Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 680
Filter
1.
Bull Exp Biol Med ; 177(5): 673-677, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39352673

ABSTRACT

The structure of left ventricular cardiomyocytes of 1 day preterm newborn rats was studied using transmission electron microscopy. It was shown that the relative area of the nucleus in cardiomyocytes of preterm rats is lower, and the relative area of the cytoplasm is higher than in full-term rats, while the relative areas of myofibrils and mitochondria do not differ. In cardiomyocytes of preterm rats damaged mitochondria, subsegmental myofibrillar contracture, and cytoplasmic swelling were found on the first postnatal day. Preterm birth in rats, in contrast to birth at term, is accompanied by the development of a number of ultrastructural damages in cardiomyocytes.


Subject(s)
Animals, Newborn , Heart Ventricles , Myocytes, Cardiac , Myofibrils , Animals , Myocytes, Cardiac/ultrastructure , Myocytes, Cardiac/pathology , Rats , Heart Ventricles/ultrastructure , Heart Ventricles/pathology , Myofibrils/ultrastructure , Myofibrils/pathology , Microscopy, Electron, Transmission , Female , Cell Nucleus/ultrastructure , Mitochondria/ultrastructure , Mitochondria/pathology , Rats, Wistar , Premature Birth/pathology
2.
Free Radic Biol Med ; 222: 493-504, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38944212

ABSTRACT

Due to an unexpected activation of different zinc (Zn) transporters in a recent prospective clinical study, we have revisited the role of Zn homeostasis and the activation of matrix metalloproteinases (MMPs) in skeletal muscle exposed to the intensive care unit (ICU) condition (immobilization and mechanical ventilation). ICU patients exposed to 12 days ICU condition were followed longitudinally with six repeated muscle biopsies while they showed a progressive preferential myosin loss, i.e., the hallmark of Critical Illness Myopathy (CIM), in parallel with the activation of Zn-transporters. In this study, we have revisited the expression of Zn-transporters and the activation of MMPs in clinical as well as in experimental studies using an established ICU model. MMPs are a group Zn-dependent endopeptidases which do not only target and cleave extracellular proteins but also intracellular proteins including multiple sarcomeric proteins. MMP-9 is of specific interest since the hallmark of CIM, the preferential myosin loss, has also been reported in dilated cardiomyopathy and coupled to MMP-9 activation. Transcriptional activation of Zn-transporters was observed in both clinical and experimental studies as well as the activation of MMPs, in particular MMP-9, in various limb and respiratory muscles in response to long-term exposure to the ICU condition. The activation of Zn-transporters was paralleled by increased Zn levels in skeletal muscle which in turn showed a negative linear correlation with the preferential myosin loss associated with CIM, offering a potential intervention strategy. Thus, activation of Zn-transporters, increased intramuscular Zn levels, and activation of the Zn-dependent MMPs are forwarded as a probable mechanism involved in CIM pathophysiology. These effects were confirmed in different rat strains subjected to a model of CIM and exacerbated by old age. This is of specific interest since old age and muscle wasting are the two factors most strongly associated with ICU mortality.


Subject(s)
Critical Illness , Muscular Diseases , Proteolysis , Zinc , Animals , Humans , Rats , Intensive Care Units , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinases/metabolism , Matrix Metalloproteinases/genetics , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Diseases/metabolism , Muscular Diseases/pathology , Muscular Diseases/genetics , Myofibrils/metabolism , Myofibrils/pathology , Zinc/metabolism
3.
Int J Rheum Dis ; 27(2): e15036, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38333999

ABSTRACT

Myofibrillar myopathies (MFMs) are a group of genetically heterogeneous diseases affecting the skeletal and cardiac muscles. Myofibrillar myopathies are characterized by focal lysis of myogenic fibers and integration of degraded myogenic fiber products into inclusion bodies, which are typically rich in desmin and many other proteins. Herein, we report a case of a 54-year-old woman who experienced bilateral thigh weakness for over three years. She was diagnosed with MFMs based on muscle biopsy findings and the presence of a novel mutation in exon 8 of the LDB3 gene. Myofibrillar myopathies caused by a mutation in the LDB3 gene are extremely uncommon and often lack distinct clinical characteristics and typically exhibit a slow disease progression. When considering a diagnosis of MFMs, particularly in complex instances of autosomal dominant myopathies where muscle biopsies do not clearly indicate MFMs, it becomes crucial for clinicians to utilize genetic test as a diagnostic tool.


