (WT) and BLAJ mice were housed at TN or room temperature (RT) and fed a WD or regular chow for 9 months. Ambulatory function, muscle histology, and protein immunoblots of skeletal muscle were assessed. RESULTS BLAJ mice at RT and fed a chow diet showed normal ambulation function similar to WT mice, whereas 90 % of BLAJ mice under WD and TN combination showed ambulatory dysfunction (P<0.001), and an up to 4.1-fold increase in quadriceps and gastrocnemius fat infiltration. Western blotting revealed decreased autophagy marker microtubules-associated protein 1 light chain 3-B (LC3BII/LC3BI) ratio and up-regulation of AKT and ribosomal protein S6 (rpS6) phosphorylation, suggesting inefficient cellular debris and protein clearance in TN BLAJ mice fed a WD. Male and female BLAJ mice under TN and WD combination showed heterogenous fibro-fatty infiltrate composition. DISCUSSION TN and WD combination exacerbates rodent LGMD2b without affecting WT mice. This improves rodent modeling of human MD and helps elucidate how metabolic abnormalities may play a causal role in muscle wasting.
Collapse
Affiliation(s)
- Graham S Donen
- University of British Columbia (UBC) Department of Anesthesiology, Pharmacology & Therapeutics, Vancouver, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, Canada
| | - Zoe White
- University of British Columbia (UBC) Department of Anesthesiology, Pharmacology & Therapeutics, Vancouver, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, Canada
| | - Elodie Sauge
- University of British Columbia (UBC) Department of Anesthesiology, Pharmacology & Therapeutics, Vancouver, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, Canada
| | - Morten Ritso
- The Biomedical Research Centre, UBC, 2222 Health Sciences Mall, Vancouver, Canada
| | - Marine Theret
- The Biomedical Research Centre, UBC, 2222 Health Sciences Mall, Vancouver, Canada
| | - John Boyd
- UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, Canada
| | - Angela M Devlin
- University of British Columbia (UBC) Department of Pediatrics, BC Children's Hospital Research Institute, Vancouver, Canada
| | - Fabio M V Rossi
- The Biomedical Research Centre, UBC, 2222 Health Sciences Mall, Vancouver, Canada
| | - Pascal Bernatchez
- University of British Columbia (UBC) Department of Anesthesiology, Pharmacology & Therapeutics, Vancouver, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, Canada
| |
Collapse
|
14
|
White Z, Theret M, Milad N, Tung LW, Chen WWH, Sirois MG, Rossi F, Bernatchez P. Cholesterol absorption blocker ezetimibe prevents muscle wasting in severe dysferlin-deficient and mdx mice. J Cachexia Sarcopenia Muscle 2022; 13:544-560. [PMID: 34927367 PMCID: PMC8818667 DOI: 10.1002/jcsm.12879] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 10/26/2021] [Accepted: 10/29/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Muscular dystrophy (MD) causes muscle wasting and is often lethal in patients due to a lack of proven therapies. In contrast, mouse models of MD are notoriously mild. We have previously shown severe human-like muscle pathology in mdx [Duchenne MD (DMD)] and dysferlin-deficient limb-girdle MD type 2B (LGMD2B) mice by inactivating the gene encoding for apolipoprotein E (ApoE), a lipid transporter synthesized by the liver, brain and adipocytes to regulate lipid and fat metabolism. Having recently established that human DMD is a novel type of primary genetic dyslipidaemia with elevated cholesterol, we sought to determine whether cholesterol could exacerbate the muscle wasting process observed in severe rodent MD. METHODS Severe mdx and dysferlin knock-out mice lacking ApoE were treated with ezetimibe (15 mg/kg/day), a clinically approved drug exhibiting few pleiotropic effects. In separate studies, dietary cholesterol was raised (from 0.2% to 2% cholesterol) in combination with experimental micro-injury and direct cholesterol injection assays. Muscles were assessed histologically for changes in collagen and adipocyte infiltration and both transcriptomic and cellular changes by RNA-seq and fluorescence-activated cell sorting analysis. RESULTS Treatment of severe DMD and LGMD2B mice with ezetimibe completely prevented clinical signs of ambulatory dysfunction (0% incidence vs. 33% for vehicle treatment; P < 0.05). Histological analyses revealed that ezetimibe-reduced fibro-fatty infiltration up to 84% and 63% in severely affected triceps (P ≤ 0.0001) and gastrocnemius (P ≤ 0.003) muscles, resulting in a respective 1.9-fold and 2.2-fold retention of healthy myofibre area (P ≤ 0.0001). Additionally, raising dietary cholesterol and thus concentrations of plasma low-density lipoprotein-associated cholesterol (by 250%; P < 0.0001) reduced overall survivability (by 100%; P < 0.001) and worsened muscle damage in the LGMD2B triceps by 767% (P < 0.03). Micro-pin-induced mechanical injury in LGMD2B mice fed a high cholesterol diet exacerbated muscle damage by 425% (P < 0.03) and increased macrophage recruitment (by 98%; P = 0.03) compared with those injured on a chow diet. Parallel RNA-seq analyses revealed that injury in cholesterol-fed mice also modulated the expression of 3671 transcripts (1953 up-regulated), with fibrogenic, inflammatory and programmed cell death-associated pathways among the most enriched. Mice lacking dysferlin also displayed heightened muscle necrosis (by 123%; P < 0.0001) following a direct intramuscular injection of cholesterol compared with control mice. CONCLUSIONS Cholesterol exacerbates rodent MD. Specific inhibition of cholesterol absorption with ezetimibe may safely attenuate human MD severity and delay death.
