Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 321
Filter
Add more filters

Country/Region as subject
Publication year range
1.
PLoS Genet ; 16(11): e1009179, 2020 11.
Article in English | MEDLINE | ID: mdl-33175853

ABSTRACT

Gene therapy approaches for DMD using recombinant adeno-associated viral (rAAV) vectors to deliver miniaturized (or micro) dystrophin genes to striated muscles have shown significant progress. However, concerns remain about the potential for immune responses against dystrophin in some patients. Utrophin, a developmental paralogue of dystrophin, may provide a viable treatment option. Here we examine the functional capacity of an rAAV-mediated microutrophin (µUtrn) therapy in the mdx4cv mouse model of DMD. We found that rAAV-µUtrn led to improvement in dystrophic histopathology & mostly restored the architecture of the neuromuscular and myotendinous junctions. Physiological studies of tibialis anterior muscles indicated peak force maintenance, with partial improvement of specific force. A fundamental question for µUtrn therapeutics is not only can it replace critical functions of dystrophin, but whether full-length utrophin impacts the therapeutic efficacy of the smaller, highly expressed µUtrn. As such, we found that µUtrn significantly reduced the spacing of the costameric lattice relative to full-length utrophin. Further, immunostaining suggested the improvement in dystrophic pathophysiology was largely influenced by favored correction of fast 2b fibers. However, unlike µUtrn, µdystrophin (µDys) expression did not show this fiber type preference. Interestingly, µUtrn was better able to protect 2a and 2d fibers in mdx:utrn-/- mice than in mdx4cv mice where the endogenous full-length utrophin was most prevalent. Altogether, these data are consistent with the role of steric hindrance between full-length utrophin & µUtrn within the sarcolemma. Understanding the stoichiometry of this effect may be important for predicting clinical efficacy.


Subject(s)
Genetic Therapy/methods , Muscle Fibers, Skeletal/pathology , Muscular Dystrophy, Duchenne/therapy , Utrophin/therapeutic use , Animals , Dependovirus/genetics , Disease Models, Animal , Dystrophin/genetics , Gene Transfer Techniques , Genetic Vectors/genetics , HEK293 Cells , Humans , Mice , Mice, Inbred mdx , Microscopy, Electron , Muscle Contraction , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/ultrastructure , Muscle, Skeletal , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Neuromuscular Junction/pathology , Neuromuscular Junction/ultrastructure , Sarcolemma/pathology , Sarcolemma/ultrastructure , Utrophin/genetics
2.
Hum Mol Genet ; 29(10): 1607-1623, 2020 06 27.
Article in English | MEDLINE | ID: mdl-32227114

ABSTRACT

Duchenne muscular dystrophy (DMD) is a lethal, X-linked disease characterized by progressive muscle degeneration. The condition is driven by nonsense and missense mutations in the dystrophin gene, leading to instability of the sarcolemma and skeletal muscle necrosis and atrophy. Resulting changes in muscle-specific gene expression that take place in dystrophin's absence remain largely uncharacterized, as they are potentially obscured by the chronic inflammation elicited by muscle damage in humans. Caenorhabditis elegans possess a mild inflammatory response that is not active in the muscle, and lack a satellite cell equivalent. This allows for the characterization of the transcriptome rearrangements affecting disease progression independently of inflammation and regeneration. In effort to better understand these dynamics, we have isolated and sequenced body muscle-specific transcriptomes from C. elegans lacking functional dystrophin at distinct stages of disease progression. We have identified an upregulation of genes involved in mitochondrial function early in disease progression, and an upregulation of genes related to muscle repair in later stages. Our results suggest that in C. elegans, dystrophin may have a signaling role early in development, and its absence may activate compensatory mechanisms that counteract muscle degradation caused by loss of dystrophin. We have also developed a temperature-based screening method for synthetic paralysis that can be used to rapidly identify genetic partners of dystrophin. Our results allow for the comprehensive identification of transcriptome changes that potentially serve as independent drivers of disease progression and may in turn allow for the identification of new therapeutic targets for the treatment of DMD.


Subject(s)
Dystrophin/genetics , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Duchenne/genetics , Transcriptome/genetics , Animals , Caenorhabditis elegans/genetics , Codon, Nonsense/genetics , Disease Models, Animal , Humans , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Duchenne/pathology , Sarcolemma/genetics , Sarcolemma/pathology
3.
Biochem Biophys Res Commun ; 540: 116-122, 2021 02 12.
Article in English | MEDLINE | ID: mdl-33472133

ABSTRACT

Mitochondrial dysfunction is considered to be a major cause of sarcopenia, defined as age-related muscle fiber atrophy and muscle weakness, as reduced mitochondrial respiration and morphological changes such as ragged red fibers (RRFs) are observed in aging muscles. However, the role of mitochondrial dysfunction in sarcopenia is not fully elucidated. Although previous studies have suggested that aging has a fiber type-specific effect on mitochondrial function, little is known about mitochondrial changes in individual fiber types. Here, we used C57BL/6NCr female mice to identify fiber type-specific pathological changes, examine the significance of pathological changes in sarcopenia, and identify possible mechanisms behind mitochondrial changes in slow-twitch soleus muscle (SOL) and fast-twitch extensor digitorum longus muscle (EDL). We observed reduced type I fiber-specific mitochondrial respiratory enzyme activity, impaired respiration, and subsarcolemmal mitochondrial accumulation in aged SOL, which was different from RRFs. These pathological alterations were not directly associated with fiber atrophy. Additionally, we found increased oxidative stress markers in aged SOL, suggesting that oxidative stress is involved in the pathological and functional changes in mitochondria. Meanwhile, obvious mitochondrial changes were not seen in aged EDL. Thus, age-related mitochondrial dysfunction is specific to the fiber type and may correlate with the muscle quality rather than the muscle mass.