Subject(s)
Myofibrils , Myopathies, Structural, Congenital , Female , Humans , Middle Aged , Myofibrils/genetics , Myofibrils/metabolism , Myofibrils/pathology , Myopathies, Structural, Congenital/diagnosis , Myopathies, Structural, Congenital/genetics , Myopathies, Structural, Congenital/metabolism , Mutation , Exons , Myocardium , Muscle, Skeletal/metabolism , Adaptor Proteins, Signal Transducing/genetics , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism
4.
J Med Genet ; 61(7): 626-632, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38413182

ABSTRACT

BACKGROUND: Congenital myopathies are a clinical, histopathological and genetic heterogeneous group of inherited muscle disorders that are defined on peculiar architectural abnormalities in the muscle fibres. Although there have been at least 33 different genetic causes of the disease, a significant percentage of congenital myopathies remain genetically unresolved. The present study aimed to report a novel TUBA4A variant in two unrelated Chinese patients with sporadic congenital myopathy. METHODS: A comprehensive strategy combining laser capture microdissection, proteomics and whole-exome sequencing was performed to identify the candidate genes. In addition, the available clinical data, myopathological changes, the findings of electrophysiological examinations and thigh muscle MRIs were also reviewed. A cellular model was established to assess the pathogenicity of the TUBA4A variant. RESULTS: We identified a recurrent novel heterozygous de novo c.679C>T (p.L227F) variant in the TUBA4A (NM_006000), encoding tubulin alpha-4A, in two unrelated patients with clinicopathologically diagnosed sporadic congenital myopathy. The prominent myopathological changes in both patients were muscle fibres with focal myofibrillar disorganisation and rimmed vacuoles. Immunofluorescence showed ubiquitin-positive TUBA4A protein aggregates in the muscle fibres with rimmed vacuoles. Overexpression of the L227F mutant TUBA4A resulted in cytoplasmic aggregates which colocalised with ubiquitin in cellular model. CONCLUSION: Our findings expanded the phenotypic and genetic manifestations of TUBA4A as well as tubulinopathies, and added a new type of congenital myopathy to be taken into consideration in the differential diagnosis.


Subject(s)
Myopathies, Structural, Congenital , Tubulin , Adult , Female , Humans , Male , Exome Sequencing , Muscle, Skeletal/pathology , Muscle, Skeletal/diagnostic imaging , Muscle, Skeletal/metabolism , Mutation , Myofibrils/pathology , Myofibrils/genetics , Myopathies, Structural, Congenital/genetics , Myopathies, Structural, Congenital/pathology , Myotonia Congenita/genetics , Myotonia Congenita/pathology , Pedigree , Tubulin/genetics
5.
Brain ; 147(8): 2867-2883, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38366623

ABSTRACT

Alterations in RNA-splicing are a molecular hallmark of several neurological diseases, including muscular dystrophies, where mutations in genes involved in RNA metabolism or characterized by alterations in RNA splicing have been described. Here, we present five patients from two unrelated families with a limb-girdle muscular dystrophy (LGMD) phenotype carrying a biallelic variant in SNUPN gene. Snurportin-1, the protein encoded by SNUPN, plays an important role in the nuclear transport of small nuclear ribonucleoproteins (snRNPs), essential components of the spliceosome. We combine deep phenotyping, including clinical features, histopathology and muscle MRI, with functional studies in patient-derived cells and muscle biopsies to demonstrate that variants in SNUPN are the cause of a new type of LGMD according to current definition. Moreover, an in vivo model in Drosophila melanogaster further supports the relevance of Snurportin-1 in muscle. SNUPN patients show a similar phenotype characterized by proximal weakness starting in childhood, restrictive respiratory dysfunction and prominent contractures, although inter-individual variability in terms of severity even in individuals from the same family was found. Muscle biopsy showed myofibrillar-like features consisting of myotilin deposits and Z-disc disorganization. MRI showed predominant impairment of paravertebral, vasti, sartorius, gracilis, peroneal and medial gastrocnemius muscles. Conservation and structural analyses of Snurportin-1 p.Ile309Ser variant suggest an effect in nuclear-cytosol snRNP trafficking. In patient-derived fibroblasts and muscle, cytoplasmic accumulation of snRNP components is observed, while total expression of Snurportin-1 and snRNPs remains unchanged, which demonstrates a functional impact of SNUPN variant in snRNP metabolism. Furthermore, RNA-splicing analysis in patients' muscle showed widespread splicing deregulation, in particular in genes relevant for muscle development and splicing factors that participate in the early steps of spliceosome assembly. In conclusion, we report that SNUPN variants are a new cause of limb girdle muscular dystrophy with specific clinical, histopathological and imaging features, supporting SNUPN as a new gene to be included in genetic testing of myopathies. These results further support the relevance of splicing-related proteins in muscle disorders.


Subject(s)
Muscular Dystrophies, Limb-Girdle , Humans , Muscular Dystrophies, Limb-Girdle/genetics , Muscular Dystrophies, Limb-Girdle/pathology , Male , Female , Adult , Animals , Muscle, Skeletal/pathology , Muscle, Skeletal/metabolism , Pedigree , Drosophila melanogaster , Myofibrils/pathology , Myofibrils/genetics , Myofibrils/metabolism , Middle Aged , Phenotype , Adolescent , Young Adult , Child
6.
Cell Calcium ; 117: 102822, 2024 01.
Article in English | MEDLINE | ID: mdl-38101154

