Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 165
Filtrer
1.
JCI Insight ; 9(13)2024 May 30.
Article de Anglais | MEDLINE | ID: mdl-38973610

RÉSUMÉ

Spinal and bulbar muscular atrophy (SBMA) is a slowly progressing disease with limited sensitive biomarkers that support clinical research. We analyzed plasma and serum samples from patients with SBMA and matched healthy controls in multiple cohorts, identifying 40 highly reproducible SBMA-associated proteins out of nearly 3,000 measured. These proteins were robustly enriched in gene sets of skeletal muscle expression and processes related to mitochondria and calcium signaling. Many proteins outperformed currently used clinical laboratory tests (e.g., creatine kinase [CK]) in distinguishing patients from controls and in their correlations with clinical and functional traits in patients. Two of the 40 proteins, Ectodysplasin A2 receptor (EDA2R) and Repulsive guidance molecule A (RGMA), were found to be associated with decreased survival and body weight in a mouse model of SBMA. In summary, we identified what we believe to be a robust and novel set of fluid protein biomarkers in SBMA that are linked with relevant disease features in patients and in a mouse model of disease. Changes in these SBMA-associated proteins could be used as an early predictor of treatment effects in clinical trials.


Sujet(s)
Marqueurs biologiques , Humains , Animaux , Marqueurs biologiques/sang , Marqueurs biologiques/métabolisme , Souris , Mâle , Femelle , Adulte d'âge moyen , Modèles animaux de maladie humaine , Muscles squelettiques/métabolisme , Adulte , Études cas-témoins , Sujet âgé , Protéines liées au GPI/sang , Protéines liées au GPI/génétique , Protéines liées au GPI/métabolisme
2.
J Clin Invest ; 134(12)2024 Jun 17.
Article de Anglais | MEDLINE | ID: mdl-38950322

RÉSUMÉ

Cytoplasmic and nuclear iron-sulfur (Fe-S) enzymes that are essential for genome maintenance and replication depend on the cytoplasmic Fe-S assembly (CIA) machinery for cluster acquisition. The core of the CIA machinery consists of a complex of CIAO1, MMS19 and FAM96B. The physiological consequences of loss of function in the components of the CIA pathway have thus far remained uncharacterized. Our study revealed that patients with biallelic loss of function in CIAO1 developed proximal and axial muscle weakness, fluctuating creatine kinase elevation, and respiratory insufficiency. In addition, they presented with CNS symptoms including learning difficulties and neurobehavioral comorbidities, along with iron deposition in deep brain nuclei, mild normocytic to macrocytic anemia, and gastrointestinal symptoms. Mutational analysis revealed reduced stability of the variants compared with WT CIAO1. Functional assays demonstrated failure of the variants identified in patients to recruit Fe-S recipient proteins, resulting in compromised activities of DNA helicases, polymerases, and repair enzymes that rely on the CIA complex to acquire their Fe-S cofactors. Lentivirus-mediated restoration of CIAO1 expression reversed all patient-derived cellular abnormalities. Our study identifies CIAO1 as a human disease gene and provides insights into the broader implications of the cytosolic Fe-S assembly pathway in human health and disease.


Sujet(s)
Ferrosulfoprotéines , Humains , Ferrosulfoprotéines/génétique , Ferrosulfoprotéines/métabolisme , Mâle , Femelle , Maladies neuromusculaires/génétique , Maladies neuromusculaires/enzymologie , Maladies neuromusculaires/métabolisme , Maladies neuromusculaires/anatomopathologie , Enfant , Noyau de la cellule/métabolisme , Noyau de la cellule/enzymologie , Noyau de la cellule/génétique , Cytoplasme/métabolisme , Cytoplasme/enzymologie , Métallochaperons
3.
JCI Insight ; 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38954467

RÉSUMÉ

Pulmonary disorders impact 40% to 80% of individuals with obesity. Respiratory muscle dysfunction is linked to these conditions; however, its pathophysiology remains largely undefined. Mice subjected to diet-induced obesity (DIO) develop diaphragmatic weakness. Increased intra-diaphragmatic adiposity and extracellular matrix (ECM) content correlate with reductions in contractile force. Thrombospondin-1 (THBS1) is an obesity-associated matricellular protein linked with muscular damage in genetic myopathies. THBS1 induces proliferation of fibro-adipogenic progenitors (FAPs) - mesenchymal cells that differentiate into adipocytes and fibroblasts. We hypothesized that THBS1 drives FAP-mediated diaphragm remodeling and contractile dysfunction in DIO. We tested this by comparing the effects of dietary challenge on diaphragms of wild-type (WT) and Thbs1 knockout (Thbs1-/-) mice. Bulk and single-cell transcriptomics demonstrated DIO-induced stromal expansion in WT diaphragms. Diaphragm FAPs displayed upregulation of ECM and TGF ß-related expression signatures and augmentation of a Thy1-expressing sub-population previously linked to type 2 diabetes. Despite similar weight gain, Thbs1-/- mice were protected from these transcriptomic changes and from obesity-induced increases in diaphragm adiposity and ECM deposition. Unlike WT controls, Thbs1-/- diaphragms maintained normal contractile force and motion after DIO challenge. These findings establish THBS1 as a necessary mediator of diaphragm stromal remodeling and contractile dysfunction in overnutrition and a potential therapeutic target in obesity-associated respiratory dysfunction.