Subject(s)
Aging/metabolism , Aging/pathology , Cell Respiration , Mitochondria/metabolism , Mitochondria/pathology , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Animals , Female , Mice , Mitochondria/enzymology , Muscle Fibers, Skeletal/enzymology , Organ Size , Oxidative Phosphorylation , Oxidative Stress , Sarcolemma/enzymology , Sarcolemma/metabolism , Sarcolemma/pathology , Sarcopenia/enzymology , Sarcopenia/metabolism , Sarcopenia/pathology
4.
Neuropathol Appl Neurobiol ; 46(6): 579-587, 2020 10.
Article in English | MEDLINE | ID: mdl-32144790

ABSTRACT

AIMS: Nakajo-Nishimura syndrome (NNS) is an autosomal recessive disease caused by biallelic mutations in the PSMB8 gene that encodes the immunoproteasome subunit ß5i. There have been only a limited number of reports on the clinicopathological features of the disease in genetically confirmed cases. METHODS: We studied clinical and pathological features of three NNS patients who all carry the homozygous p.G201V mutations in PSMB8. Patients' muscle specimens were analysed with histology and immunohistochemistry. RESULTS: All patients had episodes of typical periodic fever and skin rash, and later developed progressive muscle weakness and atrophy, similar to previous reports. Oral corticosteroid was used for treatment but showed no obvious efficacy. On muscle pathology, lymphocytes were present in the endomysium surrounding non-necrotic fibres, as well as in the perimysium perivascular area. Nearly all fibres strongly expressed MHC-I in the sarcolemma. In the eldest patient, there were abnormal protein aggregates in the sarcoplasm, immunoreactive to p62, TDP-43 and ubiquitin antibodies. CONCLUSIONS: These results suggest that inflammation, inclusion pathology and aggregation of abnormal proteins underlie the progressive clinical course of the NNS pathomechanism.


Subject(s)
Erythema Nodosum/genetics , Erythema Nodosum/pathology , Fingers/abnormalities , Inclusion Bodies/genetics , Inclusion Bodies/pathology , Myositis/genetics , Myositis/pathology , Sarcoplasmic Reticulum/pathology , Adult , Age of Onset , Child, Preschool , Exanthema/genetics , Exanthema/pathology , Female , Fever/genetics , Fever/pathology , Fingers/pathology , Genes, MHC Class I/genetics , Humans , Infant , Lymphocytes/pathology , Male , Muscle Weakness/genetics , Muscle Weakness/pathology , Mutation/genetics , Nerve Fibers/pathology , Proteasome Endopeptidase Complex/genetics , Sarcolemma/pathology , Young Adult
5.
J Mol Cell Cardiol ; 128: 51-61, 2019 03.
Article in English | MEDLINE | ID: mdl-30664850

ABSTRACT

BACKGROUND: Duchenne muscular dystrophy (DMD) is an X-linked disease characterized by skeletal muscle degeneration and a significant cardiomyopathy secondary to cardiomyocyte damage and myocardial loss. The molecular basis of DMD lies in the absence of the protein dystrophin, which plays critical roles in mechanical membrane integrity and protein localization at the sarcolemma. A popular mouse model of DMD is the mdx mouse, which lacks dystrophin and displays mild cardiac and skeletal pathology that can be exacerbated to advance the disease state. In clinical and pre-clinical studies of DMD, angiotensin signaling pathways have emerged as therapeutic targets due to their adverse influence on muscle remodeling and oxidative stress. Here we aim to establish a physiologically relevant cardiac injury model in the mdx mouse, and determine whether acute blockade of the angiotensin II type 1 receptor (AT1R) may be utilized for prevention of dystrophic injury. METHODS AND RESULTS: A single IP injection of isoproterenol (Iso, 10 mg/kg) was used to induce cardiac stress and injury in mdx and wild type (C57Bl/10) mice. Mice were euthanized 8 h, 30 h, 1 week, or 1 month following the injection, and hearts were harvested for injury evaluation. At 8 and 30 h post-injury, mdx hearts showed 2.2-fold greater serum cTnI content and 3-fold more extensive injury than wild type hearts. Analysis of hearts 1 week and 1 month after injury revealed significantly higher fibrosis in mdx hearts, with a more robust and longer-lasting immune response compared to wild type hearts. In the 30-hour group, losartan treatment initiated 1 h before Iso injection protected dystrophic hearts from cardiac damage, reducing mdx acute injury area by 2.8-fold, without any significant effect on injury in wild type hearts. However, both wild type and dystrophic hearts showed a 2-fold reduction in the magnitude of the macrophage response to injury 30 h after Iso with losartan. CONCLUSIONS: This work demonstrates that acute blockade of AT1R has the potential for robust injury prevention in a model of Iso-induced dystrophic heart injury. In addition to selectively limiting dystrophic cardiac damage, blocking AT1R may serve to limit the inflammatory nature of the immune response to injury in all hearts. Our findings strongly suggest that earlier adoption of angiotensin receptor blockers in DMD patients could limit myocardial damage and subsequent cardiomyopathy.


Subject(s)
Cardiomyopathies/drug therapy , Heart/drug effects , Muscular Dystrophy, Duchenne/drug therapy , Receptor, Angiotensin, Type 1/genetics , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Dystrophin/genetics , Heart/physiopathology , Humans , Isoproterenol/pharmacology , Losartan/pharmacology , Mice , Mice, Inbred mdx , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Sarcolemma/metabolism , Sarcolemma/pathology
6.
J Biol Chem ; 293(23): 8734-8749, 2018 06 08.
Article in English | MEDLINE | ID: mdl-29610273

ABSTRACT

G protein-coupled receptors that signal through Gαq (Gq receptors), such as α1-adrenergic receptors (α1-ARs) or angiotensin receptors, share a common proximal signaling pathway that activates phospholipase Cß1 (PLCß1), which cleaves phosphatidylinositol 4,5-bisphosphate (PIP2) to produce inositol 1,4,5-trisphosphate (IP3) and diacylglycerol. Despite these common proximal signaling mechanisms, Gq receptors produce distinct physiological responses, yet the mechanistic basis for this remains unclear. In the heart, Gq receptors are thought to induce myocyte hypertrophy through a mechanism termed excitation-transcription coupling, which provides a mechanistic basis for compartmentalization of calcium required for contraction versus IP3-dependent intranuclear calcium required for hypertrophy. Here, we identified subcellular compartmentalization of Gq-receptor signaling as a mechanistic basis for unique Gq receptor-induced hypertrophic phenotypes in cardiac myocytes. We show that α1-ARs co-localize with PLCß1 and PIP2 at the nuclear membrane. Further, nuclear α1-ARs induced intranuclear PLCß1 activity, leading to histone deacetylase 5 (HDAC5) export and a robust transcriptional response (i.e. significant up- or down-regulation of 806 genes). Conversely, we found that angiotensin receptors localize to the sarcolemma and induce sarcolemmal PLCß1 activity, but fail to promote HDAC5 nuclear export, while producing a transcriptional response that is mostly a subset of α1-AR-induced transcription. In summary, these results link Gq-receptor compartmentalization in cardiac myocytes to unique hypertrophic transcription. They suggest a new model of excitation-transcription coupling in adult cardiac myocytes that accounts for differential Gq-receptor localization and better explains distinct physiological functions of Gq receptors.


Subject(s)
Cardiomegaly/pathology , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Myocytes, Cardiac/pathology , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phospholipase C beta/metabolism , Receptors, Adrenergic, alpha-1/metabolism , Signal Transduction , Active Transport, Cell Nucleus , Animals , Cardiomegaly/genetics , Cardiomegaly/metabolism , Cell Nucleus/metabolism , Cell Nucleus/pathology , Female , GTP-Binding Protein alpha Subunits, Gq-G11/analysis , Histone Deacetylases/analysis , Histone Deacetylases/metabolism , Male , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Nuclear Envelope/metabolism , Nuclear Envelope/pathology , Phenotype , Phosphatidylinositol 4,5-Diphosphate/analysis , Phospholipase C beta/analysis , Receptors, Adrenergic, alpha-1/analysis , Sarcolemma/metabolism , Sarcolemma/pathology , Transcriptional Activation
7.
Hum Mol Genet ; 26(8): 1458-1464, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28175314

ABSTRACT

Peptidyl-tRNA hydrolase 2 (PTRH2) regulates integrin-mediated pro-survival and apoptotic signaling. PTRH2 is critical in muscle development and regulates myogenic differentiation. In humans a biallelic mutation in the PTRH2 gene causes infantile-onset multisystem disease with progressive muscle weakness. We report here that the Ptrh2 knockout mouse model recapitulates the progressive congenital muscle pathology observed in patients. Ptrh2 null mice demonstrate multiple degenerating and regenerating muscle fibers, increased central nuclei, elevated creatine kinase activity and endomysial fibrosis. This progressive muscle pathology resembles the muscular dystrophy phenotype in humans and mice lacking the α7 integrin. We demonstrate that in normal muscle Ptrh2 associates in a complex with the α7ß1 integrin at the sarcolemma and Ptrh2 expression is decreased in α7 integrin null muscle. Furthermore, Ptrh2 expression is altered in skeletal muscle of classical congenital muscular dystrophy mouse models. Ptrh2 levels were up-regulated in dystrophin deficient mdx muscle, which correlates with the elevated levels of the α7ß1 integrin observed in mdx muscle and Duchenne muscular dystrophy patients. Similar to the α7 integrin, Ptrh2 expression was decreased in laminin-α2 dyW null gastrocnemius muscle. Our data establishes a PTRH2 mutation as a novel driver of congenital muscle degeneration and identifies a potential novel target to treat muscle myopathies.


Subject(s)
Carboxylic Ester Hydrolases/genetics , Integrins/genetics , Mitochondrial Proteins/genetics , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/genetics , Animals , Carboxylic Ester Hydrolases/biosynthesis , Dystrophin/genetics , Dystrophin/metabolism , Gene Expression Regulation, Developmental , Humans , Integrins/biosynthesis , Mice , Mice, Inbred mdx , Mice, Knockout , Mitochondrial Proteins/biosynthesis , Muscle Development/genetics , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/pathology , Sarcolemma/genetics , Sarcolemma/pathology
8.
Mol Med ; 25(1): 31, 2019 07 02.
Article in English | MEDLINE | ID: mdl-31266455