ABSTRACT

Hypertrophic cardiomyopathy (HCM), the most common inherited heart disease, is frequently caused by mutations in the ß-cardiac myosin heavy chain gene (MYH7). Abnormal calcium handling and diastolic dysfunction are archetypical features of HCM caused by MYH7 gene mutations. However, the mechanism of how MYH7 mutations leads to these features remains unclear, which inhibits the development of effective therapies. Initially, cardiomyocytes were generated from induced pluripotent stem cells from an eight-year-old girl diagnosed with HCM carrying a MYH7(C.1063 G>A) heterozygous mutation(mutant-iPSC-CMs) and mutation-corrected isogenic iPSCs(control-iPSC-CMs) in the present study. Next, we compared phenotype of mutant-iPSC-CMs to that of control-iPSC-CMs, by assessing their morphology, hypertrophy-related genes expression, calcium handling, diastolic function and myofilament calcium sensitivity at days 15 and 40 respectively. Finally, to better understand increased myofilament Ca2+ sensitivity as a central mechanism of central pathogenicity in HCM, inhibition of calcium sensitivity with mavacamten can improveed cardiomyocyte hypertrophy. Mutant-iPSC-CMs exhibited enlarged areas, increased sarcomere disarray, enhanced expression of hypertrophy-related genes proteins, abnormal calcium handling, diastolic dysfunction and increased myofilament calcium sensitivity at day 40, but only significant increase in calcium sensitivity and mild diastolic dysfunction at day 15. Increased calcium sensitivity by levosimendan aggravates cardiomyocyte hypertrophy phenotypes such as expression of hypertrophy-related genes, abnormal calcium handling and diastolic dysfunction, while inhibition of calcium sensitivity significantly improves cardiomyocyte hypertrophy phenotypes in mutant-iPSC-CMs, suggesting increased myofilament calcium sensitivity is the primary mechanisms for MYH7 mutations pathogenesis. Our studies have uncovered a pathogenic mechanism of HCM caused by MYH7 gene mutations through which enhanced myofilament calcium sensitivity aggravates abnormal calcium handling and diastolic dysfunction. Correction of the myofilament calcium sensitivity was found to be an effective method for treating the development of HCM phenotype in vitro.


Subject(s)
Cardiomyopathies , Cardiomyopathy, Hypertrophic , Induced Pluripotent Stem Cells , Child , Female , Humans , Calcium/metabolism , Cardiac Myosins/genetics , Cardiac Myosins/metabolism , Cardiomyopathies/metabolism , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/pathology , Hypertrophy/metabolism , Hypertrophy/pathology , Induced Pluripotent Stem Cells/metabolism , Mutation/genetics , Myocytes, Cardiac/metabolism , Myofibrils/metabolism , Myofibrils/pathology , Myosin Heavy Chains/genetics , Myosin Heavy Chains/metabolism
7.
Am J Physiol Cell Physiol ; 325(5): C1326-C1335, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37781738

ABSTRACT

Muscular dystrophy exerts significant and dramatic impacts on affected patients, including progressive muscle wasting leading to lung and heart failure, and results in severely curtailed lifespan. Although the focus for many years has been on the dysfunction induced by the loss of function of dystrophin or related components of the striated muscle costamere, recent studies have demonstrated that accompanying pathologies, particularly muscle fibrosis, also contribute adversely to patient outcomes. A significant body of research has now shown that therapeutically targeting these accompanying pathologies via their underlying molecular mechanisms may provide novel approaches to patient management that can complement the current standard of care. In this review, we discuss the interplay between muscle fibrosis and muscular dystrophy pathology. A better understanding of these processes will contribute to improved patient care options, restoration of muscle function, and reduced patient morbidity and mortality.


Subject(s)
Heart Failure , Muscular Dystrophy, Duchenne , Humans , Muscular Dystrophy, Duchenne/pathology , Muscle, Skeletal/pathology , Fibrosis , Myofibrils/pathology , Heart Failure/pathology
8.
Proc Natl Acad Sci U S A ; 120(23): e2221244120, 2023 06 06.
Article in English | MEDLINE | ID: mdl-37252999

ABSTRACT

Missense variant Ile79Asn in human cardiac troponin T (cTnT-I79N) has been associated with hypertrophic cardiomyopathy and sudden cardiac arrest in juveniles. cTnT-I79N is located in the cTnT N-terminal (TnT1) loop region and is known for its pathological and prognostic relevance. A recent structural study revealed that I79 is part of a hydrophobic interface between the TnT1 loop and actin, which stabilizes the relaxed (OFF) state of the cardiac thin filament. Given the importance of understanding the role of TnT1 loop region in Ca2+ regulation of the cardiac thin filament along with the underlying mechanisms of cTnT-I79N-linked pathogenesis, we investigated the effects of cTnT-I79N on cardiac myofilament function. Transgenic I79N (Tg-I79N) muscle bundles displayed increased myofilament Ca2+ sensitivity, smaller myofilament lattice spacing, and slower crossbridge kinetics. These findings can be attributed to destabilization of the cardiac thin filament's relaxed state resulting in an increased number of crossbridges during Ca2+ activation. Additionally, in the low Ca2+-relaxed state (pCa8), we showed that more myosin heads are in the disordered-relaxed state (DRX) that are more likely to interact with actin in cTnT-I79N muscle bundles. Dysregulation of the myosin super-relaxed state (SRX) and the SRX/DRX equilibrium in cTnT-I79N muscle bundles likely result in increased mobility of myosin heads at pCa8, enhanced actomyosin interactions as evidenced by increased active force at low Ca2+, and increased sinusoidal stiffness. These findings point to a mechanism whereby cTnT-I79N weakens the interaction of the TnT1 loop with the actin filament, which in turn destabilizes the relaxed state of the cardiac thin filament.