4.
J Clin Invest ; 134(11)2024 May 03.
Article de Anglais | MEDLINE | ID: mdl-38702076

RÉSUMÉ

Sarcopenia burdens the older population through loss of muscle energy and mass, yet treatments to functionally rescue both parameters are lacking. The glucocorticoid prednisone remodels muscle metabolism on the basis of frequency of intake, but its mechanisms in sarcopenia are unknown. We found that once-weekly intermittent prednisone administration rescued muscle quality in aged 24-month-old mice to a level comparable to that seen in young 4-month-old mice. We discovered an age- and sex-independent glucocorticoid receptor transactivation program in muscle encompassing peroxisome proliferator-activated receptor γ coactivator 1 α (PGC1α) and its cofactor Lipin1. Treatment coordinately improved mitochondrial abundance through isoform 1 and muscle mass through isoform 4 of the myocyte-specific PGC1α, which was required for the treatment-driven increase in carbon shuttling from glucose oxidation to amino acid biogenesis. We also probed myocyte-specific Lipin1 as a nonredundant factor coaxing PGC1α upregulation to the stimulation of both oxidative and anabolic effects. Our study unveils an aging-resistant druggable program in myocytes for the coordinated rescue of energy and mass in sarcopenia.


Sujet(s)
Vieillissement , Glucocorticoïdes , Muscles squelettiques , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes , Phosphatidate phosphatase , Sarcopénie , Animaux , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/métabolisme , Coactivateur 1-alpha du récepteur gamma activé par les proliférateurs de peroxysomes/génétique , Sarcopénie/métabolisme , Sarcopénie/traitement médicamenteux , Sarcopénie/anatomopathologie , Sarcopénie/génétique , Souris , Vieillissement/métabolisme , Phosphatidate phosphatase/génétique , Phosphatidate phosphatase/métabolisme , Glucocorticoïdes/pharmacologie , Muscles squelettiques/métabolisme , Muscles squelettiques/effets des médicaments et des substances chimiques , Mâle , Modèles animaux de maladie humaine , Femelle
5.
J Clin Invest ; 2024 May 07.
Article de Anglais | MEDLINE | ID: mdl-38713532

RÉSUMÉ

Satellite cells, the stem cells of skeletal muscle tissue, hold a remarkable regeneration capacity and therapeutic potential in regenerative medicine. However, low satellite cell yield from autologous or donor-derived muscles hinders the adoption of satellite cell transplantation for the treatment of muscle diseases, including Duchenne muscular dystrophy (DMD). To address this limitation, here we investigated whether satellite cells can be derived in allogeneic or xenogeneic animal hosts. First, injection of CRISPR/Cas9-corrected mouse DMD-induced pluripotent stem cells (iPSCs) into mouse blastocysts carrying an ablation system of host satellite cells gave rise to intraspecies chimeras exclusively carrying iPSC-derived satellite cells. Furthermore, injection of genetically corrected DMD-iPSCs into rat blastocysts resulted in the formation of interspecies rat-mouse chimeras harboring mouse satellite cells. Remarkably, iPSC-derived satellite cells or derivative myoblasts produced in intraspecies or interspecies chimeras restored dystrophin expression in DMD mice following intramuscular transplantation, and contributed to the satellite cell pool. Collectively, this study demonstrates the feasibility of producing therapeutically competent stem cells across divergent animal species, raising the possibility of generating human muscle stem cells in large animals for regenerative medicine purposes.

6.
JCI Insight ; 9(11)2024 May 07.
Article de Anglais | MEDLINE | ID: mdl-38713520

RÉSUMÉ

Clinical trials delivering high doses of adeno-associated viruses (AAVs) expressing truncated dystrophin molecules (microdystrophins) are underway for Duchenne muscular dystrophy (DMD). We examined the efficiency and efficacy of this strategy with 4 microdystrophin constructs (3 in clinical trials and a variant of the largest clinical construct), in a severe mouse model of DMD, using AAV doses comparable with those in clinical trials. We achieved high levels of microdystrophin expression in striated muscles with cardiac expression approximately 10-fold higher than that observed in skeletal muscle. Significant, albeit incomplete, correction of skeletal muscle disease was observed. Surprisingly, a lethal acceleration of cardiac disease occurred with 2 of the microdystrophins. The detrimental cardiac effect appears to be caused by variable competition (dependent on microdystrophin design and expression level) between microdystrophin and utrophin at the cardiomyocyte membrane. There may also be a contribution from an overloading of protein degradation. The significance of these observations for patients currently being treated with AAV-microdystrophin therapies is unclear since the levels of expression being achieved in the DMD hearts are unknown. However, these findings suggest that microdystrophin treatments need to avoid excessively high levels of expression in the heart and that cardiac function should be carefully monitored in these patients.