Collapse
Affiliation(s)
- Zoe White
- Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia (UBC), Vancouver, BC, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| | - Marine Theret
- School of Biomedical Engineering, Department of Medical Genetics, University of British Columbia (UBC), Vancouver, BC, Canada
| | - Nadia Milad
- Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia (UBC), Vancouver, BC, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| | - Lin Wei Tung
- School of Biomedical Engineering, Department of Medical Genetics, University of British Columbia (UBC), Vancouver, BC, Canada
| | - William Wei-Han Chen
- Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia (UBC), Vancouver, BC, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| | - Martin G Sirois
- Montreal Heart Institute, Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, Canada
| | - Fabio Rossi
- School of Biomedical Engineering, Department of Medical Genetics, University of British Columbia (UBC), Vancouver, BC, Canada
| | - Pascal Bernatchez
- Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia (UBC), Vancouver, BC, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| |
Collapse
|
15
|
Bittel DC, Sreetama SC, Chandra G, Ziegler R, Nagaraju K, Van der Meulen JH, Jaiswal JK. Secreted acid sphingomyelinase as a potential gene therapy for limb girdle muscular dystrophy 2B. J Clin Invest 2022; 132:e141295. [PMID: 34981776 PMCID: PMC8718136 DOI: 10.1172/jci141295] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 11/05/2021] [Indexed: 12/14/2022] Open
Abstract
Efficient sarcolemmal repair is required for muscle cell survival, with deficits in this process leading to muscle degeneration. Lack of the sarcolemmal protein dysferlin impairs sarcolemmal repair by reducing secretion of the enzyme acid sphingomyelinase (ASM), and causes limb girdle muscular dystrophy 2B (LGMD2B). The large size of the dysferlin gene poses a challenge for LGMD2B gene therapy efforts aimed at restoring dysferlin expression in skeletal muscle fibers. Here, we present an alternative gene therapy approach targeting reduced ASM secretion, the consequence of dysferlin deficit. We showed that the bulk endocytic ability is compromised in LGMD2B patient cells, which was addressed by extracellularly treating cells with ASM. Expression of secreted human ASM (hASM) using a liver-specific adeno-associated virus (AAV) vector restored membrane repair capacity of patient cells to healthy levels. A single in vivo dose of hASM-AAV in the LGMD2B mouse model restored myofiber repair capacity, enabling efficient recovery of myofibers from focal or lengthening contraction-induced injury. hASM-AAV treatment was safe, attenuated fibro-fatty muscle degeneration, increased myofiber size, and restored muscle strength, similar to dysferlin gene therapy. These findings elucidate the role of ASM in dysferlin-mediated plasma membrane repair and to our knowledge offer the first non-muscle-targeted gene therapy for LGMD2B.
Collapse
Affiliation(s)
- Daniel C. Bittel
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, USA
| | - Sen Chandra Sreetama
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, USA
| | - Goutam Chandra
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, USA
| | - Robin Ziegler
- Rare and Neurologic Diseases Research, Sanofi, Framingham, Massachusetts, USA
| | - Kanneboyina Nagaraju
- School of Pharmacy and Pharmaceutical Sciences, SUNY Binghamton University, Binghamton, New York, USA
| | | | - Jyoti K. Jaiswal
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, USA
- Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| |
Collapse
|
16
|
Tomczyk M, Braczko A, Jablonska P, Mika A, Przyborowski K, Jedrzejewska A, Krol O, Kus F, Sledzinski T, Chlopicki S, Slominska EM, Smolenski RT. Enhanced Muscle Strength in Dyslipidemic Mice and Its Relation to Increased Capacity for Fatty Acid Oxidation. Int J Mol Sci 2021; 22:12251. [PMID: 34830135 PMCID: PMC8620496 DOI: 10.3390/ijms222212251] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/02/2021] [Accepted: 11/08/2021] [Indexed: 12/24/2022] Open
Abstract
Dyslipidemia is commonly linked to skeletal muscle dysfunction, accumulation of intramyocellular lipids, and insulin resistance. However, our previous research indicated that dyslipidemia in apolipoprotein E and low-density lipoprotein receptor double knock-out mice (ApoE/LDLR -/-) leads to improvement of exercise capacity. This study aimed to investigate in detail skeletal muscle function and metabolism in these dyslipidemic mice. We found that ApoE/LDLR -/- mice showed an increased grip strength as well as increased troponins, and Mhc2 levels in skeletal muscle. It was accompanied by the increased skeletal muscle mitochondria numbers (judged by increased citrate synthase activity) and elevated total adenine nucleotides pool. We noted increased triglycerides contents in skeletal muscles and increased serum free fatty acids (FFA) levels in ApoE/LDLR -/- mice. Importantly, Ranolazine mediated inhibition of FFA oxidation in ApoE/LDLR -/- mice led to the reduction of exercise capacity and total adenine nucleotides pool. Thus, this study demonstrated that increased capacity for fatty acid oxidation, an adaptive response to dyslipidemia leads to improved cellular energetics that translates to increased skeletal muscle strength and contributes to increased exercise capacity in ApoE/LDLR -/- mice.
Collapse
Affiliation(s)
- Marta Tomczyk
- Department of Biochemistry, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.B.); (P.J.); (A.J.); (O.K.); (F.K.); (E.M.S.)
| | - Alicja Braczko
- Department of Biochemistry, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.B.); (P.J.); (A.J.); (O.K.); (F.K.); (E.M.S.)
| | - Patrycja Jablonska
- Department of Biochemistry, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.B.); (P.J.); (A.J.); (O.K.); (F.K.); (E.M.S.)
| | - Adriana Mika
- Department of Pharmaceutical Biochemistry, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.M.); (T.S.)
| | - Kamil Przyborowski
- Jagiellonian Centre for Experimental Therapeutics, 30-348 Krakow, Poland; (K.P.); (S.C.)
| | - Agata Jedrzejewska
- Department of Biochemistry, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.B.); (P.J.); (A.J.); (O.K.); (F.K.); (E.M.S.)
| | - Oliwia Krol
- Department of Biochemistry, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.B.); (P.J.); (A.J.); (O.K.); (F.K.); (E.M.S.)
| | - Filip Kus
- Department of Biochemistry, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.B.); (P.J.); (A.J.); (O.K.); (F.K.); (E.M.S.)
- Intercollegiate Faculty of Biotechnology UG-MUG, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Tomasz Sledzinski
- Department of Pharmaceutical Biochemistry, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.M.); (T.S.)
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics, 30-348 Krakow, Poland; (K.P.); (S.C.)
- Chair of Pharmacology, Faculty of Medicine, Jagiellonian University Medical College, 30-688 Krakow, Poland
| | - Ewa M. Slominska
- Department of Biochemistry, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.B.); (P.J.); (A.J.); (O.K.); (F.K.); (E.M.S.)
| | - Ryszard T. Smolenski
- Department of Biochemistry, Medical University of Gdansk, 80-211 Gdansk, Poland; (A.B.); (P.J.); (A.J.); (O.K.); (F.K.); (E.M.S.)
| |
Collapse
|
17
|
White Z, Milad N, Sellers SL, Bernatchez P. Effect of Dysferlin Deficiency on Atherosclerosis and Plasma Lipoprotein Composition Under Normal and Hyperlipidemic Conditions. Front Physiol 2021; 12:675322. [PMID: 34366880 PMCID: PMC8339577 DOI: 10.3389/fphys.2021.675322] [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: 03/02/2021] [Accepted: 06/15/2021] [Indexed: 11/20/2022] Open
Abstract
Dysferlinopathies are a group of muscle disorders caused by mutations to dysferlin, a transmembrane protein involved in membrane patching events following physical damage to skeletal myofibers. We documented dysferlin expression in vascular tissues including non-muscle endothelial cells, suggesting that blood vessels may have an endogenous repair system that helps promote vascular homeostasis. To test this hypothesis, we generated dysferlin-null mice lacking apolipoprotein E (ApoE), a common model of atherosclerosis, dyslipidemia and endothelial injury when stressed with a high fat, and cholesterol-rich diet. Despite high dysferlin expression in mouse and human atheromatous plaques, loss of dysferlin did not affect atherosclerotic burden as measured in the aortic root, arch, thoracic, and abdominal aortic regions. Interestingly, we observed that dysferlin-null mice exhibit lower plasma high-density lipoprotein cholesterol (HDL-C) levels than their WT controls at all measured stages of the disease process. Western blotting revealed abundant dysferlin expression in protein extracts from mouse livers, the main regulator of plasma lipoprotein levels. Despite abnormal lipoprotein levels, Dysf/ApoE double knockout mice responded to cholesterol absorption blockade with lower total cholesterol and blunted atherosclerosis. Our study suggests that dysferlin does not protect against atherosclerosis or participate in cholesterol absorption blockade but regulates basal plasma lipoprotein composition. Dysferlinopathic patients may be dyslipidemic without greater atherosclerotic burden while remaining responsive to cholesterol absorption blockade.
Collapse
Affiliation(s)
- Zoe White
- Department of Anesthesiology, Pharmacology, and Therapeutics, The University of British Columbia, Vancouver, BC, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| | - Nadia Milad
- Department of Anesthesiology, Pharmacology, and Therapeutics, The University of British Columbia, Vancouver, BC, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| | - Stephanie L Sellers
- Department of Anesthesiology, Pharmacology, and Therapeutics, The University of British Columbia, Vancouver, BC, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| | - Pascal Bernatchez
- Department of Anesthesiology, Pharmacology, and Therapeutics, The University of British Columbia, Vancouver, BC, Canada.,UBC Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada
| |
Collapse
|
18
|
Vázquez J, Lefeuvre C, Escobar RE, Luna Angulo AB, Miranda Duarte A, Delia Hernandez A, Brisset M, Carlier RY, Leturcq F, Durand-Canard MC, Nicolas G, Laforet P, Malfatti E. Phenotypic Spectrum of Myopathies with Recessive Anoctamin-5 Mutations. J Neuromuscul Dis 2021; 7:443-451. [PMID: 32925086 DOI: 10.3233/jnd-200515] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Biallelic variants in Anoctamin 5 (ANO5) gene are causative of limb-girdle muscular dystrophy (LGMD) R12 anoctamin5-related, non-dysferlin Miyoshi-like distal myopathy (MMD3), and asymptomatic hyperCKemia. OBJECTIVE To describe clinic, histologic, genetic and imaging features, of ANO5 mutated patients. METHODS Five patients, four from France (P1, P2, P3 and P4) and one from Mexico (P5), from four families were included. P1 and P2, belonging to group 1, had normal muscle strength; Group 2, P3, P4 and P5, presented with muscular weakness. Muscle strength was measured by manual muscle testing, Medical Research Council (MRC) grades 1/5 to 5/5. Laboratory exams included serum CK levels, nerve conduction studies (NCS)/needle electromyography (EMG), pulmonary function tests, EKG and cardiac ultrasound. ANO5 molecular screening was performed with different approaches. RESULTS Group 1 patients showed myalgias with hyperCKemia or isolated hyperCKemia. Group 2 patients presented with limb-girdle or proximo-distal muscular weakness. Serum CK levels ranged from 897 to 5000 UI/L. Muscle biopsy analysis in P4 and P5 showed subsarcolemmal mitochondrial aggregates. Electron microscopy confirmed mitochondrial proliferation and revealed discontinuity of the sarcolemmal membrane. Muscle MRI showed asymmetrical fibro-fatty substitution predominant in the lower limbs.P1 and P2 were compound heterozygous for c.191dupA (p.Asn64Lysfs*15) and c.1898 + G>A; P3 was homozygous for the c.692G>T. (p.Gly231Val); P4 harbored a novel biallelic homozygous exons 1-7 ANO5 gene deletion, and P5 was homozygous for a c.172 C > T (p.(Arg 58 Trp)) ANO5 pathogenic variant. CONCLUSIONS Our cohort confirms the wide clinical variability and enlarge the genetic spectrum of ANO5-related myopathies.