Subject(s)
Myofibrils , Troponin T , Humans , Myofibrils/genetics , Myofibrils/pathology , Troponin T/genetics , Troponin T/chemistry , Actins/genetics , Mutation , Actin Cytoskeleton/genetics , Myosins , Calcium
9.
Mol Metab ; 66: 101643, 2022 12.
Article in English | MEDLINE | ID: mdl-36400401

ABSTRACT

OBJECTIVE: Skeletal muscle is a pivotal organ for the coordination of systemic metabolism, constituting one of the largest storage site for glucose, lipids and amino acids. Tight temporal orchestration of protein breakdown in times of fasting has to be balanced with preservation of muscle mass and function. However, the molecular mechanisms that control the fasting response in muscle are poorly understood. METHODS: We now have identified a role for the peroxisome proliferator-activated receptor γ coactivator 1ß (PGC-1ß) in the regulation of catabolic pathways in this context in muscle-specific loss-of-function mouse models. RESULTS: Muscle-specific knockouts for PGC-1ß experience mitigated muscle atrophy in fasting, linked to reduced expression of myostatin, atrogenes, activation of AMP-dependent protein kinase (AMPK) and other energy deprivation signaling pathways. At least in part, the muscle fasting response is modulated by a negative effect of PGC-1ß on the nuclear factor of activated T-cells 1 (NFATC1). CONCLUSIONS: Collectively, these data highlight the complex regulation of muscle metabolism and reveal a new role for muscle PGC-1ß in the control of proteostasis in fasting.


Subject(s)
Muscle, Skeletal , Transcription Factors , Animals , Mice , Fasting/physiology , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Atrophy/metabolism , Myofibrils/pathology , Transcription Factors/genetics , Transcription Factors/metabolism
10.
J Invest Dermatol ; 142(1): 114-123.e8, 2022 01.
Article in English | MEDLINE | ID: mdl-34274346

ABSTRACT

Dupuytren's disease (DD) is a common, progressive fibroproliferative disease affecting the palmar fascia of the hands, causing fingers to irreversibly flex toward the palm with significant loss of function. Surgical treatments are limited; therefore, effective new therapies for DD are urgently required. To identify the key cellular and molecular pathways driving DD, we employed single-cell RNA sequencing, profiling the transcriptomes of 35,250 human single cells from DD, nonpathogenic fascia, and healthy dermis. We identify a DD-specific population of pathogenic PDPN+/FAP+ mesenchymal cells displaying an elevated expression of fibrillar collagens and profibrogenic genes. In silico trajectory analysis reveals resident fibroblasts to be the source of this pathogenic population. To resolve the processes governing DD progression, genes differentially expressed during fibroblast differentiation were identified, including upregulated TNFRSF12A and transcription factor SCX. Knockdown of SCX and blockade of TNFRSF12A inhibited the proliferation and altered the profibrotic gene expression of cultured human FAP+ mesenchymal cells, demonstrating a functional role for these genes in DD. The power of single-cell RNA sequencing is utilized to identify the major pathogenic mesenchymal subpopulations driving DD and the key molecular pathways regulating the DD-specific myofibroblast phenotype. Using this precision medicine approach, inhibition of TNFRSF12A has shown potential clinical utility in the treatment of DD.


Subject(s)
Dermis/physiology , Dupuytren Contracture/genetics , Fibroblasts/physiology , Mesenchymal Stem Cells/physiology , Myofibrils/pathology , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , Endopeptidases/metabolism , Fibrosis/genetics , Gene Expression Profiling , Gene Knockdown Techniques , Humans , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Sequence Analysis, RNA , Single-Cell Analysis , TWEAK Receptor/genetics , TWEAK Receptor/metabolism
11.
J Ethnopharmacol ; 287: 114963, 2022 Apr 06.
Article in English | MEDLINE | ID: mdl-34971733

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Glabridin, extracted from Glycyrrhiza glabra L., is widely used for the treatment of hyperpigmentation because of its anti-inflammatory and antioxidant activities and its ability to inhibit melanin synthesis. This led to the strict regulation of its quality and safety. However, traditional quality control methods used for plant extracts cannot reflect the product quality owing to multiple unknown impurities, which necessitates the further analysis of impurities. AIM OF THE STUDY: The study identified the toxic impurities of glabridin and their toxicological mechanism. MATERIALS AND METHODS: In total, 10 glabridin samples from different sources were quantified using high-performance liquid chromatography. Sample toxicities were evaluated using zebrafish and cell models. To identify impurities, samples with different toxicity were analyzed by ultra-high-performance liquid chromatography coupled with quadrupole-Orbitrap mass spectrometry. The toxicity of related impurities was verified in the zebrafish model. Phalloidin stain was used to evaluate subtle changes in myofibril alignment. RESULTS: Although glabridin content in the samples was similar, there were significant differences in toxicity. The results were verified using four different mammalian cell lines. Higher contents of glabrone and glabrol were identified in the sample with the highest toxicity. In the zebrafish model, the addition of glabrol reduced the LC50 of glabridin to 9.224, 6.229, and 5.370 µM at 48, 72, and 96 h post-fertilization, respectively, whereas glabrone did not have any toxic effect. Phalloidin staining indicated that a glabrol impurity exacerbates the myotoxicity of glabridin in zebrafish embryos. CONCLUSION: Glabrol, but not glabrone, was identified as a key impurity that increased glabridin toxicity. This finding indicates that controlling glabrol content is necessary during glabridin product production.