Sujet(s)
Thérapie génétique , Myopathie de Duchenne , Animaux , Humains , Mâle , Souris , Dependovirus/génétique , Modèles animaux de maladie humaine , Dystrophine/génétique , Thérapie génétique/méthodes , Vecteurs génétiques/administration et posologie , Vecteurs génétiques/génétique , Souris de lignée mdx , Muscles squelettiques/métabolisme , Myopathie de Duchenne/thérapie , Myopathie de Duchenne/génétique , Myopathie de Duchenne/métabolisme , Myocytes cardiaques/métabolisme , Utrophine/génétique , Utrophine/métabolisme
7.
J Clin Invest ; 134(9)2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38690726

RÉSUMÉ

Proline substitutions within the coiled-coil rod region of the ß-myosin gene (MYH7) are the predominant mutations causing Laing distal myopathy (MPD1), an autosomal dominant disorder characterized by progressive weakness of distal/proximal muscles. We report that the MDP1 mutation R1500P, studied in what we believe to be the first mouse model for the disease, adversely affected myosin motor activity despite being in the structural rod domain that directs thick filament assembly. Contractility experiments carried out on isolated mutant muscles, myofibrils, and myofibers identified muscle fatigue and weakness phenotypes, an increased rate of actin-myosin detachment, and a conformational shift of the myosin heads toward the more reactive disordered relaxed (DRX) state, causing hypercontractility and greater ATP consumption. Similarly, molecular analysis of muscle biopsies from patients with MPD1 revealed a significant increase in sarcomeric DRX content, as observed in a subset of myosin motor domain mutations causing hypertrophic cardiomyopathy. Finally, oral administration of MYK-581, a small molecule that decreases the population of heads in the DRX configuration, significantly improved the limited running capacity of the R1500P-transgenic mice and corrected the increased DRX state of the myofibrils from patients. These studies provide evidence of the molecular pathogenesis of proline rod mutations and lay the groundwork for the therapeutic advancement of myosin modulators.


Sujet(s)
Substitution d'acide aminé , Myopathies distales , Proline , Animaux , Souris , Humains , Proline/génétique , Proline/métabolisme , Myopathies distales/génétique , Myopathies distales/métabolisme , Myopathies distales/anatomopathologie , Mutation faux-sens , Myosines cardiaques/génétique , Myosines cardiaques/métabolisme , Chaînes lourdes de myosine/génétique , Chaînes lourdes de myosine/métabolisme , Chaînes lourdes de myosine/composition chimique , Femelle , Mâle , Souris transgéniques , Contraction musculaire/génétique , Muscles squelettiques/métabolisme , Muscles squelettiques/anatomopathologie
8.
JCI Insight ; 9(10)2024 Apr 23.
Article de Anglais | MEDLINE | ID: mdl-38652558

RÉSUMÉ

Chronic kidney disease (CKD) causes accumulation of uremic metabolites that negatively affect skeletal muscle. Tryptophan-derived uremic metabolites are agonists of the aryl hydrocarbon receptor (AHR), which has been shown to be activated in CKD. This study investigated the role of the AHR in skeletal muscle pathology of CKD. Compared with controls with normal kidney function, AHR-dependent gene expression (CYP1A1 and CYP1B1) was significantly upregulated in skeletal muscle of patients with CKD, and the magnitude of AHR activation was inversely correlated with mitochondrial respiration. In mice with CKD, muscle mitochondrial oxidative phosphorylation (OXPHOS) was markedly impaired and strongly correlated with the serum level of tryptophan-derived uremic metabolites and AHR activation. Muscle-specific deletion of the AHR substantially improved mitochondrial OXPHOS in male mice with the greatest uremic toxicity (CKD + probenecid) and abolished the relationship between uremic metabolites and OXPHOS. The uremic metabolite/AHR/mitochondrial axis in skeletal muscle was verified using muscle-specific AHR knockdown in C57BL/6J mice harboring a high-affinity AHR allele, as well as ectopic viral expression of constitutively active mutant AHR in mice with normal renal function. Notably, OXPHOS changes in AHRmKO mice were present only when mitochondria were fueled by carbohydrates. Further analyses revealed that AHR activation in mice led to significantly increased pyruvate dehydrogenase kinase 4 (Pdk4) expression and phosphorylation of pyruvate dehydrogenase enzyme. These findings establish a uremic metabolite/AHR/Pdk4 axis in skeletal muscle that governs mitochondrial deficits in carbohydrate oxidation during CKD.


Sujet(s)
Souris de lignée C57BL , Muscles squelettiques , Phosphorylation oxydative , Pyruvate dehydrogenase acetyl-transferring kinase , Récepteurs à hydrocarbure aromatique , Insuffisance rénale chronique , Tryptophane , Animaux , Récepteurs à hydrocarbure aromatique/métabolisme , Récepteurs à hydrocarbure aromatique/génétique , Souris , Mâle , Insuffisance rénale chronique/métabolisme , Tryptophane/métabolisme , Muscles squelettiques/métabolisme , Humains , Pyruvate dehydrogenase acetyl-transferring kinase/métabolisme , Pyruvate dehydrogenase acetyl-transferring kinase/génétique , Urémie/métabolisme , Mitochondries du muscle/métabolisme , Facteurs de transcription à motif basique hélice-boucle-hélice/métabolisme , Facteurs de transcription à motif basique hélice-boucle-hélice/génétique , Femelle , Souris knockout , Cytochrome P-450 CYP1B1/métabolisme , Cytochrome P-450 CYP1B1/génétique , Cytochrome P-450 CYP1A1/métabolisme , Cytochrome P-450 CYP1A1/génétique , Adulte d'âge moyen , Métabolisme énergétique , Modèles animaux de maladie humaine
9.
JCI Insight ; 9(6)2024 Mar 05.
Article de Anglais | MEDLINE | ID: mdl-38516893

RÉSUMÉ

Tubular aggregate myopathy (TAM) and Stormorken syndrome (STRMK) are clinically overlapping disorders characterized by childhood-onset muscle weakness and a variable occurrence of multisystemic signs, including short stature, thrombocytopenia, and hyposplenism. TAM/STRMK is caused by gain-of-function mutations in the Ca2+ sensor STIM1 or the Ca2+ channel ORAI1, both of which regulate Ca2+ homeostasis through the ubiquitous store-operated Ca2+ entry (SOCE) mechanism. Functional experiments in cells have demonstrated that the TAM/STRMK mutations induce SOCE overactivation, resulting in excessive influx of extracellular Ca2+. There is currently no treatment for TAM/STRMK, but SOCE is amenable to manipulation. Here, we crossed Stim1R304W/+ mice harboring the most common TAM/STRMK mutation with Orai1R93W/+ mice carrying an ORAI1 mutation partially obstructing Ca2+ influx. Compared with Stim1R304W/+ littermates, Stim1R304W/+Orai1R93W/+ offspring showed a normalization of bone architecture, spleen histology, and muscle morphology; an increase of thrombocytes; and improved muscle contraction and relaxation kinetics. Accordingly, comparative RNA-Seq detected more than 1,200 dysregulated genes in Stim1R304W/+ muscle and revealed a major restoration of gene expression in Stim1R304W/+Orai1R93W/+ mice. Altogether, we provide physiological, morphological, functional, and molecular data highlighting the therapeutic potential of ORAI1 inhibition to rescue the multisystemic TAM/STRMK signs, and we identified myostatin as a promising biomarker for TAM/STRMK in humans and mice.


Sujet(s)
Anomalies des plaquettes , Dyslexie , Ichtyose , Migraines , Myopathies congénitales structurales , Protéine ORAI1 , Rate , Animaux , Souris , Calcium/métabolisme , Érythrocytes anormaux , Migraines/traitement médicamenteux , Myosis/traitement médicamenteux , Myosis/génétique , Myosis/métabolisme , Fatigue musculaire , Myopathies congénitales structurales/traitement médicamenteux , Myopathies congénitales structurales/génétique , Myopathies congénitales structurales/métabolisme , Protéine ORAI1/génétique , Protéine ORAI1/métabolisme , Rate/métabolisme , Rate/malformations
10.
JCI Insight ; 9(8)2024 Mar 14.
Article de Anglais | MEDLINE | ID: mdl-38483507

RÉSUMÉ

The polymerization of myosin molecules into thick filaments in muscle sarcomeres is essential for cardiac contractility, with the attenuation of interactions between the heads of myosin molecules within the filaments being proposed to result in hypercontractility, as observed in hypertrophic cardiomyopathy (HCM). However, experimental evidence demonstrates that the structure of these giant macromolecular complexes is highly dynamic, with molecules exchanging between the filaments and a pool of soluble molecules on the minute timescale. Therefore, we sought to test the hypothesis that the enhancement of interactions between the heads of myosin molecules within thick filaments limits the mobility of myosin by taking advantage of mavacamten, a small molecule approved for the treatment of HCM. Myosin molecules were labeled in vivo with a green fluorescent protein (GFP) and imaged in intact hearts using multiphoton microscopy. Treatment of the intact hearts with mavacamten resulted in an unexpected > 5-fold enhancement in GFP-myosin mobility within the sarcomere. In vitro biochemical assays suggested that mavacamten enhanced the mobility of GFP-myosin by increasing the solubility of myosin molecules, through the stabilization of a compact/folded conformation of the molecules, once disassociated from the thick filaments. These findings provide alternative insight into the mechanisms by which molecules exchange into and out of thick filaments and have implications for how mavacamten may affect cardiac contractility.


Sujet(s)
Benzylamines , Myocarde , Sarcomères , Solubilité , Uracile/analogues et dérivés , Animaux , Sarcomères/métabolisme , Myocarde/métabolisme , Souris , Myosines/métabolisme , Pliage des protéines , Protéines à fluorescence verte/métabolisme , Protéines à fluorescence verte/génétique , Cardiomyopathie hypertrophique/métabolisme , Contraction myocardique , Humains , Mâle
11.
JCI Insight ; 9(8)2024 Mar 26.
Article de Anglais | MEDLINE | ID: mdl-38530354

RÉSUMÉ

Skeletal muscle wasting results from numerous pathological conditions affecting both the musculoskeletal and nervous systems. A unifying feature of these pathologies is the upregulation of members of the E3 ubiquitin ligase family, resulting in increased proteolytic degradation of target proteins. Despite the critical role of E3 ubiquitin ligases in regulating muscle mass, the specific proteins they target for degradation and the mechanisms by which they regulate skeletal muscle homeostasis remain ill-defined. Here, using zebrafish loss-of-function models combined with in vivo cell biology and proteomic approaches, we reveal a role of atrogin-1 in regulating the levels of the endoplasmic reticulum chaperone BiP. Loss of atrogin-1 resulted in an accumulation of BiP, leading to impaired mitochondrial dynamics and a subsequent loss in muscle fiber integrity. We further implicated a disruption in atrogin-1-mediated BiP regulation in the pathogenesis of Duchenne muscular dystrophy. We revealed that BiP was not only upregulated in Duchenne muscular dystrophy, but its inhibition using pharmacological strategies, or by upregulating atrogin-1, significantly ameliorated pathology in a zebrafish model of Duchenne muscular dystrophy. Collectively, our data implicate atrogin-1 and BiP in the pathogenesis of Duchenne muscular dystrophy and highlight atrogin-1's essential role in maintaining muscle homeostasis.


Sujet(s)
Modèles animaux de maladie humaine , Chaperonne BiP du réticulum endoplasmique , Homéostasie , Protéines du muscle , Muscles squelettiques , Myopathie de Duchenne , SKP cullin F-box protein ligases , Danio zébré , Animaux , SKP cullin F-box protein ligases/métabolisme , SKP cullin F-box protein ligases/génétique , Protéines du muscle/métabolisme , Protéines du muscle/génétique , Muscles squelettiques/métabolisme , Muscles squelettiques/anatomopathologie , Myopathie de Duchenne/métabolisme , Myopathie de Duchenne/anatomopathologie , Myopathie de Duchenne/génétique , Humains , Chaperonne BiP du réticulum endoplasmique/métabolisme , Protéines du choc thermique/métabolisme , Protéines du choc thermique/génétique , Protéines de poisson-zèbre/métabolisme , Protéines de poisson-zèbre/génétique , Réticulum endoplasmique/métabolisme , Dynamique mitochondriale
12.
J Clin Invest ; 134(7)2024 Feb 01.
Article de Anglais | MEDLINE | ID: mdl-38300705

RÉSUMÉ

Stromal interaction molecule 1 (STIM1) is a Ca2+ sensor located in the sarcoplasmic reticulum (SR) of skeletal muscle, where it is best known for its role in store-operated Ca2+ entry (SOCE). Genetic syndromes resulting from STIM1 mutations are recognized as a cause of muscle weakness and atrophy. Here, we focused on a gain-of-function mutation that occurs in humans and mice (STIM1+/D84G mice), in which muscles exhibited constitutive SOCE. Unexpectedly, this constitutive SOCE did not affect global Ca2+ transients, SR Ca2+ content, or excitation-contraction coupling (ECC) and was therefore unlikely to underlie the reduced muscle mass and weakness observed in these mice. Instead, we demonstrate that the presence of D84G STIM1 in the nuclear envelope of STIM1+/D84G muscle disrupted nuclear-cytosolic coupling, causing severe derangement in nuclear architecture, DNA damage, and altered lamina A-associated gene expression. Functionally, we found that D84G STIM1 reduced the transfer of Ca2+ from the cytosol to the nucleus in myoblasts, resulting in a reduction of [Ca2+]N. Taken together, we propose a novel role for STIM1 in the nuclear envelope that links Ca2+ signaling to nuclear stability in skeletal muscle.


Sujet(s)
Faiblesse musculaire , Enveloppe nucléaire , Molécule-1 d'interaction stromale , Animaux , Humains , Souris , Calcium/métabolisme , Signalisation calcique , Faiblesse musculaire/génétique , Faiblesse musculaire/métabolisme , Muscles squelettiques/métabolisme , Mutation , Protéines tumorales/génétique , Protéines tumorales/métabolisme , Enveloppe nucléaire/génétique , Enveloppe nucléaire/métabolisme , Protéine ORAI1/génétique , Molécule-1 d'interaction stromale/génétique , Molécule-1 d'interaction stromale/métabolisme
13.
JCI Insight ; 9(4)2024 Feb 22.
Article de Anglais | MEDLINE | ID: mdl-38385748

RÉSUMÉ

BACKGROUNDWhile the benefits of statin therapy on atherosclerotic cardiovascular disease are clear, patients often experience mild to moderate skeletal myopathic symptoms, the mechanism for which is unknown. This study investigated the potential effect of high-dose atorvastatin therapy on skeletal muscle mitochondrial function and whole-body aerobic capacity in humans.METHODSEight overweight (BMI, 31.9 ± 2.0) but otherwise healthy sedentary adults (4 females, 4 males) were studied before (day 0) and 14, 28, and 56 days after initiating atorvastatin (80 mg/d) therapy.RESULTSMaximal ADP-stimulated respiration, measured in permeabilized fiber bundles from muscle biopsies taken at each time point, declined gradually over the course of atorvastatin treatment, resulting in > 30% loss of skeletal muscle mitochondrial oxidative phosphorylation capacity by day 56. Indices of in vivo muscle oxidative capacity (via near-infrared spectroscopy) decreased by 23% to 45%. In whole muscle homogenates from day 0 biopsies, atorvastatin inhibited complex III activity at midmicromolar concentrations, whereas complex IV activity was inhibited at low nanomolar concentrations.CONCLUSIONThese findings demonstrate that high-dose atorvastatin treatment elicits a striking progressive decline in skeletal muscle mitochondrial respiratory capacity, highlighting the need for longer-term dose-response studies in different patient populations to thoroughly define the effect of statin therapy on skeletal muscle health.FUNDINGNIH R01 AR071263.


Sujet(s)
Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase , Maladies musculaires , Mâle , Adulte , Femelle , Humains , Atorvastatine/pharmacologie , Atorvastatine/métabolisme , Inhibiteurs de l'hydroxyméthylglutaryl-CoA réductase/pharmacologie , Mitochondries du muscle/métabolisme , Muscles squelettiques/métabolisme , Mitochondries , Maladies musculaires/métabolisme
14.
Article de Anglais | MEDLINE | ID: mdl-38317053

RÉSUMÉ

OBJECTIVE: To better understand the pathogenesis of juvenile dermatomyositis (JDM), we examined the effect of the cytokines type I interferons (IFN I) and JAK inhibitor drugs (JAKi) on gene expression in bioengineered pediatric skeletal muscle. METHODS: Myoblasts from 3 healthy pediatric donors were used to create three-dimensional skeletal muscle units termed myobundles. Myobundles were treated with IFN I, either IFNα or IFNß. A subset of IFNß-exposed myobundles was treated with JAKi tofacitinib or baricitinib. RNA sequencing analysis was performed on all myobundles. RESULTS: Seventy-six myobundles were analyzed. Principal component analysis showed donor-specific clusters of gene expression across IFNα and IFNß-exposed myobundles in a dose-dependent manner. Both cytokines upregulated interferon response and proinflammatory genes; however, IFNß led to more significant upregulation. Key downregulated pathways involved oxidative phosphorylation, fatty acid metabolism and myogenesis genes. Addition of tofacitinib or baricitinib moderated the gene expression induced by IFNß, with partial reversal of upregulated inflammatory and downregulated myogenesis pathways. Baricitinib altered genetic profiles more than tofacitinib. CONCLUSION: IFNß leads to more pro-inflammatory gene upregulation than IFNα, correlating to greater decrease in contractile protein gene expression and reduced contractile force. JAK inhibitors, baricitinib more so than tofacitinib, partially reverse IFN I-induced genetic changes. Increased IFN I exposure in healthy bioengineered skeletal muscle leads to IFN-inducible gene expression, inflammatory pathway enrichment, and myogenesis gene downregulation, consistent with what is observed in JDM.

15.
JCI Insight ; 9(3)2024 Jan 04.
Article de Anglais | MEDLINE | ID: mdl-38175727

RÉSUMÉ

The Murphy Roths Large (MRL) mouse strain has "super-healing" properties that enhance recovery from injury. In mice, the DBA/2J strain intensifies many aspects of muscular dystrophy, so we evaluated the ability of the MRL strain to suppress muscular dystrophy in the Sgcg-null mouse model of limb girdle muscular dystrophy. A comparative analysis of Sgcg-null mice in the DBA/2J versus MRL strains showed greater myofiber regeneration, with reduced structural degradation of muscle in the MRL strain. Transcriptomic profiling of dystrophic muscle indicated strain-dependent expression of extracellular matrix (ECM) and TGF-ß signaling genes. To investigate the MRL ECM, cellular components were removed from dystrophic muscle sections to generate decellularized myoscaffolds. Decellularized myoscaffolds from dystrophic mice in the protective MRL strain had significantly less deposition of collagen and matrix-bound TGF-ß1 and TGF-ß3 throughout the matrix. Dystrophic myoscaffolds from the MRL background, but not the DBA/2J background, were enriched in myokines like IGF-1 and IL-6. C2C12 myoblasts seeded onto decellularized matrices from Sgcg-/- MRL and Sgcg-/- DBA/2J muscles showed the MRL background induced greater myoblast differentiation compared with dystrophic DBA/2J myoscaffolds. Thus, the MRL background imparts its effect through a highly regenerative ECM, which is active even in muscular dystrophy.


Sujet(s)
Dystrophies musculaires des ceintures , Dystrophies musculaires , Souris , Animaux , Souris de lignée DBA , Dystrophies musculaires/génétique , Muscles , Matrice extracellulaire , Souris knockout
16.
J Clin Invest ; 134(1)2024 Jan 02.
Article de Anglais | MEDLINE | ID: mdl-38165038

RÉSUMÉ

Myotonic dystrophy type 1 (DM1) involves misregulated alternative splicing for specific genes. We used exon or nucleotide deletion to mimic altered splicing of genes central to muscle excitation-contraction coupling in mice. Mice with forced skipping of exon 29 in the CaV1.1 calcium channel combined with loss of ClC-1 chloride channel function displayed markedly reduced lifespan, whereas other combinations of splicing mimics did not affect survival. The Ca2+/Cl- bi-channelopathy mice exhibited myotonia, weakness, and impairment of mobility and respiration. Chronic administration of the calcium channel blocker verapamil rescued survival and improved force generation, myotonia, and respiratory function. These results suggest that Ca2+/Cl- bi-channelopathy contributes to muscle impairment in DM1 and is potentially mitigated by common clinically available calcium channel blockers.


Sujet(s)
Canalopathies , Myotonie , Dystrophie myotonique , Souris , Animaux , Dystrophie myotonique/traitement médicamenteux , Dystrophie myotonique/génétique , Dystrophie myotonique/métabolisme , Calcium/métabolisme , Chlorures/métabolisme , Myotonie/métabolisme , Vérapamil/pharmacologie , Vérapamil/métabolisme , Canalopathies/génétique , Canalopathies/métabolisme , Épissage alternatif , Canaux chlorure/génétique , Canaux chlorure/métabolisme , Muscles squelettiques/métabolisme
17.
Rheumatology (Oxford) ; 63(1): 209-217, 2024 Jan 04.
Article de Anglais | MEDLINE | ID: mdl-37094222

RÉSUMÉ

OBJECTIVE: To investigate pathogenic mechanisms underlying JDM, we defined the effect of type I IFN, IFN-α and IFN-ß, on pediatric skeletal muscle function and expression of myositis-related proteins using an in vitro engineered human skeletal muscle model (myobundle). METHODS: Primary myoblasts were isolated from three healthy pediatric donors and used to create myobundles that mimic functioning skeletal muscle in structural architecture and physiologic function. Myobundles were exposed to 0, 5, 10 or 20 ng/ml IFN-α or IFN-ß for 7 days and then functionally tested under electrical stimulation and analyzed immunohistochemically for structural and myositis-related proteins. Additionally, IFN-ß-exposed myobundles were treated with Janus kinase inhibitors (JAKis) tofacitinib and baricitinib. These myobundles were also analyzed for contractile force and immunohistochemistry. RESULTS: IFN-ß, but not IFN-α, was associated with decreased contractile tetanus force and slowed twitch kinetics. These effects were reversed by tofacitinib and baricitinib. Type I IFN paradoxically reduced myobundle fatigue, which did not reverse after JAKi. Additionally, type I IFN correlated with MHC I upregulation, which normalized after JAKi treatment, but expression of myositis-specific autoantigens Mi-2, melanocyte differentiation-associated protein 5 and the endoplasmic reticulum stress marker GRP78 were variable and donor specific after type I IFN exposure. CONCLUSION: IFN-α and IFN-ß have distinct effects on pediatric skeletal muscle and these effects can partially be reversed by JAKi treatment. This is the first study illustrating effective use of a three-dimensional human skeletal muscle model to investigate JDM pathogenesis and test novel therapeutics.


Sujet(s)
Dermatomyosite , Interféron de type I , Maladies musculaires , Myosite , Humains , Enfant , Dermatomyosite/anatomopathologie , Muscles squelettiques/anatomopathologie , Myosite/anatomopathologie , Maladies musculaires/anatomopathologie
18.
JCI Insight ; 9(2)2024 Jan 23.
Article de Anglais | MEDLINE | ID: mdl-38051584

RÉSUMÉ

Cachexia is a debilitating skeletal muscle wasting condition for which we currently lack effective treatments. In the context of cancer, certain chemotherapeutics cause DNA damage and cellular senescence. Senescent cells exhibit chronic activation of the transcription factor NF-κB, a known mediator of the proinflammatory senescence-associated secretory phenotype (SASP) and skeletal muscle atrophy. Thus, targeting NF-κB represents a logical therapeutic strategy to alleviate unintended consequences of genotoxic drugs. Herein, we show that treatment with the IKK/NF-κB inhibitor SR12343 during a course of chemotherapy reduces markers of cellular senescence and the SASP in liver, skeletal muscle, and circulation and, correspondingly, attenuates features of skeletal muscle pathology. Lastly, we demonstrate that SR12343 mitigates chemotherapy-induced reductions in body weight, lean mass, fat mass, and muscle strength. These findings support senescent cells as a promising druggable target to counteract the SASP and skeletal muscle wasting in the context of chemotherapy.


Sujet(s)
Antinéoplasiques , Facteur de transcription NF-kappa B , Humains , Facteur de transcription NF-kappa B/métabolisme , Transduction du signal , Cachexie/induit chimiquement , Cachexie/traitement médicamenteux , Sénothérapie , Amyotrophie/induit chimiquement , Amyotrophie/traitement médicamenteux , Antinéoplasiques/effets indésirables
19.
J Clin Invest ; 134(2)2024 Jan 16.
Article de Anglais | MEDLINE | ID: mdl-37962957

RÉSUMÉ

Heterozygous (HET) truncating variant mutations in the TTN gene (TTNtvs), encoding the giant titin protein, are the most common genetic cause of dilated cardiomyopathy (DCM). However, the molecular mechanisms by which TTNtv mutations induce DCM are controversial. Here, we studied 127 clinically identified DCM human cardiac samples with next-generation sequencing (NGS), high-resolution gel electrophoresis, Western blot analysis, and super-resolution microscopy in order to dissect the structural and functional consequences of TTNtv mutations. The occurrence of TTNtv was found to be 15% in the DCM cohort. Truncated titin proteins matching, by molecular weight, the gene sequence predictions were detected in the majority of the TTNtv+ samples. Full-length titin was reduced in TTNtv+ compared with TTNtv- samples. Proteomics analysis of washed myofibrils and stimulated emission depletion (STED) super-resolution microscopy of myocardial sarcomeres labeled with sequence-specific anti-titin antibodies revealed that truncated titin was structurally integrated into the sarcomere. Sarcomere length-dependent anti-titin epitope position, shape, and intensity analyses pointed at possible structural defects in the I/A junction and the M-band of TTNtv+ sarcomeres, which probably contribute, possibly via faulty mechanosensor function, to the development of manifest DCM.


Sujet(s)
Cardiomyopathie dilatée , Connectine , Humains , Cardiomyopathie dilatée/génétique , Connectine/génétique , Connectine/métabolisme , Coeur , Sarcomères/génétique , Sarcomères/métabolisme
20.
JCI Insight ; 9(2)2024 Jan 23.
Article de Anglais | MEDLINE | ID: mdl-38032735

RÉSUMÉ

Hyperuricemia is implicated in numerous pathologies, but the mechanisms underlying uric acid production are poorly understood. Using a combination of mouse studies, cell culture studies, and human serum samples, we sought to determine the cellular source of uric acid. In mice, fasting and glucocorticoid treatment increased serum uric acid and uric acid release from ex vivo-incubated skeletal muscle. In vitro, glucocorticoids and the transcription factor FoxO3 increased purine nucleotide degradation and purine release from differentiated muscle cells, which coincided with the transcriptional upregulation of AMP deaminase 3, a rate-limiting enzyme in adenine nucleotide degradation. Heavy isotope tracing during coculture experiments revealed that oxidation of muscle purines to uric acid required their transfer from muscle cells to a cell type that expresses xanthine oxidoreductase, such as endothelial cells. Last, in healthy women, matched for age and body composition, serum uric acid was greater in individuals scoring below average on standard physical function assessments. Together, these studies reveal skeletal muscle purine degradation is an underlying driver of uric acid production, with the final step of uric acid production occurring primarily in a nonmuscle cell type. This suggests that skeletal muscle fiber purine degradation may represent a therapeutic target to reduce serum uric acid and treat numerous pathologies.


Sujet(s)
Cellules endothéliales , Acide urique , Humains , Femelle , Souris , Animaux , Acide urique/métabolisme , Cellules endothéliales/métabolisme , Xanthine dehydrogenase , Muscles squelettiques/métabolisme , Oxydoréduction
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...