Collapse
Affiliation(s)
- José Vázquez
- Department of Medical Genetics, National Rehabilitation Institute, "Luis Guillermo Ibarra Ibarra", México.,APHP, Department of Neurology, Raymond Poincaré Hospital, North-East-Ile-de-France Neuromuscular Pathology Reference Center, U 1179 INSERM, University Saint Quentin en Yvelines Versailles; Paris-Saclay, France
| | - Claire Lefeuvre
- APHP, Department of Neurology, Raymond Poincaré Hospital, North-East-Ile-de-France Neuromuscular Pathology Reference Center, U 1179 INSERM, University Saint Quentin en Yvelines Versailles; Paris-Saclay, France
| | - Rosa Elena Escobar
- Department of Electromyography and Muscle Dystrophies, National Rehabilitation Institute, "Luis Guillermo Ibarra Ibarra", México
| | | | - Antonio Miranda Duarte
- Department of Medical Genetics, National Rehabilitation Institute, "Luis Guillermo Ibarra Ibarra", México
| | - Alma Delia Hernandez
- Department of Pathology, National Rehabilitation Institute, "Luis Guillermo Ibarra Ibarra", México
| | - Marion Brisset
- APHP, Department of Neurology, Raymond Poincaré Hospital, North-East-Ile-de-France Neuromuscular Pathology Reference Center, U 1179 INSERM, University Saint Quentin en Yvelines Versailles; Paris-Saclay, France
| | - Robert-Yves Carlier
- APHP, GH U. Paris Saclay, DMU Smart Imaging, Department of Radiology, Raymond Poincaré teaching Hospital, 104 Bld R. Poincaré, 92380 Garches, France; U 1179 INSERM, Université Paris-Saclay
| | - France Leturcq
- APHP, Department of Genetics, Cochin Hospital, Paris, France
| | - Marie-Christine Durand-Canard
- APHP, Service of Physiological Explorations Raymond Poincaré Hospital, 104 Bld Raymond Poincaré, 92380 Garches, France
| | - Guillaume Nicolas
- APHP, Department of Neurology, Raymond Poincaré Hospital, North-East-Ile-de-France Neuromuscular Pathology Reference Center, U 1179 INSERM, University Saint Quentin en Yvelines Versailles; Paris-Saclay, France
| | - Pascal Laforet
- APHP, Department of Neurology, Raymond Poincaré Hospital, North-East-Ile-de-France Neuromuscular Pathology Reference Center, U 1179 INSERM, University Saint Quentin en Yvelines Versailles; Paris-Saclay, France
| | - Edoardo Malfatti
- APHP, Department of Neurology, Raymond Poincaré Hospital, North-East-Ile-de-France Neuromuscular Pathology Reference Center, U 1179 INSERM, University Saint Quentin en Yvelines Versailles; Paris-Saclay, France
| |
Collapse
|
19
|
Huang JP, Cheng ML, Wang CH, Huang SS, Hsieh PS, Chang CC, Kuo CY, Chen KH, Hung LM. Therapeutic potential of cPLA2 inhibitor to counteract dilated-cardiomyopathy in cholesterol-treated H9C2 cardiomyocyte and MUNO rat. Pharmacol Res 2020; 160:105201. [PMID: 32942017 DOI: 10.1016/j.phrs.2020.105201] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/03/2020] [Accepted: 09/08/2020] [Indexed: 01/25/2023]
Abstract
BACKGROUND AND PURPOSE The pathogenesis of cardiomyopathy in metabolically unhealthy obesity (MUO) has been well studied. However, the pathogenesis of cardiomyopathy typically associated with high cholesterol levels in metabolically unhealthy nonobesity (MUNO) remains unclear. We investigated whether cholesterol-generated LysoPCs contribute to cardiomyopathy and the role of cytosolic phospholipase A2 (cPLA2) inhibitor in cholesterol-induced MUNO. EXPERIMENTAL APPROACH Cholesterol diet was performed in Sprague-Dawley rats that were fed either regular chow (C), or high cholesterol chow (HC), or HC diet with 10 % fructose in drinking water (HCF) for 12 weeks. LysoPCs levels were subsequently measured in rats and in MUNO human patients. The effects of cholesterol-mediated LysoPCs on cardiac injury, and the action of cPLA2 inhibitor, AACOCF3, were further assessed in H9C2 cardiomyocytes. KEY RESULTS HC and HCF rats fed cholesterol diets demonstrated a MUNO-phenotype and cholesterol-induced dilated cardiomyopathy (DCM). Upregulated levels of LysoPCs were found in rat myocardium and the plasma in MUNO human patients. Further testing in H9C2 cardiomyocytes revealed that cholesterol-induced atrophy and death of cardiomyocytes was due to mitochondrial dysfunction and conditions favoring DCM (i.e. reduced mRNA expression of ANF, BNP, DSP, and atrogin-1), and that AACOCF3 counteracted the cholesterol-induced DCM phenotype. CONCLUSION AND IMPLICATIONS Cholesterol-induced MUNO-DCM phenotype was counteracted by cPLA2 inhibitor, which is potentially useful for the treatment of LysoPCs-associated DCM in MUNO.
Collapse
Affiliation(s)
- Jiung-Pang Huang
- Department and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan.
| | - Mei-Ling Cheng
- Department and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan.
| | - Chao-Hung Wang
- Heart Failure Center, Division of Cardiology, Department of Internal Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan.
| | - Shiang-Suo Huang
- Department of Pharmacology, Chung Shan Medical University, Taichung, Taiwan.
| | - Po-Shiuan Hsieh
- Department of Physiology and Biophysics, National Defense Medical Center, Taipei, Taiwan.
| | - Chih-Chun Chang
- Department of Clinical Pathology, Far Eastern Memorial Hospital, New Taipei, Taiwan.
| | - Chao-Yu Kuo
- Department and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Kuan-Hsing Chen
- Kidney Research Center, Chang Gung Memorial Hospital, Linkou, Taiwan.
| | - Li-Man Hung
- Department and Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Kidney Research Center, Chang Gung Memorial Hospital, Linkou, Taiwan; Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
20
|
White Z, Hakim CH, Theret M, Yang NN, Rossi F, Cox D, Francis GA, Straub V, Selby K, Panagiotopoulos C, Duan D, Bernatchez P. High prevalence of plasma lipid abnormalities in human and canine Duchenne and Becker muscular dystrophies depicts a new type of primary genetic dyslipidemia. J Clin Lipidol 2020; 14:459-469.e0. [PMID: 32593511 DOI: 10.1016/j.jacl.2020.05.098] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 05/13/2020] [Accepted: 05/15/2020] [Indexed: 02/06/2023]
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD) are allelic X-linked recessive muscle diseases caused by mutations in the DMD gene, with DMD being the more severe form. We have recently shown that increased plasma low-density lipoprotein-associated cholesterol causes severe muscle wasting in the mdx mouse, a mild DMD model, which suggested that plasma lipids may play a critical role in DMD. We have also observed that loss of dystrophin in mice causes unexpected elevations in plasma lipoprotein levels. OBJECTIVE The objectives of the study were to determine whether patients with DMD and BMD also present with clinically relevant plasma lipoprotein abnormalities and to mitigate the presence of confounders (medications and lifestyle) by analyzing the plasma from patients with DMD/BMD and unmedicated dogs with DMD, the most relevant model of DMD. METHODS Levels of low-density lipoprotein-associated cholesterol, high-density lipoprotein cholesterol, and triglycerides were analyzed in patients with DMD and BMD and female carriers. Samples from unmedicated, ambulatory dogs with DMD, unaffected carriers, and normal controls were also analyzed. RESULTS We report that 97% and 64% of all pediatric patients with DMD (33 of 36) and BMD (6 of 11) are dyslipidemic, along with an unusually high incidence in adult patients with BMD. All dogs with DMD showed plasma lipid abnormalities that progressively worsened with age. Most strikingly, unaffected carrier dogs also showed plasma lipid abnormalities similar to affected dogs with DMD. Dyslipidemia is likely not secondary to liver damage as unaffected carriers showed no plasma aminotransferase elevation. CONCLUSIONS The high incidence of plasma lipid abnormalities in dystrophin-deficient plasma may depict a new type of genetic dyslipidemia. Abnormal lipid levels in dystrophinopathic samples in the absence of muscle damage suggest a primary state of dyslipidemia. Whether dyslipidemia plays a causal role in patients with DMD warrants further investigation, which could lead to new diagnostic and therapeutic options.
Collapse
Affiliation(s)
- Zoe White
- Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia (UBC), Vancouver, BC, Canada; Centre for Heart & Lung Innovation, St. Paul's Hospital, Vancouver, Canada
| | - Chady H Hakim
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO; National Center for Advancing Translational Sciences, NIH, Rockville, MD
| | | | - N Nora Yang
- National Center for Advancing Translational Sciences, NIH, Rockville, MD
| | - Fabio Rossi
- Biomedical Research Centre, UBC, Vancouver, Canada
| | - Dan Cox
- Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
| | - Gordon A Francis
- Centre for Heart & Lung Innovation, St. Paul's Hospital, Vancouver, Canada; Department of Medicine, UBC, Vancouver, Canada
| | - Volker Straub
- Newcastle University Translational and Clinical Research Institute, Newcastle upon Tyne, UK
| | - Kathryn Selby
- Department of Pediatrics, University of British Columbia (UBC), BC Children's Hospital Research Institute, Vancouver, Canada
| | - Constadina Panagiotopoulos
- Department of Pediatrics, University of British Columbia (UBC), BC Children's Hospital Research Institute, Vancouver, Canada
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO; Department of Pediatrics, University of British Columbia (UBC), BC Children's Hospital Research Institute, Vancouver, Canada; Department of Neurology, University of Missouri, Columbia, MO; Department of Bioengineering, Faculty of Medicine, University of Missouri, Columbia, MO; Department of Biomedical Sciences, College of Veterinary Medicine, University of Missouri, Columbia, MO.
| | - Pascal Bernatchez
- Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia (UBC), Vancouver, BC, Canada; Centre for Heart & Lung Innovation, St. Paul's Hospital, Vancouver, Canada.
| |
Collapse
|
21
|
Kravtsova VV, Bouzinova EV, Chibalin AV, Matchkov VV, Krivoi II. Isoform-specific Na,K-ATPase and membrane cholesterol remodeling in motor endplates in distinct mouse models of myodystrophy. Am J Physiol Cell Physiol 2020; 318:C1030-C1041. [PMID: 32293933 DOI: 10.1152/ajpcell.00453.2019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Na,K-ATPase is a membrane transporter that is critically important for skeletal muscle function. Mdx and Bla/J mice are the experimental models of Duchenne muscular dystrophy and dysferlinopathy that are known to differ in the molecular mechanism of the pathology. This study examines the function of α1- and α2-Na,K-ATPase isozymes in respiratory diaphragm and postural soleus muscles from mdx and Bla/J mice compared with control С57Bl/6 mice. In diaphragm muscles, the motor endplate structure was severely disturbed (manifested by defragmentation) in mdx mice only. The endplate membrane of both Bla/J and mdx mice was depolarized due to specific loss of the α2-Na,K-ATPase electrogenic activity and its decreased membrane abundance. Total FXYD1 subunit (modulates Na,K-ATPase activity) abundance was decreased in both mouse models. However, the α2-Na,K-ATPase protein content as well as mRNA expression were specifically and significantly reduced only in mdx mice. The endplate membrane cholesterol redistribution was most pronounced in mdx mice. Soleus muscles from Bla/J and mdx mice demonstrated reduction of the α2-Na,K-ATPase membrane abundance and mRNA expression similar to the diaphragm muscles. In contrast to diaphragm, the α2-Na,K-ATPase protein content was altered in both Bla/J and mdx mice; membrane cholesterol re-distribution was not observed. Thus, the α2-Na,K-ATPase is altered in both Bla/J and mdx mouse models of chronic muscle pathology. However, despite some similarities, the α2-Na,K-ATPase and cholesterol abnormalities are more pronounced in mdx mice.
Collapse
Affiliation(s)
- Violetta V Kravtsova
- Department of General Physiology, St. Petersburg State University, St. Petersburg, Russia
| | | | | | | | - Igor I Krivoi
- Department of General Physiology, St. Petersburg State University, St. Petersburg, Russia
| |
Collapse
|
22
|
Abstract
Ferlins are multiple-C2-domain proteins involved in Ca2+-triggered membrane dynamics within the secretory, endocytic and lysosomal pathways. In bony vertebrates there are six ferlin genes encoding, in humans, dysferlin, otoferlin, myoferlin, Fer1L5 and 6 and the long noncoding RNA Fer1L4. Mutations in DYSF (dysferlin) can cause a range of muscle diseases with various clinical manifestations collectively known as dysferlinopathies, including limb-girdle muscular dystrophy type 2B (LGMD2B) and Miyoshi myopathy. A mutation in MYOF (myoferlin) was linked to a muscular dystrophy accompanied by cardiomyopathy. Mutations in OTOF (otoferlin) can be the cause of nonsyndromic deafness DFNB9. Dysregulated expression of any human ferlin may be associated with development of cancer. This review provides a detailed description of functions of the vertebrate ferlins with a focus on muscle ferlins and discusses the mechanisms leading to disease development.
Collapse
|
23
|
van Putten M, Lloyd EM, de Greef JC, Raz V, Willmann R, Grounds MD. Mouse models for muscular dystrophies: an overview. Dis Model Mech 2020; 13:dmm043562. [PMID: 32224495 PMCID: PMC7044454 DOI: 10.1242/dmm.043562] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Muscular dystrophies (MDs) encompass a wide variety of inherited disorders that are characterized by loss of muscle tissue associated with a progressive reduction in muscle function. With a cure lacking for MDs, preclinical developments of therapeutic approaches depend on well-characterized animal models that recapitulate the specific pathology in patients. The mouse is the most widely and extensively used model for MDs, and it has played a key role in our understanding of the molecular mechanisms underlying MD pathogenesis. This has enabled the development of therapeutic strategies. Owing to advancements in genetic engineering, a wide variety of mouse models are available for the majority of MDs. Here, we summarize the characteristics of the most commonly used mouse models for a subset of highly studied MDs, collated into a table. Together with references to key publications describing these models, this brief but detailed overview would be useful for those interested in, or working with, mouse models of MD.
Collapse
Affiliation(s)
- Maaike van Putten
- Leiden University Medical Center, Department of Human Genetics, Leiden, 2333 ZA, The Netherlands
| | - Erin M Lloyd
- The University of Western Australia, School of Human Sciences, Perth 6009, Australia
| | - Jessica C de Greef
- Leiden University Medical Center, Department of Human Genetics, Leiden, 2333 ZA, The Netherlands
| | - Vered Raz
- Leiden University Medical Center, Department of Human Genetics, Leiden, 2333 ZA, The Netherlands
| | | | - Miranda D Grounds
- The University of Western Australia, School of Human Sciences, Perth 6009, Australia
| |
Collapse
|
24
|
Rodriguez A, Trigatti BL, Mineo C, Knaack D, Wilkins JT, Sahoo D, Asztalos BF, Mora S, Cuchel M, Pownall HJ, Rosales C, Bernatchez P, Ribeiro Martins da Silva A, Getz GS, Barber JL, Shearer GC, Zivkovic AM, Tietge UJF, Sacks FM, Connelly MA, Oda MN, Davidson WS, Sorci-Thomas MG, Vaisar T, Ruotolo G, Vickers KC, Martel C. Proceedings of the Ninth HDL (High-Density Lipoprotein) Workshop: Focus on Cardiovascular Disease. Arterioscler Thromb Vasc Biol 2019; 39:2457-2467. [PMID: 31597448 DOI: 10.1161/atvbaha.119.313340] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The HDL (high-density lipoprotein) Workshop was established in 2009 as a forum for candid discussions among academic basic scientists, clinical investigators, and industry researchers about the role of HDL in cardiovascular disease. This ninth HDL Workshop was held on May 16 to 17, 2019 in Boston, MA, and included outstanding oral presentations from established and emerging investigators. The Workshop featured 5 sessions with topics that tackled the role of HDL in the vasculature, its structural complexity, its role in health and disease states, and its interaction with the intestinal microbiome. The highlight of the program was awarding the Jack Oram Award to the distinguished professor emeritus G.S. Getz from the University of Chicago. The tenth HDL Workshop will be held on May 2020 in Chicago and will continue the focus on intellectually stimulating presentations by established and emerging investigators on novel roles of HDL in cardiovascular and noncardiovascular health and disease states.
Collapse
Affiliation(s)
- Annabelle Rodriguez
- From the Center for Vascular Biology, Department of Cell Biology, University of Connecticut Health, Farmington (A.R.)
| | - Bernardo L Trigatti
- Department of Biochemistry and Biomedical Sciences, McMaster University, and Thrombosis and Atherosclerosis Research Institute, McMaster University and Hamilton Health Sciences, Hamilton, ON, Canada (B.L.T.)
| | - Chieko Mineo
- Center for Pulmonary and Vascular Biology, Department of Pediatrics and Cell Biology, University of Texas Southwestern Medical Center, Dallas (C.M.)
| | - Darcy Knaack
- Department of Biochemistry (D.K., D.S.), Medical College of Wisconsin, Milwaukee
| | - John T Wilkins
- Division of Cardiology, Departments of Medicine and of Preventive Medicine, Northwestern University, Chicago, IL (J.T.W.)
| | - Daisy Sahoo
- Department of Biochemistry (D.K., D.S.), Medical College of Wisconsin, Milwaukee.,Division of Endocrinology (D.S., M.G.S.-T.), Medical College of Wisconsin, Milwaukee
| | - Bela F Asztalos
- Human Nutrition Research Center, Tufts University, Boston, MA (B.F.A.)
| | - Samia Mora
- Center for Lipid Metabolomics, Divisions of Preventive and Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (S.M.)
| | - Marina Cuchel
- Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (M.C.)
| | - Henry J Pownall
- Institute for Academic Medicine, Houston Methodist, Weill Cornell Medical College, Houston, TX (H.J.P., C.R.)
| | - Corina Rosales
- Institute for Academic Medicine, Houston Methodist, Weill Cornell Medical College, Houston, TX (H.J.P., C.R.)
| | - Pascal Bernatchez
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Heart and Lung Innovation Centre, St Paul's Hospital, Vancouver, BC, Canada (P.B.)
| | | | - Godfrey S Getz
- Department of Pathology, University of Chicago, IL (G.S.G.)
| | - Jacob L Barber
- Department of Exercise Science, University of South Carolina, Columbia (J.L.B.)
| | - Gregory C Shearer
- Department Nutritional Sciences, The Pennsylvania State University, University Park (G.C.S.)
| | | | - Uwe J F Tietge
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden (U.J.F.T.).,Clinical Chemistry, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden (U.J.F.T.)
| | - Frank M Sacks
- Harvard T.H. Chan School of Public Health, Boston, MA (F.M.S.)
| | - Margery A Connelly
- Laboratory Corporation of America Holdings (LabCorp), Morrisville, NC (M.A.C.)
| | | | - W Sean Davidson
- Department of Pathology and Laboratory Medicine, University of Cincinnati, OH (W.S.D.)
| | - Mary G Sorci-Thomas
- Division of Endocrinology (D.S., M.G.S.-T.), Medical College of Wisconsin, Milwaukee
| | - Tomas Vaisar
- UW Medicine Diabetes Institute, University of Washington, Seattle (T.V.)
| | | | - Kasey C Vickers
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN (K.C.V.)
| | - Catherine Martel
- Montreal Heart Institute, Montreal and Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada (C.M.)
| |
Collapse
|
25
|
White Z, Milad N, Tehrani AY, Chen WWH, Donen G, Sellers SL, Bernatchez P. Angiotensin II receptor blocker losartan exacerbates muscle damage and exhibits weak blood pressure-lowering activity in a dysferlin-null model of Limb-Girdle muscular dystrophy type 2B. PLoS One 2019; 14:e0220903. [PMID: 31404091 PMCID: PMC6690544 DOI: 10.1371/journal.pone.0220903] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Accepted: 07/25/2019] [Indexed: 01/01/2023] Open
Abstract
There is no cure or beneficial management option for Limb-Girdle muscular dystrophy (MD) type 2B (LGMD2B). Losartan, a blood pressure (BP) lowering angiotensin II (AngII) receptor type 1 (ATR1) blocker (ARB) with unique anti-transforming growth factor-β (TGF-β) properties, can protect muscles in various types of MD such as Duchenne MD, suggesting a potential benefit for LGMD2B patients. Herein, we show in a mild, dysferlin-null mouse model of LGMD2B that losartan increased quadriceps muscle fibrosis (142%; P<0.0001). In a severe, atherogenic diet-fed model of LGMD2B recently described by our group, losartan further exacerbated dysferlin-null mouse muscle wasting in quadriceps and triceps brachii, two muscles typically affected by LGMD2B, by 40% and 51%, respectively (P<0.05). Lower TGF-β signalling was not observed with losartan, therefore plasma levels of atherogenic lipids known to aggravate LGMD2B severity were investigated. We report that losartan increased both plasma triglycerides and cholesterol concentrations in dysferlin-null mice. Other protective properties of losartan, such as increased nitric oxide release and BP lowering, were also reduced in the absence of dysferlin expression. Our data suggest that LGMD2B patients may show some resistance to the primary BP-lowering effects of losartan along with accelerated muscle wasting and dyslipidemia. Hence, we urge caution on the use of ARBs in this population as their ATR1 pathway may be dysfunctional.
Collapse
Affiliation(s)
- Zoe White
- University of British Columbia (UBC) Department of Anesthesiology, Pharmacology & Therapeutics, Vancouver, Canada
- UBC Centre for Heart Lung Innovation & St. Paul’s Hospital, Vancouver, Canada
- * E-mail: (ZW); (PB)
| | - Nadia Milad
- University of British Columbia (UBC) Department of Anesthesiology, Pharmacology & Therapeutics, Vancouver, Canada
- UBC Centre for Heart Lung Innovation & St. Paul’s Hospital, Vancouver, Canada
| | - Arash Y. Tehrani
- University of British Columbia (UBC) Department of Anesthesiology, Pharmacology & Therapeutics, Vancouver, Canada
- UBC Centre for Heart Lung Innovation & St. Paul’s Hospital, Vancouver, Canada
| | - William Wei-Han Chen
- University of British Columbia (UBC) Department of Anesthesiology, Pharmacology & Therapeutics, Vancouver, Canada
- UBC Centre for Heart Lung Innovation & St. Paul’s Hospital, Vancouver, Canada
| | - Graham Donen
- University of British Columbia (UBC) Department of Anesthesiology, Pharmacology & Therapeutics, Vancouver, Canada
- UBC Centre for Heart Lung Innovation & St. Paul’s Hospital, Vancouver, Canada
| | - Stephanie L. Sellers
- University of British Columbia (UBC) Department of Anesthesiology, Pharmacology & Therapeutics, Vancouver, Canada
- UBC Centre for Heart Lung Innovation & St. Paul’s Hospital, Vancouver, Canada
| | - Pascal Bernatchez
- University of British Columbia (UBC) Department of Anesthesiology, Pharmacology & Therapeutics, Vancouver, Canada
- UBC Centre for Heart Lung Innovation & St. Paul’s Hospital, Vancouver, Canada
- * E-mail: (ZW); (PB)
| |
Collapse
|
26
|
Wang J, Khodabukus A, Rao L, Vandusen K, Abutaleb N, Bursac N. Engineered skeletal muscles for disease modeling and drug discovery. Biomaterials 2019; 221:119416. [PMID: 31419653 DOI: 10.1016/j.biomaterials.2019.119416] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 08/01/2019] [Accepted: 08/05/2019] [Indexed: 01/04/2023]
Abstract
Skeletal muscle is the largest organ of human body with several important roles in everyday movement and metabolic homeostasis. The limited ability of small animal models of muscle disease to accurately predict drug efficacy and toxicity in humans has prompted the development in vitro models of human skeletal muscle that fatefully recapitulate cell and tissue level functions and drug responses. We first review methods for development of three-dimensional engineered muscle tissues and organ-on-a-chip microphysiological systems and discuss their potential utility in drug discovery research and development of new regenerative therapies. Furthermore, we describe strategies to increase the functional maturation of engineered muscle, and motivate the importance of incorporating multiple tissue types on the same chip to model organ cross-talk and generate more predictive drug development platforms. Finally, we review the ability of available in vitro systems to model diseases such as type II diabetes, Duchenne muscular dystrophy, Pompe disease, and dysferlinopathy.
Collapse
Affiliation(s)
- Jason Wang
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | - Lingjun Rao
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Keith Vandusen
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Nadia Abutaleb
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
27
|
Ishiba R, Santos ALF, Almeida CF, Caires LC, Ribeiro AF, Ayub-Guerrieri D, Fernandes SA, Souza LS, Vainzof M. Faster regeneration associated to high expression of Fam65b and Hdac6 in dysferlin-deficient mouse. J Mol Histol 2019; 50:375-387. [DOI: 10.1007/s10735-019-09834-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 06/10/2019] [Indexed: 11/27/2022]
|
28
|
Haynes VR, Keenan SN, Bayliss J, Lloyd EM, Meikle PJ, Grounds MD, Watt MJ. Dysferlin deficiency alters lipid metabolism and remodels the skeletal muscle lipidome in mice. J Lipid Res 2019; 60:1350-1364. [PMID: 31203232 DOI: 10.1194/jlr.m090845] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 05/14/2019] [Indexed: 12/15/2022] Open
Abstract
Defects in the gene coding for dysferlin, a membrane-associated protein, affect many tissues, including skeletal muscles, with a resultant myopathy called dysferlinopathy. Dysferlinopathy manifests postgrowth with a progressive loss of skeletal muscle function, early intramyocellular lipid accumulation, and a striking later replacement of selective muscles by adipocytes. To better understand the changes underpinning this disease, we assessed whole-body energy homeostasis, skeletal muscle fatty acid metabolism, lipolysis in adipose tissue, and the skeletal muscle lipidome using young adult dysferlin-deficient male BLAJ mice and age-matched C57Bl/6J WT mice. BLAJ mice had increased lean mass and reduced fat mass associated with increased physical activity and increased adipose tissue lipolysis. Skeletal muscle fatty acid metabolism was remodeled in BLAJ mice, characterized by a partitioning of fatty acids toward storage rather than oxidation. Lipidomic analysis identified marked changes in almost all lipid classes examined in the skeletal muscle of BLAJ mice, including sphingolipids, phospholipids, cholesterol, and most glycerolipids but, surprisingly, not triacylglycerol. These observations indicate that an early manifestation of dysferlin deficiency is the reprogramming of skeletal muscle and adipose tissue lipid metabolism, which is likely to contribute to the progressive adverse histopathology in dysferlinopathies.
Collapse
Affiliation(s)
- Vanessa R Haynes
- Department of Physiology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Australia
| | - Stacey N Keenan
- Department of Physiology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Australia
| | - Jackie Bayliss
- Department of Physiology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Australia
| | - Erin M Lloyd
- School of Human Sciences University of Western Australia, Perth, Australia
| | - Peter J Meikle
- Metabolomics Laboratory Baker Heart Institute, Melbourne, Australia
| | - Miranda D Grounds
- School of Human Sciences University of Western Australia, Perth, Australia
| | - Matthew J Watt
- Department of Physiology, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Australia
| |
Collapse
|
29
|
Dysferlin-deficiency has greater impact on function of slow muscles, compared with fast, in aged BLAJ mice. PLoS One 2019; 14:e0214908. [PMID: 30970035 PMCID: PMC6457631 DOI: 10.1371/journal.pone.0214908] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 03/24/2019] [Indexed: 12/26/2022] Open
Abstract
Dysferlinopathies are a form of muscular dystrophy caused by gene mutations resulting in deficiency of the protein dysferlin. Symptoms manifest later in life in a muscle specific manner, although the pathomechanism is not well understood. This study compared the impact of dysferlin-deficiency on in vivo and ex vivo muscle function, and myofibre type composition in slow (soleus) and fast type (extensor digitorum longus; EDL) muscles using male dysferlin-deficient (dysf-/-) BLAJ mice aged 10 months, compared with wild type (WT) C57Bl/6J mice. There was a striking increase in muscle mass of BLAJ soleus (+25%) (p<0.001), with no strain differences in EDL mass, compared with WT. In vivo measures of forelimb grip strength and wheel running capacity showed no strain differences. Ex vivo measures showed the BLAJ soleus had faster twitch contraction (-21%) and relaxation (-20%) times, and delayed post fatigue recovery (ps<0.05); whereas the BLAJ EDL had a slower relaxation time (+11%) and higher maximum rate of force production (+25%) (ps<0.05). Similar proportions of MHC isoforms were evident in the soleus muscles of both strains (ps>0.05); however, for the BLAJ EDL, there was an increased proportion of type IIx MHC isoform (+5.5%) and decreased type IIb isoform (-5.5%) (ps<0.01). This identification of novel differences in the impact of dysferlin-deficiency on slow and fast twitch muscles emphasises the importance of evaluating myofibre type specific effects to provide crucial insight into the mechanisms responsible for loss of function in dysferlinopathies; this is critical for the development of targeted future clinical therapies.
Collapse
|
30
|
Cholesterol and the Safety Factor for Neuromuscular Transmission. Int J Mol Sci 2019; 20:ijms20051046. [PMID: 30823359 PMCID: PMC6429197 DOI: 10.3390/ijms20051046] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 02/23/2019] [Accepted: 02/24/2019] [Indexed: 12/12/2022] Open
Abstract
A present review is devoted to the analysis of literature data and results of own research. Skeletal muscle neuromuscular junction is specialized to trigger the striated muscle fiber contraction in response to motor neuron activity. The safety factor at the neuromuscular junction strongly depends on a variety of pre- and postsynaptic factors. The review focuses on the crucial role of membrane cholesterol to maintain a high efficiency of neuromuscular transmission. Cholesterol metabolism in the neuromuscular junction, its role in the synaptic vesicle cycle and neurotransmitter release, endplate electrogenesis, as well as contribution of cholesterol to the synaptogenesis, synaptic integrity, and motor disorders are discussed.
Collapse
|