Subject(s)
Flavonoids/toxicity , Glycyrrhiza/chemistry , Isoflavones/toxicity , Myofibrils/drug effects , Phenols/toxicity , Animals , Cell Line , Cell Line, Tumor , Chromatography, High Pressure Liquid , Embryo, Nonmammalian/drug effects , Female , Flavonoids/chemistry , Humans , Isoflavones/chemistry , Male , Mass Spectrometry , Mice , Myofibrils/pathology , Phenols/chemistry , Plant Extracts/chemistry , Plant Extracts/toxicity , Quality Control , Zebrafish
12.
Cardiovasc Res ; 118(1): 241-253, 2022 01 07.
Article in English | MEDLINE | ID: mdl-33135063

ABSTRACT

AIMS: Dilated cardiomyopathy (DCM) is associated with mutations in many genes encoding sarcomere proteins. Truncating mutations in the titin gene TTN are the most frequent. Proteomic and functional characterizations are required to elucidate the origin of the disease and the pathogenic mechanisms of TTN-truncating variants. METHODS AND RESULTS: We isolated myofibrils from DCM hearts carrying truncating TTN mutations and measured the Ca2+ sensitivity of force and its length dependence. Simultaneous measurement of force and adenosine triphosphate (ATP) consumption in skinned cardiomyocytes was also performed. Phosphorylation levels of troponin I (TnI) and myosin binding protein-C (MyBP-C) were manipulated using protein kinase A and λ phosphatase. mRNA sequencing was employed to overview gene expression profiles. We found that Ca2+ sensitivity of myofibrils carrying TTN mutations was significantly higher than in myofibrils from donor hearts. The length dependence of the Ca2+ sensitivity was absent in DCM myofibrils with TTN-truncating variants. No significant difference was found in the expression level of TTN mRNA between the DCM and donor groups. TTN exon usage and splicing were also similar. However, we identified down-regulation of genes encoding Z-disk proteins, while the atrial-specific regulatory myosin light chain gene, MYL7, was up-regulated in DCM patients with TTN-truncating variants. CONCLUSION: Titin-truncating mutations lead to decreased length-dependent activation and increased elasticity of myofibrils. Phosphorylation levels of TnI and MyBP-C seen in the left ventricles are essential for the length-dependent changes in Ca2+ sensitivity in healthy donors, but they are reduced in DCM patients with TTN-truncating variants. A decrease in expression of Z-disk proteins may explain the observed decrease in myofibril passive stiffness and length-dependent activation.


Subject(s)
Cardiomyopathy, Dilated/metabolism , Carrier Proteins/metabolism , Connectin/metabolism , Myocardial Contraction , Myocytes, Cardiac/metabolism , Myofibrils/metabolism , Troponin I/metabolism , Adult , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Dilated/physiopathology , Connectin/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Genetic Predisposition to Disease , Humans , Kinetics , Male , Middle Aged , Mutation , Myofibrils/pathology , Phenotype , Phosphoprotein Phosphatases/metabolism , Phosphorylation , Viral Proteins/metabolism , Young Adult
13.
J Am Coll Cardiol ; 78(24): 2439-2453, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34886965

ABSTRACT

BACKGROUND: Duchenne muscular dystrophy is associated with progressive deterioration in left ventricular (LV) function. The golden retriever muscular dystrophy (GRMD) dog model recapitulates the pathology and clinical manifestations of Duchenne muscular dystrophy. Importantly, they develop progressive LV dysfunction starting at early age. OBJECTIVES: The authors tested the cardioprotective effect of chronic administration of the ARM036, a small molecule that stabilizes the closed conformation of the cardiac sarcoplasmic reticulum ryanodine receptor/calcium release channel (RyR2) in young GRMD-dogs. METHODS: Two-month-old GRMD-dogs were treated with ARM036 or placebo for 4 months. Healthy-dogs of the same genetic background served as controls. Cardiac function was evaluated by conventional and 2-dimensional speckle-tracking echocardiography. Cardiac cellular and molecular analyses were performed at 6 months old. RESULTS: Conventional echocardiography showed normal LV dimensions and ejection fraction in 6-month-old GRMD dogs. Interestingly, 2-dimensional speckle-tracking echocardiography revealed decreased global longitudinal strain and the presence of hypokinetic segments in placebo-treated GRMD dogs. Single-channel measurements revealed higher RyR2 open probability at low resting Ca2+ in GRMD cardiomyocytes than in controls. ARM036 prevented those in vivo and in vitro dysfunctions in GRMD dogs. Myofilament Ca2+-sensitivity was increased in permeabilized GRMD cardiomyocytes at short sarcomere length. ARM036 had no effect on this parameter. Cross-bridge cycling kinetics were altered in GRMD myocytes and recovered with ARM036 treatment, which coincided with the level of myosin binding protein-C-S glutathionylation. CONCLUSIONS: GRMD-dogs exhibit early LV dysfunction associated with altered myofilament contractile properties. These abnormalities were prevented pharmacologically by stabilizing RyR2 with ARM036.


Subject(s)
Muscular Dystrophy, Duchenne/complications , Ryanodine Receptor Calcium Release Channel/metabolism , Ventricular Dysfunction, Left/etiology , Ventricular Function, Left/physiology , Animals , Biopsy , Disease Models, Animal , Dogs , Echocardiography , Muscular Dystrophy, Duchenne/diagnosis , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myofibrils/metabolism , Myofibrils/pathology , Sarcoplasmic Reticulum/metabolism , Sarcoplasmic Reticulum/pathology , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/pathology
14.
Int J Mol Sci ; 22(20)2021 Oct 15.
Article in English | MEDLINE | ID: mdl-34681770

ABSTRACT

Long-term exercise induces physiological cardiac adaptation, a condition referred to as athlete's heart. Exercise tolerance is known to be associated with decreased cardiac passive stiffness. Passive stiffness of the heart muscle is determined by the giant elastic protein titin. The adult cardiac muscle contains two titin isoforms: the more compliant N2BA and the stiffer N2B. Titin-based passive stiffness may be controlled by altering the expression of the different isoforms or via post-translational modifications such as phosphorylation. Currently, there is very limited knowledge about titin's role in cardiac adaptation during long-term exercise. Our aim was to determine the N2BA/N2B ratio and post-translational phosphorylation of titin in the left ventricle and to correlate the changes with the structure and transverse stiffness of cardiac sarcomeres in a rat model of an athlete's heart. The athlete's heart was induced by a 12-week-long swim-based training. In the exercised myocardium the N2BA/N2B ratio was significantly increased, Ser11878 of the PEVK domain was hypophosphorlyated, and the sarcomeric transverse elastic modulus was reduced. Thus, the reduced passive stiffness in the athlete's heart is likely caused by a shift towards the expression of the longer cardiac titin isoform and a phosphorylation-induced softening of the PEVK domain which is manifested in a mechanical rearrangement locally, within the cardiac sarcomere.


Subject(s)
Cardiomegaly, Exercise-Induced/genetics , Connectin/genetics , Myofibrils/metabolism , Adaptation, Physiological/physiology , Animals , Connectin/chemistry , Connectin/metabolism , Disease Models, Animal , Elastic Modulus/physiology , Heart/physiology , Male , Myocardial Contraction/genetics , Myocardium/metabolism , Myocardium/pathology , Myofibrils/pathology , Myofibrils/physiology , Physical Conditioning, Animal/physiology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Rats , Rats, Wistar , Sarcomeres/pathology , Sarcomeres/physiology
15.
ESC Heart Fail ; 8(6): 5178-5191, 2021 12.
Article in English | MEDLINE | ID: mdl-34486814

ABSTRACT

AIMS: The aim of the present study was to consider whether the ultrastructural features of cardiomyocytes in dilated cardiomyopathy can be used to guide genetic testing. METHODS AND RESULTS: Endomyocardial biopsy and whole-exome sequencing were performed in 32 consecutive sporadic dilated cardiomyopathy patients [51.0 (40.0-64.0) years, 75% men] in initial phases of decompensated heart failure. The predicted pathogenicity of ultrarare (minor allele frequency ≤0.0005), non-synonymous variants was determined using the American College of Medical Genetics guidelines. Focusing on 75 cardiomyopathy-susceptibility and 41 arrhythmia-susceptibility genes, we identified 404 gene variants, of which 15 were considered pathogenic or likely pathogenic in 14 patients (44% of 32). There were five sarcomeric gene variants (29% of 17 variants) found in five patients (16% of 32), involving a variant of MYBPC3 and four variants of TTN. A patient with an MYBPC3 variant showed disorganized sarcomeres, three patients with TTN variants located in the region encoding the A-band domain showed sparse sarcomeres, and a patient with a TTN variant in encoding the I-band domain showed disrupted sarcomeres. The distribution of diffuse myofilament lysis depended on the causal genes; three patients with the same TMEM43 variant had diffuse myofilament lysis near nuclei (P = 0.011), while two patients with different DSP variants had lysis in the peripheral areas of cardiomyocytes (P = 0.033). CONCLUSIONS: Derangement patterns of myofilament and subcellular distribution of myofilament lysis might implicate causal genes. Large-scale studies are required to confirm whether these ultrastructural findings are related to the causative genes.


Subject(s)
Cardiomyopathy, Dilated , Heart Failure , Myocardium , Adult , Cardiomyopathy, Dilated/diagnosis , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Carrier Proteins/genetics , Connectin/genetics , Desmoplakins/genetics , Female , Genetic Testing , Heart Failure/diagnosis , Heart Failure/genetics , Humans , Male , Middle Aged , Myocardium/ultrastructure , Myofibrils/pathology , Sarcomeres/genetics , Sarcomeres/pathology
16.
Mol Biol Cell ; 32(18): 1690-1706, 2021 08 19.
Article in English | MEDLINE | ID: mdl-34081531

ABSTRACT

Dilated cardiomyopathy (DCM), a life-threatening disease characterized by pathological heart enlargement, can be caused by myosin mutations that reduce contractile function. To better define the mechanistic basis of this disease, we employed the powerful genetic and integrative approaches available in Drosophila melanogaster. To this end, we generated and analyzed the first fly model of human myosin-induced DCM. The model reproduces the S532P human ß-cardiac myosin heavy chain DCM mutation, which is located within an actin-binding region of the motor domain. In concordance with the mutation's location at the actomyosin interface, steady-state ATPase and muscle mechanics experiments revealed that the S532P mutation reduces the rates of actin-dependent ATPase activity and actin binding and increases the rate of actin detachment. The depressed function of this myosin form reduces the number of cross-bridges during active wing beating, the power output of indirect flight muscles, and flight ability. Further, S532P mutant hearts exhibit cardiac dilation that is mutant gene dose-dependent. Our study shows that Drosophila can faithfully model various aspects of human DCM phenotypes and suggests that impaired actomyosin interactions in S532P myosin induce contractile deficits that trigger the disease.


Subject(s)
Actomyosin/metabolism , Cardiomyopathy, Dilated/genetics , Drosophila Proteins/genetics , Mutation , Myosin Heavy Chains/genetics , Actins/metabolism , Animals , Animals, Genetically Modified , Cardiac Myosins/genetics , Cardiomyopathy, Dilated/physiopathology , Disease Models, Animal , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Flight, Animal , Humans , Locomotion , Muscle, Skeletal/physiopathology , Myofibrils/pathology , Myosin Heavy Chains/metabolism
17.
Am J Physiol Cell Physiol ; 321(1): C94-C103, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33979211

ABSTRACT

Cellular senescence is the irreversible arrest of normally dividing cells and is driven by cell cycle inhibitory proteins such as p16, p21, and p53. When cells enter senescence, they secrete a host of proinflammatory factors known as the senescence-associated secretory phenotype, which has deleterious effects on surrounding cells and tissues. Little is known of the role of senescence in Duchenne muscular dystrophy (DMD), the fatal X-linked neuromuscular disorder typified by chronic inflammation, extracellular matrix remodeling, and a progressive loss in muscle mass and function. Here, we demonstrate using C57-mdx (8-wk-old) and D2-mdx (4-wk-old and 8-wk-old) mice, two mouse models of DMD, that cells displaying canonical markers of senescence are found within the skeletal muscle. Eight-week-old D2-mdx mice, which display severe muscle pathology, had greater numbers of senescent cells associated with areas of inflammation, which were mostly Cdkn1a-positive macrophages, whereas in C57-mdx muscle, senescent populations were endothelial cells and macrophages localized to newly regenerated myofibers. Interestingly, this pattern was similar to cardiotoxin (CTX)-injured wild-type (WT) muscle, which experienced a transient senescent response. Dystrophic muscle demonstrated significant upregulations in senescence pathway genes [Cdkn1a (p21), Cdkn2a (p16INK4A), and Trp53 (p53)], which correlated with the quantity of senescence-associated ß-galactosidase (SA-ß-Gal)-positive cells. These results highlight an underexplored role for cellular senescence in murine dystrophic muscle.


Subject(s)
Cellular Senescence/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Endothelial Cells/metabolism , Macrophages/metabolism , Muscle, Skeletal/metabolism , Muscular Dystrophy, Animal/genetics , Animals , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Disease Models, Animal , Dystrophin/deficiency , Dystrophin/genetics , Endothelial Cells/pathology , Gene Expression Regulation , Humans , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal/metabolism , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/pathology , Myofibrils/metabolism , Myofibrils/pathology , Signal Transduction , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , beta-Galactosidase/genetics , beta-Galactosidase/metabolism
18.
J Biol Chem ; 296: 100228, 2021.
Article in English | MEDLINE | ID: mdl-33814345

ABSTRACT

The conserved C-terminal end segment of troponin I (TnI) plays a critical role in regulating muscle relaxation. This function is retained in the isolated C-terminal 27 amino acid peptide (residues 184-210) of human cardiac TnI (HcTnI-C27): When added to skinned muscle fibers, HcTnI-C27 reduces the Ca2+-sensitivity of activated myofibrils and facilitates relaxation without decreasing the maximum force production. However, the underlying mechanism of HcTnI-C27 function is unknown. We studied the conformational preferences of HcTnI-C27 and a myopathic mutant, Arg192His, (HcTnI-C27-H). Both peptides were mainly disordered in aqueous solution with a nascent helix involving residues from Trp191 to Ile195, as shown by NMR analysis and molecular dynamics simulations. The population of nascent helix was smaller in HcTnI-C27-H than in HcTnI-C27, as shown by circular dichroism (CD) titrations. Fluorescence and isothermal titration calorimetry (ITC) showed that both peptides bound tropomyosin (αTm), with a detectably higher affinity (∼10 µM) of HcTnI-C27 than that of HcTnI-C27-H (∼15 µM), consistent with an impaired Ca2+-desensitization effect of the mutant peptide on skinned muscle strips. Upon binding to αTm, HcTnI-C27 acquired a weakly stable helix-like conformation involving residues near Trp191, as shown by transferred nuclear Overhauser effect spectroscopy and hydrogen/deuterium exchange experiments. With the potent Ca2+-desensitization effect of HcTnI-C27 on skinned cardiac muscle from a mouse model of hypertrophic cardiomyopathy, the data support that the C-terminal end domain of TnI can function as an isolated peptide with the intrinsic capacity of binding tropomyosin, providing a promising therapeutic approach to selectively improve diastolic function of the heart.


Subject(s)
Cardiomyopathy, Hypertrophic/genetics , Muscle Fibers, Skeletal/metabolism , Myofibrils/metabolism , Peptides/chemistry , Tropomyosin/metabolism , Troponin I/chemistry , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites , Calcium/metabolism , Cardiomyopathy, Hypertrophic/metabolism , Cardiomyopathy, Hypertrophic/pathology , Cardiomyopathy, Hypertrophic/prevention & control , Disease Models, Animal , Gene Expression , Humans , Kinetics , Mice , Molecular Docking Simulation , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/pathology , Muscle Relaxation , Mutation , Myofibrils/drug effects , Myofibrils/pathology , Peptides/genetics , Peptides/metabolism , Peptides/pharmacology , Protein Binding , Protein Conformation, alpha-Helical , Protein Interaction Domains and Motifs , Sequence Alignment , Sequence Homology, Amino Acid , Substrate Specificity , Tropomyosin/chemistry , Tropomyosin/genetics , Troponin I/genetics , Troponin I/metabolism
19.
Biomed Pharmacother ; 135: 111176, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33401224

ABSTRACT

We intended to examine the molecular mechanism of action of isorhamnetin (IHN) to regulate the pathway of insulin signaling. Molecular analysis, immunofluorescence, and histopathological examination were used to assess the anti-hyperglycemic and insulin resistance lowering effects of IHN in streptozotocin /high fat diet-induced type 2 diabetes using Wistar rats. At the microscopic level, treatment with IHN resulted in the restoration of myofibrils uniform arrangement and adipose tissue normal architecture. At the molecular level, treatment with IHN at three different doses showed a significant decrease in m-TOR, IGF1-R & LncRNA-RP11-773H22.4. expression and it up-regulated the expression of AKT2 mRNA, miR-1, and miR-3163 in both skeletal muscle and adipose tissue. At the protein level, IHN treated group showed a discrete spread with a moderate faint expression of m-TOR in skeletal muscles as well as adipose tissues. We concluded that IHN could be used in the in ameliorating insulin resistance associated with type 2 diabetes mellitus.


Subject(s)
Adipose Tissue/drug effects , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/pharmacology , Insulin Resistance , Insulin/blood , Myofibrils/drug effects , Quercetin/analogs & derivatives , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/pathology , Male , MicroRNAs/genetics , MicroRNAs/metabolism , Myofibrils/metabolism , Myofibrils/pathology , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Quercetin/pharmacology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Rats, Wistar , Receptor, IGF Type 1/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
20.
J Cardiovasc Pharmacol ; 77(3): 280-290, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33109927

ABSTRACT

ABSTRACT: Because cardiotoxicity is one of the leading causes of drug failure and attrition, the design of new protocols and technologies to assess proarrhythmic risks on cardiac cells is in continuous development by different laboratories. Current methodologies use electrical, intracellular Ca2+, or contractility assays to evaluate cardiotoxicity. Increasingly, the human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) are the in vitro tissue model used in commercial assays because it is believed to recapitulate many aspects of human cardiac physiology. In this work, we demonstrate that the combination of a contractility and voltage measurements, using video-based imaging and fluorescence microscopy, on hiPSC-CMs allows the investigation of mechanistic links between electrical and mechanical effects in an assay design that can address medium throughput scales necessary for drug screening, offering a view of the mechanisms underlying drug toxicity. To assess the accuracy of this novel technique, 10 commercially available inotropic drugs were tested (5 positive and 5 negative). Included were drugs with simple and specific mechanisms, such as nifedipine, Bay K8644, and blebbistatin, and others with a more complex action such as isoproterenol, pimobendan, digoxin, and amrinone, among others. In addition, the results provide a mechanism for the toxicity of itraconazole in a human model, a drug with reported side effects on the heart. The data demonstrate a strong negative inotropic effect because of the blockade of L-type Ca2+ channels and additional action on the cardiac myofilaments. We can conclude that the combination of contractility and action potential measurements can provide wider mechanistic knowledge of drug cardiotoxicity for preclinical assays.


Subject(s)
Arrhythmias, Cardiac/chemically induced , Excitation Contraction Coupling/drug effects , Fluorescent Dyes/chemistry , Induced Pluripotent Stem Cells/drug effects , Microscopy, Fluorescence , Microscopy, Video , Myocardial Contraction/drug effects , Myocytes, Cardiac/drug effects , Pyridinium Compounds/chemistry , Action Potentials/drug effects , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/physiopathology , Calcium Channels, L-Type/drug effects , Calcium Channels, L-Type/metabolism , Cardiotoxicity , Cell Differentiation , Cells, Cultured , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myofibrils/drug effects , Myofibrils/metabolism , Myofibrils/pathology , Risk Assessment , Time Factors , Toxicity Tests
SELECTION OF CITATIONS
SEARCH DETAIL