ABSTRACT

BACKGROUND: Delocalization of neuronal nitric oxide synthase (nNOS) from the sarcolemma leads to functional muscle ischemia. This contributes to the pathogenesis in cachexia, aging and muscular dystrophy. Mutations in the gene encoding dystrophin result in Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD). In many BMD patients and DMD patients that have been converted to BMD by gene therapy, sarcolemmal nNOS is missing due to the lack of dystrophin nNOS-binding domain. METHODS: Dystrophin spectrin-like repeats 16 and 17 (R16/17) is the sarcolemmal nNOS localization domain. Here we explored whether R16/17 protein therapy can restore nNOS to the sarcolemma and prevent functional ischemia in transgenic mice which expressed an R16/17-deleted human micro-dystrophin gene in the dystrophic muscle. The palmitoylated R16/17.GFP fusion protein was conjugated to various cell-penetrating peptides and produced in the baculovirus-insect cell system. The best fusion protein was delivered to the transgenic mice and functional muscle ischemia was quantified. RESULTS: Among five candidate cell-penetrating peptides, the mutant HIV trans-acting activator of transcription (TAT) protein transduction domain (mTAT) was the best in transferring the R16/17.GFP protein to the muscle. Systemic delivery of the mTAT.R16/17.GFP protein to micro-dystrophin transgenic mice successfully restored sarcolemmal nNOS without inducing T cell infiltration. More importantly, R16/17 protein therapy effectively prevented treadmill challenge-induced force loss and improved muscle perfusion during contraction. CONCLUSIONS: Our results suggest that R16/17 protein delivery is a highly promising therapy for muscle diseases involving sarcolemmal nNOS delocalizaton.


Subject(s)
Muscle, Skeletal/metabolism , Nitric Oxide Synthase Type I/metabolism , Sarcolemma/metabolism , Utrophin/metabolism , Animals , Humans , Mice , Mice, Transgenic , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/therapy , Mutation/genetics , Nitric Oxide Synthase Type I/genetics , Nitric Oxide Synthase Type I/pharmacology , Protein Binding/genetics , Sarcolemma/genetics , Sarcolemma/pathology , Utrophin/genetics
9.
J Pathol ; 246(4): 433-446, 2018 12.
Article in English | MEDLINE | ID: mdl-30066461

ABSTRACT

Skeletal muscle atrophy following unloading or immobilization represents a major invalidating event in bedridden patients. Among mechanisms involved in atrophy development, a controversial role is played by neuronal NOS (nNOS; NOS1), whose dysregulation at the protein level and/or subcellular distribution also characterizes other neuromuscular disorders. This study aimed to investigate unloading-induced changes in nNOS before any evidence of myofiber atrophy, using vastus lateralis biopsies obtained from young healthy subjects after a short bed-rest and rat soleus muscles after exposure to short unloading periods. Our results showed that (1) changes in nNOS subcellular distribution using NADPH-diaphorase histochemistry to detect enzyme activity were observed earlier than using immunofluorescence to visualize the protein; (2) loss of active nNOS from the physiological subsarcolemmal localization occurred before myofiber atrophy, i.e. in 8-day bed-rest biopsies and in 6 h-unloaded rat soleus, and was accompanied by increased nNOS activity in the sarcoplasm; (3) nNOS (Nos1) transcript and protein levels decreased significantly in the rat soleus after 6 h and 1 day unloading, respectively, to return to ambulatory levels after 4 and 7 days of unloading, respectively; (4) unloading-induced nNOS redistribution appeared dependent on mitochondrial-derived oxidant species, indirectly measured by tropomyosin disulfide bonds which had increased significantly in the rat soleus already after a 6 h-unloading bout; (5) activity of displaced nNOS molecules is required for translocation of the FoxO3 transcription factor to myofiber nuclei. FoxO3 nuclear localization in rat soleus increased after 6 h unloading (about four-fold the ambulatory level), whereas it did not when nNOS expression and activity were inhibited in vivo before and during 6 h unloading. In conclusion, this study demonstrates that the redistribution of active nNOS molecules from sarcolemma to sarcoplasm not only is ahead of the atrophy of unloaded myofibers, and is induced by increased production of mitochondrial superoxide anion, but also drives FoxO3 activation to initiate muscle atrophy. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Muscular Atrophy/enzymology , Nitric Oxide Synthase Type I/metabolism , Oxidative Stress , Quadriceps Muscle/enzymology , Sarcolemma/enzymology , Animals , Bed Rest , Disease Models, Animal , Down-Regulation , Female , Forkhead Box Protein O3/metabolism , Healthy Volunteers , Hindlimb Suspension , Humans , Male , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Muscular Atrophy/physiopathology , NADP/metabolism , Nitric Oxide Synthase Type I/genetics , Protein Transport , Quadriceps Muscle/pathology , Quadriceps Muscle/physiopathology , Rats, Wistar , Sarcolemma/pathology , Superoxides/metabolism , Time Factors
10.
Molecules ; 24(9)2019 May 07.
Article in English | MEDLINE | ID: mdl-31067690

ABSTRACT

Out-of-hospital sudden cardiac arrest is a major public health problem with an overall survival of less than 5%. Upon cardiac arrest, cessation of coronary blood flow rapidly leads to intense myocardial ischemia and activation of the sarcolemmal Na+-H+ exchanger isoform-1 (NHE-1). NHE-1 activation drives Na+ into cardiomyocytes in exchange for H+ with its exchange rate intensified upon reperfusion during the resuscitation effort. Na+ accumulates in the cytosol driving Ca2+ entry through the Na+-Ca2+ exchanger, eventually causing cytosolic and mitochondrial Ca2+ overload and worsening myocardial injury by compromising mitochondrial bioenergetic function. We have reported clinically relevant myocardial effects elicited by NHE-1 inhibitors given during resuscitation in animal models of ventricular fibrillation (VF). These effects include: (a) preservation of left ventricular distensibility enabling hemodynamically more effective chest compressions, (b) return of cardiac activity with greater electrical stability reducing post-resuscitation episodes of VF, (c) less post-resuscitation myocardial dysfunction, and (d) attenuation of adverse myocardial effects of epinephrine; all contributing to improved survival in animal models. Mechanistically, NHE-1 inhibition reduces adverse effects stemming from Na+-driven cytosolic and mitochondrial Ca2+ overload. We believe the preclinical work herein discussed provides a persuasive rationale for examining the potential role of NHE-1 inhibitors for cardiac resuscitation in humans.


Subject(s)
Heart Arrest/drug therapy , Myocardial Ischemia/genetics , Sodium-Hydrogen Exchangers/genetics , Ventricular Fibrillation/drug therapy , Calcium/metabolism , Calcium Signaling/drug effects , Calcium Signaling/genetics , Heart Arrest/genetics , Heart Arrest/pathology , Humans , Models, Animal , Myocardial Ischemia/drug therapy , Myocardial Ischemia/pathology , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Sarcolemma/metabolism , Sarcolemma/pathology , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sodium-Hydrogen Exchangers/metabolism , Ventricular Fibrillation/genetics , Ventricular Fibrillation/pathology
11.
Hum Mol Genet ; 25(10): 1900-1911, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26911675

ABSTRACT

Limb-girdle muscular dystrophies are a genetically diverse group of diseases characterized by chronic muscle wasting and weakness. Recessive mutations in ANO5 (TMEM16E) have been directly linked to several clinical phenotypes including limb-girdle muscular dystrophy type 2L and Miyoshi myopathy type 3, although the pathogenic mechanism has remained elusive. ANO5 is a member of the Anoctamin/TMEM16 superfamily that encodes both ion channels and regulators of membrane phospholipid scrambling. The phenotypic overlap of ANO5 myopathies with dysferlin-associated muscular dystrophies has inspired the hypothesis that ANO5, like dysferlin, may be involved in the repair of muscle membranes following injury. Here we show that Ano5-deficient mice have reduced capacity to repair the sarcolemma following laser-induced damage, exhibit delayed regeneration after cardiotoxin injury and suffer from defective myoblast fusion necessary for the proper repair and regeneration of multinucleated myotubes. Together, these data suggest that ANO5 plays an important role in sarcolemmal membrane dynamics. Genbank Mouse Genome Informatics accession no. 3576659.


Subject(s)
Chloride Channels/genetics , Distal Myopathies/genetics , Muscular Atrophy/genetics , Muscular Dystrophies, Limb-Girdle/genetics , Animals , Anoctamins , Disease Models, Animal , Humans , Mice , Mice, Knockout , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/pathology , Muscular Dystrophies, Limb-Girdle/pathology , Mutation , Sarcolemma/pathology
12.
Hum Mol Genet ; 25(16): 3555-3563, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27378686

ABSTRACT

In preclinical models for Duchenne muscular dystrophy, dystrophin restoration during adeno-associated virus (AAV)-U7-mediated exon-skipping therapy was shown to decrease drastically after six months in treated muscles. This decline in efficacy is strongly correlated with the loss of the therapeutic AAV genomes, probably due to alterations of the dystrophic myofiber membranes. To improve the membrane integrity of the dystrophic myofibers at the time of AAV-U7 injection, mdx muscles were pre-treated with a single dose of the peptide-phosphorodiamidate morpholino (PPMO) antisense oligonucleotides that induced temporary dystrophin expression at the sarcolemma. The PPMO pre-treatment allowed efficient maintenance of AAV genomes in mdx muscles and enhanced the AAV-U7 therapy effect with a ten-fold increase of the protein level after 6 months. PPMO pre-treatment was also beneficial to AAV-mediated gene therapy with transfer of micro-dystrophin cDNA into muscles. Therefore, avoiding vector genome loss after AAV injection by PPMO pre-treatment would allow efficient long-term restoration of dystrophin and the use of lower and thus safer vector doses for Duchenne patients.


Subject(s)
Dystrophin/genetics , Genetic Therapy , Morpholinos/administration & dosage , Muscular Dystrophy, Animal/therapy , Muscular Dystrophy, Duchenne/therapy , Oligonucleotides, Antisense/administration & dosage , Animals , Dependovirus/genetics , Exons/genetics , Gene Transfer Techniques , Genetic Vectors/administration & dosage , Humans , Mice, Inbred mdx , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Duchenne/genetics , Sarcolemma/drug effects , Sarcolemma/pathology
13.
Hum Mol Genet ; 25(24): 5395-5406, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27798107

ABSTRACT

Duchenne muscular dystrophy (DMD) is a genetic disorder that causes progressive muscle weakness, ultimately leading to early mortality in affected teenagers and young adults. Previous work from our lab has shown that a small transmembrane protein called sarcospan (SSPN) can enhance the recruitment of adhesion complex proteins to the cell surface. When human SSPN is expressed at three-fold levels in mdx mice, this increase in adhesion complex abundance improves muscle membrane stability, preventing many of the histopathological changes associated with DMD. However, expressing higher levels of human SSPN (ten-fold transgenic expression) causes a severe degenerative muscle phenotype in wild-type mice. Since SSPN-mediated stabilization of the sarcolemma represents a promising therapeutic strategy in DMD, it is important to determine whether SSPN can be introduced at high levels without toxicity. Here, we show that mouse SSPN (mSSPN) can be overexpressed at 30-fold levels in wild-type mice with no deleterious effects. In mdx mice, mSSPN overexpression improves dystrophic pathology and sarcolemmal stability. We show that these mice exhibit increased resistance to eccentric contraction-induced damage and reduced fatigue following exercise. mSSPN overexpression improved pulmonary function and reduced dystrophic histopathology in the diaphragm. Together, these results demonstrate that SSPN overexpression is well tolerated in mdx mice and improves sarcolemma defects that underlie skeletal muscle and pulmonary dysfunction in DMD.


Subject(s)
Carrier Proteins/genetics , Membrane Proteins/genetics , Muscular Dystrophy, Duchenne/genetics , Neoplasm Proteins/genetics , Sarcolemma/genetics , Animals , Carrier Proteins/biosynthesis , Disease Models, Animal , Gene Expression Regulation/genetics , Humans , Lung Diseases/genetics , Lung Diseases/pathology , Membrane Proteins/biosynthesis , Mice , Mice, Inbred mdx , Mice, Transgenic , Muscle Contraction/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Neoplasm Proteins/biosynthesis , Sarcolemma/pathology
14.
Biochim Biophys Acta Mol Basis Dis ; 1864(5 Pt B): 1923-1930, 2018 May.
Article in English | MEDLINE | ID: mdl-29066284

ABSTRACT

Amylin is a pancreatic ß-cell hormone co-secreted with insulin, plays a role in normal glucose homeostasis, and forms amyloid in the pancreatic islets of individuals with type-2 diabetes. Aggregated amylin is also found in blood and extra-pancreatic tissues, including myocardium. Myocardial amylin accumulation is associated with myocyte Ca2+ dysregulation in diabetic rats expressing human amylin. Whether deposition of amylin in the heart is a consequence of or a contributor to diabetic cardiomyopathy remains unknown. We used amylin knockout (AKO) mice intravenously infused with either human amylin (i.e, the aggregated form) or non-amyloidogenic (i.e., monomeric) rodent amylin to test the hypothesis that aggregated amylin accumulates in the heart in the absence of diabetes. AKO mice infused with human amylin, but not rodent amylin, showed amylin deposits in the myocardium. Cardiac amylin level was larger in males compared to females. Sarcolemmal Ca2+ leak and Ca2+ transients were increased in myocytes isolated from males infused with human amylin while no significant changes occurred in either females injected with human amylin or in rat amylin-infused mice. In isolated cardiac myocytes, the amylin receptor antagonist AC-187 did not effectively block the interaction of amylin with the sarcolemma. In conclusion, circulating aggregated amylin accumulates preferentially in male vs. female hearts and its effects on myocyte Ca2+ cycling do not require diabetic remodeling of the myocardium. This article is part of a Special issue entitled Cardiac adaptations to obesity, diabetes and insulin resistance, edited by Professors Jan F.C. Glatz, Jason R.B. Dyck and Christine Des Rosiers.


Subject(s)
Calcium Signaling , Calcium/metabolism , Diabetic Cardiomyopathies/metabolism , Islet Amyloid Polypeptide/metabolism , Myocytes, Cardiac/metabolism , Sarcolemma/metabolism , Ventricular Remodeling , Animals , Diabetic Cardiomyopathies/genetics , Diabetic Cardiomyopathies/pathology , Diabetic Cardiomyopathies/physiopathology , Disease Models, Animal , Female , Inflammation Mediators/blood , Infusions, Intravenous , Interleukin-1beta/blood , Islet Amyloid Polypeptide/administration & dosage , Islet Amyloid Polypeptide/deficiency , Islet Amyloid Polypeptide/genetics , Male , Mice, Knockout , Myocytes, Cardiac/pathology , Protein Aggregates , Protein Aggregation, Pathological , Sarcolemma/pathology , Sex Factors
15.
Muscle Nerve ; 58(2): 286-292, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29603301

ABSTRACT

INTRODUCTION: GNE myopathy is an adult-onset muscle disorder characterized by impaired sialylation of (muscle) glycans, detectable by lectin histochemistry. We describe a standardized method to quantify (lectin-) fluorescence in muscle sections, applicable for diagnosis and response to therapy for GNE myopathy. METHODS: Muscle sections were fluorescently labeled with the sialic acid-binding Sambucus nigra agglutinin (SNA) lectin and antibodies to sarcolemma residence protein caveolin-3 (CAV-3). Entire tissue sections were imaged in tiles and fluorescence was quantified. RESULTS: SNA fluorescence co-localizing with CAV-3 was ∼50% decreased in GNE myopathy biopsies compared with muscle-matched controls, confirming previous qualitative results. DISCUSSION: This quantitative fluorescence method can accurately determine sialylation status of GNE myopathy muscle biopsies. This method is adaptable for expression of other membrane-associated muscle proteins, and may be of benefit for disorders in which therapeutic changes in expression are subtle and difficult to assess by other methods. Muscle Nerve 58: 286-292, 2018.


Subject(s)
Distal Myopathies/pathology , Lectins , Muscle, Skeletal/pathology , Adult , Caveolin 3/genetics , Distal Myopathies/genetics , Female , Fluorescent Dyes , Humans , Image Processing, Computer-Assisted , Male , Microscopy, Confocal , Middle Aged , Plant Lectins , Ribosome Inactivating Proteins , Sarcolemma/pathology , Sarcolemma/ultrastructure
16.
J Biol Chem ; 291(19): 9920-8, 2016 May 06.
Article in English | MEDLINE | ID: mdl-26966179

ABSTRACT

Duchenne muscular dystrophy (DMD) is an X-linked recessive disease caused by mutations in the gene encoding dystrophin. Loss of dystrophin protein compromises the stability of the sarcolemma membrane surrounding each muscle cell fiber, leading to membrane ruptures and leakiness that induces myofiber necrosis, a subsequent inflammatory response, and progressive tissue fibrosis with loss of functional capacity. Cathepsin S (Ctss) is a cysteine protease that is actively secreted in areas of tissue injury and ongoing inflammation, where it participates in extracellular matrix remodeling and healing. Here we show significant induction of Ctss expression and proteolytic activity following acute muscle injury or in muscle from mdx mice, a model of DMD. To examine the functional ramifications associated with greater Ctss expression, the Ctss gene was deleted in the mdx genetic background, resulting in protection from muscular dystrophy pathogenesis that included reduced myofiber turnover and histopathology, reduced fibrosis, and improved running capacity. Mechanistically, deletion of the Ctss gene in the mdx background significantly increased myofiber sarcolemmal membrane stability with greater expression and membrane localization of utrophin, integrins, and ß-dystroglycan, which anchor the membrane to the basal lamina and underlying cytoskeletal proteins. Consistent with these results, skeletal muscle-specific transgenic mice overexpressing Ctss showed increased myofiber necrosis, muscle histopathology, and a functional deficit reminiscent of muscular dystrophy. Hence, Ctss induction during muscular dystrophy is a pathologic event that partially underlies disease pathogenesis, and its inhibition might serve as a new therapeutic strategy in DMD.


Subject(s)
Cathepsins/biosynthesis , Gene Expression Regulation, Developmental , Muscle Fibers, Skeletal/enzymology , Muscular Dystrophy, Animal/enzymology , Muscular Dystrophy, Duchenne/enzymology , Animals , Cytoskeleton/enzymology , Cytoskeleton/genetics , Cytoskeleton/pathology , Mice , Mice, Inbred mdx , Mice, Knockout , Muscle Fibers, Skeletal/pathology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/pathology , Necrosis , Proteolysis , Sarcolemma/enzymology , Sarcolemma/genetics , Sarcolemma/pathology
17.
Biochim Biophys Acta ; 1863(9): 2267-79, 2016 09.
Article in English | MEDLINE | ID: mdl-27286750

ABSTRACT

Defect in membrane repair contributes to the development of limb girdle muscular dystrophy type 2B (LGMD2B) and Miyoshi myopathy. In healthy skeletal muscle, unraveling membrane repair mechanisms requires to establish an exhaustive list of the components of the resealing machinery. Here we show that human myotubes rendered deficient for Annexin-A5 (AnxA5) suffer from a severe defect in membrane resealing. This defect is rescued by the addition of recombinant AnxA5 while an AnxA5 mutant, which is unable to form 2D protein arrays, has no effect. Using correlative light and electron microscopy, we show that AnxA5 binds to the edges of the torn membrane, as early as a few seconds after sarcolemma injury, where it probably self-assembles into 2D arrays. In addition, we observed that membrane resealing is associated with the presence of a cluster of lipid vesicles at the wounded site. AnxA5 is present at the surface of these vesicles and may thus participate in plugging the cell membrane disruption. Finally, we show that AnxA5 behaves similarly in myotubes from a muscle cell line established from a patient suffering from LGMD2B, a myopathy due to dysferlin mutations, which indicates that trafficking of AnxA5 during sarcolemma damage is independent of the presence of dysferlin.


Subject(s)
Annexin A5/metabolism , Cell Membrane/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Wound Healing , Adult , Annexin A5/ultrastructure , Cell Line , Dysferlin , Extracellular Space/metabolism , Humans , Lasers , Lipid Bilayers/metabolism , Membrane Proteins/deficiency , Membrane Proteins/metabolism , Muscle Fibers, Skeletal/ultrastructure , Muscle Proteins/deficiency , Muscle Proteins/metabolism , Muscular Dystrophies, Limb-Girdle/metabolism , Muscular Dystrophies, Limb-Girdle/pathology , Mutation/genetics , Myoblasts/metabolism , Myoblasts/pathology , Recombinant Proteins/metabolism , Sarcolemma/pathology , Subcellular Fractions/metabolism
18.
Biochim Biophys Acta ; 1863(7 Pt B): 1839-47, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26578114

ABSTRACT

Cardiac transverse tubules (t-tubules) are specific membrane organelles critical in calcium signaling and excitation-contraction coupling required for beat-to-beat heart contraction. T-tubules are highly branched and form an interconnected network that penetrates the myocyte interior to form junctions with the sarcoplasmic reticulum. T-tubules are selectively enriched with specific ion channels and proteins crucial in calcium transient development necessary in excitation-contraction coupling, thus t-tubules are a key component of cardiac myocyte function. In this review, we focus primarily on two proteins concentrated within the t-tubular network, the L-type calcium channel (LTCC) and associated membrane anchor protein, bridging integrator 1 (BIN1). Here, we provide an overview of current knowledge in t-tubule morphology, composition, microdomains, as well as the dynamics of the t-tubule network. Secondly, we highlight multiple aspects of BIN1-dependent t-tubule function, which includes forward trafficking of LTCCs to t-tubules, LTCC clustering at t-tubule surface, microdomain organization and regulation at t-tubule membrane, and the formation of a slow diffusion barrier within t-tubules. Lastly, we describe progress in characterizing how acquired human heart failure can be attributed to abnormal BIN1 transcription and associated t-tubule remodeling. Understanding BIN1-regulated cardiac t-tubule biology in human heart failure management has the dual benefit of promoting progress in both biomarker development and therapeutic target identification. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Integration of Developmental and Environmental Cues in the Heart edited by Marcus Schaub and Hughes Abriel.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Calcium Channels, L-Type/metabolism , Membrane Microdomains/metabolism , Myocytes, Cardiac/metabolism , Nuclear Proteins/metabolism , Sarcolemma/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Calcium Signaling , Genetic Predisposition to Disease , Heart Failure/genetics , Heart Failure/metabolism , Heart Failure/pathology , Heart Failure/physiopathology , Humans , Membrane Microdomains/genetics , Membrane Potentials , Myocytes, Cardiac/pathology , Protein Binding , Protein Transport , Risk Factors , Sarcolemma/pathology , Transcription, Genetic
19.
Hum Mol Genet ; 24(22): 6428-45, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26358775

ABSTRACT

Duchenne muscular dystrophy (DMD) is a genetic disease characterized by progressive muscle degeneration due to mutations in the dystrophin gene. In spite of great advances in the design of curative treatments, most patients currently receive palliative therapies with steroid molecules such as prednisone or deflazacort thought to act through their immunosuppressive properties. These molecules only slightly slow down the progression of the disease and lead to severe side effects. Fundamental research is still needed to reveal the mechanisms involved in the disease that could be exploited as therapeutic targets. By studying a Caenorhabditis elegans model for DMD, we show here that dystrophin-dependent muscle degeneration is likely to be cell autonomous and affects the muscle cells the most involved in locomotion. We demonstrate that muscle degeneration is dependent on exercise and force production. Exhaustive studies by electron microscopy allowed establishing for the first time the chronology of subcellular events occurring during the entire process of muscle degeneration. This chronology highlighted the crucial role for dystrophin in stabilizing sarcomeric anchoring structures and the sarcolemma. Our results suggest that the disruption of sarcomeric anchoring structures and sarcolemma integrity, observed at the onset of the muscle degeneration process, triggers subcellular consequences that lead to muscle cell death. An ultra-structural analysis of muscle biopsies from DMD patients suggested that the chronology of subcellular events established in C. elegans models the pathogenesis in human. Finally, we found that the loss of sarcolemma integrity was greatly reduced after prednisone treatment suggesting a role for this molecule in plasma membrane stabilization.


Subject(s)
Muscular Dystrophy, Duchenne/pathology , Sarcolemma/ultrastructure , Sarcomeres/pathology , Animals , Animals, Genetically Modified , Caenorhabditis elegans , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/metabolism , Disease Models, Animal , Dystrophin/genetics , Dystrophin/metabolism , Humans , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Mutation , Sarcolemma/metabolism , Sarcolemma/pathology , Sarcomeres/metabolism , Sarcomeres/ultrastructure
20.
Cardiovasc Diabetol ; 16(1): 155, 2017 12 04.
Article in English | MEDLINE | ID: mdl-29202762

ABSTRACT

Cardiovascular disease, predominantly ischemic heart disease (IHD), is the leading cause of death in diabetes mellitus (DM). In addition to eliciting cardiomyopathy, DM induces a 'wicked triumvirate': (i) increasing the risk and incidence of IHD and myocardial ischemia; (ii) decreasing myocardial tolerance to ischemia-reperfusion (I-R) injury; and (iii) inhibiting or eliminating responses to cardioprotective stimuli. Changes in ischemic tolerance and cardioprotective signaling may contribute to substantially higher mortality and morbidity following ischemic insult in DM patients. Among the diverse mechanisms implicated in diabetic impairment of ischemic tolerance and cardioprotection, changes in sarcolemmal makeup may play an overarching role and are considered in detail in the current review. Observations predominantly in animal models reveal DM-dependent changes in membrane lipid composition (cholesterol and triglyceride accumulation, fatty acid saturation vs. reduced desaturation, phospholipid remodeling) that contribute to modulation of caveolar domains, gap junctions and T-tubules. These modifications influence sarcolemmal biophysical properties, receptor and phospholipid signaling, ion channel and transporter functions, contributing to contractile and electrophysiological dysfunction, cardiomyopathy, ischemic intolerance and suppression of protective signaling. A better understanding of these sarcolemmal abnormalities in types I and II DM (T1DM, T2DM) can inform approaches to limiting cardiomyopathy, associated IHD and their consequences. Key knowledge gaps include details of sarcolemmal changes in models of T2DM, temporal patterns of lipid, microdomain and T-tubule changes during disease development, and the precise impacts of these diverse sarcolemmal modifications. Importantly, exercise, dietary, pharmacological and gene approaches have potential for improving sarcolemmal makeup, and thus myocyte function and stress-resistance in this ubiquitous metabolic disorder.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetic Cardiomyopathies/metabolism , Membrane Microdomains/metabolism , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/metabolism , Myocytes, Cardiac/metabolism , Animals , Anticholesteremic Agents/therapeutic use , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 1/physiopathology , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/physiopathology , Diabetic Cardiomyopathies/pathology , Diabetic Cardiomyopathies/physiopathology , Diabetic Cardiomyopathies/prevention & control , Diet/adverse effects , Disease Models, Animal , Energy Metabolism , Exercise , Humans , Hypoglycemic Agents/adverse effects , Membrane Lipids/metabolism , Membrane Microdomains/drug effects , Membrane Microdomains/pathology , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Prognosis , Protective Factors , Risk Factors , Sarcolemma/metabolism , Sarcolemma/pathology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL