Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 13.378
Filter
1.
Front Biosci (Schol Ed) ; 16(2): 12, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38939976

ABSTRACT

Several inherited metabolic fatty acid disorders present with myopathies. Skeletal muscle accounts for 40% of the body and is important for metabolism, exercise, and movement. Muscle energy failure is manifested by metabolic crises with muscle weakness, sometimes associated with muscle fatigue and failure resulting in acute necrosis or rhabdomyolysis/myoglobinuria episodes. Lack of energy leads to muscle necrosis. Other presentations are weakness and myalgias with lipid storage myopathies in the biopsy. The biomarkers of such disorders are acyl-carnitine with various profiles and need to be carefully evaluated to plan supplementary therapy and specific diets. If red flags are not distinctly followed and diagnosed in time they might lead to a metabolic or cardiac failure.


Subject(s)
Carnitine , Lipid Metabolism, Inborn Errors , Muscular Diseases , Humans , Muscular Diseases/metabolism , Muscular Diseases/therapy , Muscular Diseases/genetics , Carnitine/metabolism , Carnitine/analogs & derivatives , Lipid Metabolism, Inborn Errors/genetics , Lipid Metabolism, Inborn Errors/therapy , Lipid Metabolism, Inborn Errors/metabolism , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophies
3.
J Neurol ; 271(7): 4724-4726, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38900296
4.
Sci Rep ; 14(1): 14757, 2024 06 26.
Article in English | MEDLINE | ID: mdl-38926599

ABSTRACT

Muscular dystrophy is a group of genetic disorders that lead to muscle wasting and loss of muscle function. Identifying genetic modifiers that alleviate symptoms or enhance the severity of a primary disease helps to understand mechanisms behind disease pathology and facilitates discovery of molecular targets for therapy. Several muscular dystrophies are caused by genetic defects in the components of the dystrophin-glycoprotein adhesion complex (DGC). Thrombospondin-4 overexpression has been shown to mitigate dystrophic disease in mouse models for Duchenne muscular dystrophy (dystrophin deficiency) and limb-girdle muscular dystrophy type 2F (LGMD2F, δ-sarcoglycan deficiency), while deletion of the thrombospondin-4 gene exacerbated the diseases. Hence, thrombospondin-4 has been considered a candidate molecule for therapy of muscular dystrophies involving the DGC. We have investigated whether thrombospondin-4 could act as a genetic modifier for other DGC-associated diseases: limb-girdle muscular dystrophy type 2E (LGMD2E, ß-sarcoglycan deficiency) and laminin α2 chain-deficient muscular dystrophy (LAMA2-RD). Deletion of the thrombospondin-4 gene in mouse models for LGMD2E and LAMA2-RD, respectively, did not result in worsening of the dystrophic phenotype. Loss of thrombospondin-4 did not enhance sarcolemma damage and did not impair trafficking of transmembrane receptors integrin α7ß1 and dystroglycan in double knockout muscles. Our results suggest that thrombospondin-4 might not be a relevant therapeutic target for all muscular dystrophies involving the DGC. This data also demonstrates that molecular pathology between very similar diseases like LGMD2E and 2F can differ significantly.


Subject(s)
Laminin , Mice, Knockout , Sarcoglycans , Thrombospondins , Animals , Laminin/metabolism , Laminin/genetics , Laminin/deficiency , Sarcoglycans/genetics , Sarcoglycans/deficiency , Sarcoglycans/metabolism , Mice , Thrombospondins/genetics , Thrombospondins/metabolism , Thrombospondins/deficiency , Disease Models, Animal , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Gene Deletion , Muscular Dystrophies/genetics , Muscular Dystrophies/metabolism , Muscular Dystrophies/pathology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Animal/pathology
5.
Int J Mol Sci ; 25(11)2024 May 27.
Article in English | MEDLINE | ID: mdl-38892025

ABSTRACT

Pathogenic variants in LMNA have been associated with a wide spectrum of muscular conditions: the laminopathies. LMNA-related congenital muscular dystrophy is a laminopathy characterised by the early onset of symptoms and often leads to a fatal outcome at young ages. Children face a heightened risk of malignant arrhythmias. No established paediatric protocols for managing this condition are available. We review published cases and provide insights into disease progression in two twin sisters with LMNA-related muscular dystrophy. Our objective is to propose a cardiac surveillance and management plan tailored specifically for paediatric patients. We present a family of five members, including two twin sisters with LMNA-related muscular dystrophy. A comprehensive neuromuscular and cardiac work-up was performed in all family members. Genetic analysis using massive sequencing technology was performed in both twins. Clinical assessment showed that only the twins showed diagnoses of LMNA-related muscular dystrophy. Follow-up showed an early onset of symptoms and life-threatening arrhythmias, with differing disease progressions despite both twins passing away. Genetic analysis identified a de novo rare missense deleterious variant in the LMNA gene. Other additional rare variants were identified in genes associated with myasthenic syndrome. Early-onset neuromuscular symptoms could be related to a prognosis of worse life-threatening arrhythmias in LMNA related muscular dystrophy. Being a carrier of other rare variants may be a modifying factor in the progression of the phenotype, although further studies are needed. There is a pressing need for specific cardiac recommendations tailored to the paediatric population to mitigate the risk of malignant arrhythmias.


Subject(s)
Lamin Type A , Muscular Dystrophies , Twins, Monozygotic , Humans , Lamin Type A/genetics , Twins, Monozygotic/genetics , Female , Muscular Dystrophies/genetics , Muscular Dystrophies/therapy , Male , Child , Pedigree , Child, Preschool , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/etiology
6.
Int J Mol Sci ; 25(9)2024 May 03.
Article in English | MEDLINE | ID: mdl-38732204

ABSTRACT

The extraocular muscles (EOMs) possess unique characteristics that set them apart from other skeletal muscles. These muscles, responsible for eye movements, exhibit remarkable resistance to various muscular dystrophies and aging, presenting a significant contrast to the vulnerability of skeletal muscles to these conditions. In this review, we delve into the cellular and molecular underpinnings of the distinct properties of EOMs. We explore their structural complexity, highlighting differences in fiber types, innervation patterns, and developmental origins. Notably, EOM fibers express a diverse array of myosin heavy-chain isoforms, retaining embryonic forms into adulthood. Moreover, their motor innervation is characterized by a high ratio of nerve fibers to muscle fibers and the presence of unique neuromuscular junctions. These features contribute to the specialized functions of EOMs, including rapid and precise eye movements. Understanding the mechanisms behind the resilience of EOMs to disease and aging may offer insights into potential therapeutic strategies for treating muscular dystrophies and myopathies affecting other skeletal muscles.


Subject(s)
Aging , Oculomotor Muscles , Humans , Oculomotor Muscles/physiology , Aging/physiology , Animals , Muscular Dystrophies , Neuromuscular Junction/physiology , Neuromuscular Junction/metabolism , Muscle, Skeletal/physiology , Muscle, Skeletal/metabolism
7.
BMC Anesthesiol ; 24(1): 173, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38730355

ABSTRACT

BACKGROUND: Bethlem Myopathy is a collagen VI-related myopathy presenting as a rare hereditary muscular disorder with progressive muscular weakness and joint contractures. Despite its milder clinical course relative to other myopathies, anaesthetic management can be challenging. High arched palates and fixed flexion deformities may contribute to a difficult airway. A progressive decline in pulmonary function can present later into adulthood. This respiratory decline can carry secondary cardiovascular consequences due to the progressive nature of restrictive lung disease, including right sided heart disease and pulmonary hypertension. We describe a case of a male patient with Bethlem Myopathy undergoing anaesthesia, to contribute to the limited body of literature on this condition and enhance awareness and guidance amongst anaesthesiologists on approaching patients with this condition. This is the first case report within the literature of its kind. CASE PRESENTATION: This case details a 33-year-old male with Bethlem Myopathy undergoing tonsillectomy. Diagnosed in childhood following developmental delays, the patient had no prior anaesthetic exposure and no family history of anaesthetic complications. Anaesthetic induction was achieved without complications, avoiding depolarizing muscle relaxants and careful airway management. Extreme care was taken in patient positioning to prevent complications. The surgery proceeded without incident and muscle paralysis was reversed with Suggammadex, resulting in no adverse post-operative respiratory complications. The patient was discharged on the first post-operative day without any respiratory or cardiovascular compromise. CONCLUSIONS: Bethlem Myopathy, while often exhibiting a mild clinical course, can present anaesthetic challenges. Awareness of potential complications including a difficult airway, cardiovascular and respiratory implications as well as the need for specialised monitoring and positioning is crucial to ensure a safe peri-operative course.


Subject(s)
Tonsillectomy , Humans , Male , Adult , Tonsillectomy/methods , Anesthesia/methods , Contracture/surgery , Elective Surgical Procedures , Muscular Dystrophies/complications , Muscular Dystrophies/surgery , Muscular Dystrophies/congenital
8.
Int J Mol Sci ; 25(10)2024 May 13.
Article in English | MEDLINE | ID: mdl-38791328

ABSTRACT

Inherited muscular diseases (MDs) are genetic degenerative disorders typically caused by mutations in a single gene that affect striated muscle and result in progressive weakness and wasting in affected individuals. Cardiac muscle can also be involved with some variability that depends on the genetic basis of the MD (Muscular Dystrophy) phenotype. Heart involvement can manifest with two main clinical pictures: left ventricular systolic dysfunction with evolution towards dilated cardiomyopathy and refractory heart failure, or the presence of conduction system defects and serious life-threatening ventricular arrhythmias. The two pictures can coexist. In these cases, heart transplantation (HTx) is considered the most appropriate option in patients who are not responders to the optimized standard therapeutic protocols. However, cardiac transplant is still considered a relative contraindication in patients with inherited muscle disorders and end-stage cardiomyopathies. High operative risk related to muscle impairment and potential graft involvement secondary to the underlying myopathy have been the two main reasons implicated in the generalized reluctance to consider cardiac transplant as a viable option. We report an overview of cardiac involvement in MDs and its possible association with the underlying molecular defect, as well as a systematic review of HTx outcomes in patients with MD-related end-stage dilated cardiomyopathy, published so far in the literature.


Subject(s)
Cardiomyopathy, Dilated , Heart Transplantation , Muscular Dystrophies , Humans , Cardiomyopathy, Dilated/surgery , Heart Transplantation/methods , Muscular Dystrophies/complications
9.
Life Sci Alliance ; 7(8)2024 Aug.
Article in English | MEDLINE | ID: mdl-38749543

ABSTRACT

Phosphatidylcholine (PC) is the major membrane phospholipid in most eukaryotic cells. Bi-allelic loss of function variants in CHKB, encoding the first step in the synthesis of PC, is the cause of a rostrocaudal muscular dystrophy in both humans and mice. Loss of sarcolemma integrity is a hallmark of muscular dystrophies; however, how this occurs in the absence of choline kinase function is not known. We determine that in Chkb -/- mice there is a failure of the α7ß1 integrin complex that is specific to affected muscle. We observed that in Chkb -/- hindlimb muscles there is a decrease in sarcolemma association/abundance of the PI(4,5)P2 binding integrin complex proteins vinculin, and α-actinin, and a decrease in actin association with the sarcolemma. In cells, pharmacological inhibition of choline kinase activity results in internalization of a fluorescent PI(4,5)P2 reporter from discrete plasma membrane clusters at the cell surface membrane to cytosol, this corresponds with a decreased vinculin localization at plasma membrane focal adhesions that was rescued by overexpression of CHKB.


Subject(s)
Choline Kinase , Integrins , Mice, Knockout , Muscular Dystrophies , Sarcolemma , Vinculin , Animals , Mice , Vinculin/metabolism , Vinculin/genetics , Muscular Dystrophies/metabolism , Muscular Dystrophies/genetics , Integrins/metabolism , Choline Kinase/metabolism , Choline Kinase/genetics , Sarcolemma/metabolism , Humans , Focal Adhesions/metabolism , Cell Membrane/metabolism , Actinin/metabolism , Actinin/genetics , Muscle, Skeletal/metabolism , Phosphatidylinositol 4,5-Diphosphate/metabolism , Actins/metabolism , Disease Models, Animal
10.
Sci Rep ; 14(1): 11225, 2024 05 16.
Article in English | MEDLINE | ID: mdl-38755190

ABSTRACT

Muscular dystrophies (MDs) are inherited genetic diseases causing weakness and degeneration of muscles. The distribution of muscle weakness differs between MDs, involving distal muscles or proximal muscles. While the mutations in most of the MD-associated genes lead to either distal or proximal onset, there are also genes whose mutations can cause both types of onsets. We hypothesized that the genes associated with different MD onsets code proteins with distinct cellular functions. To investigate this, we collected the MD-associated genes and assigned them to three onset groups: genes mutated only in distal onset dystrophies, genes mutated only in proximal onset dystrophies, and genes mutated in both types of onsets. We then systematically evaluated the cellular functions of these gene sets with computational strategies based on functional enrichment analysis and biological network analysis. Our analyses demonstrate that genes mutated in either distal or proximal onset MDs code proteins linked with two distinct sets of cellular processes. Interestingly, these two sets of cellular processes are relevant for the genes that are associated with both onsets. Moreover, the genes associated with both onsets display high centrality and connectivity in the network of muscular dystrophy genes. Our findings support the hypothesis that the proteins associated with distal or proximal onsets have distinct functional characteristics, whereas the proteins associated with both onsets are multifunctional.


Subject(s)
Muscle Weakness , Muscular Dystrophies , Mutation , Humans , Muscular Dystrophies/genetics , Muscle Weakness/genetics , Gene Regulatory Networks , Computational Biology/methods , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Muscle, Skeletal/pathology
11.
Muscle Nerve ; 70(2): 273-278, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38783566

ABSTRACT

INTRODUCTION/AIMS: Early diagnosis of a chronic neuromuscular disease such as muscular dystrophy (MD) generally excludes an individual from active-duty military service. However, it is not known whether veterans are sometimes diagnosed with milder forms of MD at a later timepoint. We aimed to determine the prevalence of MD in a veterans health system. METHODS: We abstracted clinical and genetic test data on patients who received care for a diagnosis of MD at the North Florida/South Georgia Veterans Health System between 2008 and 2021. We then determined which of these individuals would meet criteria for a definite diagnosis of MD, based on electrodiagnostic testing, muscle biopsy, and genetic testing of the individual or an affected first degree relative. RESULTS: We identified 12 patients with definite MD and 36 with possible or probable MD. The definite cases included myotonic dystrophy type 1 (4), myotonic dystrophy type 2 (3), oculopharyngeal MD (2), Becker MD (1), distal MD (1), and facioscapulohumeral MD (1). At least five of the cases classified as definite developed symptoms after discharge from active duty. DISCUSSION: Clinicians who care for veterans should be knowledgeable about, and have access to, diagnostic testing and treatment options for MD. When conducting MD surveillance, it is important to include veterans health systems as a data source. Mild cases of MD and those of later onset appear to be compatible in some cases with successful completion of military service.


Subject(s)
Muscular Dystrophies , Veterans , Humans , Male , Middle Aged , Female , Adult , Muscular Dystrophies/diagnosis , Muscular Dystrophies/epidemiology , Muscular Dystrophies/genetics , Aged , Veterans Health , Prevalence
14.
Neuromuscul Disord ; 39: 19-23, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38691940

ABSTRACT

LAMA2-related muscular dystrophy is caused by pathogenic variants of the alpha2 subunit of Laminin. This common form of muscular dystrophy is characterized by elevated CK >1000IU/L, dystrophic changes on muscle biopsy, complete or partial absence of merosin staining, and both central and peripheral nervous system involvement. Advancements in genomic testing using NGS and wider application of RNA sequencing has expanded our knowledge of novel non-coding pathogenic variants in LAMA2. RNA sequencing is an increasingly utilized technique to directly analyze the transcriptome, through creation of a complementary DNA (cDNA) from the transcript within a tissue sample. Here we describe a homozygous deep intronic variant that produces a novel splice junction in LAMA2 identified by RNA sequencing analysis in a patient with a clinical phenotype in keeping with LAMA2-related muscular dystrophy. Furthermore, in this case merosin staining was retained suggestive of a functional deficit.


Subject(s)
Introns , Laminin , Muscular Dystrophies , Sequence Analysis, RNA , Humans , Laminin/genetics , Introns/genetics , Muscular Dystrophies/genetics , Muscular Dystrophies/pathology , Muscular Dystrophies/diagnosis , Male , Phenotype , Mutation , Female
15.
Sci Rep ; 14(1): 10774, 2024 05 11.
Article in English | MEDLINE | ID: mdl-38729999

ABSTRACT

Muscular dystrophies (MD) are a group of genetic neuromuscular disorders that cause progressive weakness and loss of muscles over time, influencing 1 in 3500-5000 children worldwide. New and exciting treatment options have led to a critical need for a clinical post-marketing surveillance tool to confirm the efficacy and safety of these treatments after individuals receive them in a commercial setting. For MDs, functional gait assessment is a common approach to evaluate the efficacy of the treatments because muscle weakness is reflected in individuals' walking patterns. However, there is little incentive for the family to continue to travel for such assessments due to the lack of access to specialty centers. While various existing sensing devices, such as cameras, force plates, and wearables can assess gait at home, they are limited by privacy concerns, area of coverage, and discomfort in carrying devices, which is not practical for long-term, continuous monitoring in daily settings. In this study, we introduce a novel functional gait assessment system using ambient floor vibrations, which is non-invasive and scalable, requiring only low-cost and sparsely deployed geophone sensors attached to the floor surface, suitable for in-home usage. Our system captures floor vibrations generated by footsteps from patients while they walk around and analyzes such vibrations to extract essential gait health information. To enhance interpretability and reliability under various sensing scenarios, we translate the signal patterns of floor vibration to pathological gait patterns related to MD, and develop a hierarchical learning algorithm that aggregates insights from individual footsteps to estimate a person's overall gait performance. When evaluated through real-world experiments with 36 subjects (including 15 patients with MD), our floor vibration sensing system achieves a 94.8% accuracy in predicting functional gait stages for patients with MD. Our approach enables accurate, accessible, and scalable functional gait assessment, bringing MD progressive tracking into real life.


Subject(s)
Gait , Muscular Dystrophies , Vibration , Humans , Child , Gait/physiology , Muscular Dystrophies/physiopathology , Muscular Dystrophies/diagnosis , Muscular Dystrophies/therapy , Male , Female , Gait Analysis/methods , Gait Analysis/instrumentation , Adolescent
16.
Brain Nerve ; 76(4): 375-386, 2024 Apr.
Article in Japanese | MEDLINE | ID: mdl-38589282

ABSTRACT

Many muscle disease names are mostly based on muscle pathology findings. Naturally, muscle pathology is important in the diagnosis of muscle diseases. Moreover, in recent years, extensive genetic analysis and autoantibody testing for myositis have been applied clinically, although muscle biopsies are less performed. However, muscle pathology should be proactively considered when a single gene presents multiple phenotypes, when variants of unknown pathological significance are detected, or in cases of autoimmune myositis that may be misdiagnosed as muscular dystrophy.


Subject(s)
Autoimmune Diseases , Muscular Diseases , Muscular Dystrophies , Myositis , Humans , Muscular Diseases/diagnosis , Muscular Diseases/genetics , Muscular Dystrophies/pathology , Muscles/pathology , Muscle, Skeletal/pathology
17.
Matrix Biol ; 129: 44-58, 2024 May.
Article in English | MEDLINE | ID: mdl-38582404

ABSTRACT

Extracellular matrix (ECM) pathologic remodeling underlies many disorders, including muscular dystrophy. Tissue decellularization removes cellular components while leaving behind ECM components. We generated "on-slide" decellularized tissue slices from genetically distinct dystrophic mouse models. The ECM of dystrophin- and sarcoglycan-deficient muscles had marked thrombospondin 4 deposition, while dysferlin-deficient muscle had excess decorin. Annexins A2 and A6 were present on all dystrophic decellularized ECMs, but annexin matrix deposition was excessive in dysferlin-deficient muscular dystrophy. Muscle-directed viral expression of annexin A6 resulted in annexin A6 in the ECM. C2C12 myoblasts seeded onto decellularized matrices displayed differential myoblast mobility and fusion. Dystrophin-deficient decellularized matrices inhibited myoblast mobility, while dysferlin-deficient decellularized matrices enhanced myoblast movement and differentiation. Myoblasts treated with recombinant annexin A6 increased mobility and fusion like that seen on dysferlin-deficient decellularized matrix and demonstrated upregulation of ECM and muscle cell differentiation genes. These findings demonstrate specific fibrotic signatures elicit effects on myoblast activity.


Subject(s)
Cell Differentiation , Cell Movement , Dysferlin , Extracellular Matrix , Myoblasts , Sarcoglycans , Animals , Myoblasts/metabolism , Myoblasts/cytology , Extracellular Matrix/metabolism , Mice , Sarcoglycans/genetics , Sarcoglycans/metabolism , Dysferlin/genetics , Dysferlin/metabolism , Muscular Dystrophies/genetics , Muscular Dystrophies/metabolism , Muscular Dystrophies/pathology , Dystrophin/genetics , Dystrophin/metabolism , Annexin A2/genetics , Annexin A2/metabolism , Decorin/genetics , Decorin/metabolism , Cell Line , Disease Models, Animal , Muscle, Skeletal/metabolism
19.
Hell J Nucl Med ; 27(1): 66-67, 2024.
Article in English | MEDLINE | ID: mdl-38629820

ABSTRACT

Technetium-99m-methoxy isobutyl isonitrile (99mTc-MIBI) myocardial perfusion imaging (MPI) is a functional imaging method with relatively poor specificity but high sensitivity. We present 48-year-old man with cardiac involvement due to muscular dystrophies (MD). Myocardial perfusion imaging rest images revealed regional myocardial perfusion decrease in multiple walls, enlarged heart and decreased left ventricular systolic function. The lesion location of MPI was consistent with that seen on CMR. Our case showed MPI was useful for detection and evaluation of the MD patient with cardiac involvement. In addition, imaging findings in combination with clinical history and other data are important. The case highlight is thevalue of MPI in myocardiopathy.


Subject(s)
Muscular Dystrophies , Myocardial Perfusion Imaging , Humans , Male , Middle Aged , Muscular Dystrophies/diagnostic imaging , Muscular Dystrophies/complications , Technetium Tc 99m Sestamibi , Radiopharmaceuticals
20.
Physiol Rep ; 12(8): e16020, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38658362

ABSTRACT

Desminopathy R350P is a human myopathy that is characterized by the progressive loss of muscle fiber organization. This results in the loss of muscle size, mobility, and strength. In desminopathy, inflammation affects muscle homeostasis and repair, and contributes to progressive muscle deterioration. Mitochondria morphology was also suggested to affect desminopathy progression. Epicatechin (Epi)-a natural compound found in cacao-has been proposed to regulate inflammatory signaling and mitochondria morphology in human and animal models. Hence, we hypothesize chronic Epi consumption to improve inflammatory pathway and mitochondria morphology in the peripheral blood mononuclear cells (PBMCs) of a desminopathy R350P patient. We found that 12 weeks of Epi consumption partially restored TRL4 signaling, indicative of inflammatory signaling and mitochondria morphology in the desminopathy patient. Moreover, Epi consumption improved blood health parameters, including reduced HOMA-IR and IL-6 levels in the desminopathy patient. This indicates that Epi consumption could be a useful tool to slow disease progression in desminopathy patients.


Subject(s)
Catechin , Leukocytes, Mononuclear , Mitochondria , Humans , Catechin/pharmacology , Catechin/administration & dosage , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/drug effects , Mitochondria/metabolism , Mitochondria/drug effects , Mitochondria/pathology , Male , Muscular Dystrophies/metabolism , Muscular Dystrophies/pathology , Muscular Dystrophies/drug therapy , Muscular Dystrophies/genetics , Adult , Female , Inflammation/metabolism , Inflammation/pathology , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cardiomyopathies/drug therapy , Desmin/metabolism , Desmin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL