Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Respir Res ; 24(1): 305, 2023 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-38057814

RESUMO

INTRODUCTION: Biomarkers are needed to inform the choice of biologic therapy in patients with asthma given the increasing number of biologics. We aimed to identify proteins associated with response to omalizumab and mepolizumab. METHODS: Aptamer-based proteomic profiling (SomaScan) was used to assess 1437 proteins from 51 patients with moderate to severe asthma who received omalizumab (n = 29) or mepolizumab (n = 22). Response was defined as the change in asthma-related exacerbations in the 12 months following therapy initiation. All models were adjusted for age, sex, and pre-treatment exacerbation rate. Additionally, body mass index was included in the omalizumab model and eosinophil count in the mepolizumab model. We evaluated the association between molecular signatures and response using negative binomial regression correcting for the false discovery rate (FDR) and gene set enrichment analyses (GSEA) to identify associated pathways. RESULTS: Over two-thirds of patients were female. The average age for omalizumab patients was 42 years and 57 years for mepolizumab. At baseline, the average exacerbation rate was 1.5/year for omalizumab and 2.4/year for mepolizumab. Lower levels of LOXL2 (unadjusted p: 1.93 × 10E-05, FDR-corrected: 0.028) and myostatin (unadjusted: 3.87 × 10E-05, FDR-corrected: 0.028) were associated with better response to mepolizumab. Higher levels of CD9 antigen (unadjusted: 5.30 × 10E-07, FDR-corrected: 0.0006) and MUC1 (unadjusted: 1.15 × 10E-06, FDR-corrected: 0.0006) were associated with better response to omalizumab, and LTB4R (unadjusted: 1.12 × 10E-06, FDR-corrected: 0.0006) with worse response. Protein-protein interaction network modeling showed an enrichment of the TNF- and NF-kB signaling pathways for patients treated with mepolizumab and multiple pathways involving MAPK, including the FcER1 pathway, for patients treated with omalizumab. CONCLUSIONS: This study provides novel fundamental data on proteins associated with response to mepolizumab or omalizumab in severe asthma and warrants further validation as potential biomarkers for therapy selection.


Assuntos
Antiasmáticos , Asma , Humanos , Feminino , Adulto , Masculino , Omalizumab/uso terapêutico , Omalizumab/efeitos adversos , Miostatina/uso terapêutico , Proteômica , Asma/diagnóstico , Asma/tratamento farmacológico , Asma/induzido quimicamente , Biomarcadores , Mucina-1
2.
BMC Pediatr ; 22(1): 632, 2022 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-36329412

RESUMO

Spinal muscular atrophy (5q-SMA; SMA), a genetic neuromuscular condition affecting spinal motor neurons, is caused by defects in both copies of the SMN1 gene that produces survival motor neuron (SMN) protein. The highly homologous SMN2 gene primarily expresses a rapidly degraded isoform of SMN protein that causes anterior horn cell degeneration, progressive motor neuron loss, skeletal muscle atrophy and weakness. Severe cases result in limited mobility and ventilatory insufficiency. Untreated SMA is the leading genetic cause of death in young children. Recently, three therapeutics that increase SMN protein levels in patients with SMA have provided incremental improvements in motor function and developmental milestones and prevented the worsening of SMA symptoms. While the therapeutic approaches with Spinraza®, Zolgensma®, and Evrysdi® have a clinically significant impact, they are not curative. For many patients, there remains a significant disease burden. A potential combination therapy under development for SMA targets myostatin, a negative regulator of muscle mass and strength. Myostatin inhibition in animal models increases muscle mass and function. Apitegromab is an investigational, fully human, monoclonal antibody that specifically binds to proforms of myostatin, promyostatin and latent myostatin, thereby inhibiting myostatin activation. A recently completed phase 2 trial demonstrated the potential clinical benefit of apitegromab by improving or stabilizing motor function in patients with Type 2 and Type 3 SMA and providing positive proof-of-concept for myostatin inhibition as a target for managing SMA. The primary goal of this manuscript is to orient physicians to the evolving landscape of SMA treatment.


Assuntos
Atrofia Muscular Espinal , Miostatina , Animais , Criança , Pré-Escolar , Humanos , Neurônios Motores/metabolismo , Atrofia Muscular Espinal/genética , Atrofia Muscular Espinal/terapia , Miostatina/genética , Miostatina/metabolismo , Miostatina/uso terapêutico , Ensaios Clínicos Fase II como Assunto
3.
Int J Chron Obstruct Pulmon Dis ; 17: 2383-2399, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36185172

RESUMO

Objective: Skeletal muscle dysfunction is an important comorbidity in patients with chronic obstructive pulmonary disease (COPD), and is associated with poor quality of life and reduced survival, but the mechanisms involved remain elusive. Ferroptosis is a newly discovered type of cell death resulting from iron-dependent lipid peroxide accumulation. The purpose of this study was to examine whether ferroptosis is involved in COPD-associated skeletal muscle dysfunction. Methods: A mouse model of COPD was established after 24 weeks of cigarette smoke (CS) exposure, and mRNA sequencing, hematoxylin-eosin (H&E) staining, immunostaining (IF), RT-PCR, and Western blot were utilized to identify the changes in gastrocnemius muscles. In vitro, C2C12 myotubes were treated with CS extract (CSE) and evaluated for ferroptosis-related molecules. The pathways regulating ferroptosis were then explored in CSE-stimulated myotubes. Results: Compared with controls, COPD mice showed an enriched ferroptosis pathway. Gpx4 was decreased, while hypoxia-inducible factor (Hif) 2α was increased, at gene and protein levels. A reduced level of GSH, but increased cell death, Fe2+, lipid ROS, LPO, and 4-HNE were observed in COPD mice or in CSE-stimulated C2C12 myotubes, which could be ameliorated by ferroptosis inhibitors. The expression of myostatin (MSTN) was enhanced in COPD mice and CSE-stimulated myotubes. MSTN up-regulated HIF2α expression and led to ferroptosis in myotubes, whereas inhibition of MSTN binding to its receptor or inhibition/knockdown of HIF2α resulted in decreased cell death, and partially restored GPX4 and GSH. Conclusion: CS exposure induced ferroptosis in vivo and in vitro. Mechanistically, CS-exposure upregulated MSTN which further induced ferroptosis through HIF2α in skeletal muscles, which may contribute to muscle dysfunction through impairing metabolic capacity and decreasing muscle fiber numbers, revealing a potential novel therapeutic target for COPD-related skeletal muscle dysfunction.


Assuntos
Ferroptose , Doença Pulmonar Obstrutiva Crônica , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Amarelo de Eosina-(YS)/metabolismo , Amarelo de Eosina-(YS)/uso terapêutico , Hematoxilina/metabolismo , Hematoxilina/uso terapêutico , Ferro , Peróxidos Lipídicos/metabolismo , Camundongos , Músculo Esquelético/metabolismo , Miostatina/metabolismo , Miostatina/uso terapêutico , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico , Qualidade de Vida , RNA Mensageiro/metabolismo , Espécies Reativas de Oxigênio/metabolismo
4.
Clin Pharmacol Ther ; 112(6): 1291-1302, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36104012

RESUMO

Myostatin, a negative regulator of skeletal muscle growth, is a therapeutic target in muscle-wasting diseases. Domagrozumab, a humanized recombinant monoclonal antibody, binds myostatin and inhibits activity. Domagrozumab was investigated in a phase II trial (NCT02310763) as a potential treatment for boys with Duchenne muscular dystrophy (DMD). Pharmacokinetic/pharmacodynamic (PK/PD) modeling is vital in clinical trial design, particularly for determining dosing regimens in pediatric populations. This analysis sought to establish the PK/PD relationship between free domagrozumab and total myostatin concentrations in pediatric patients with DMD using a prior semimechanistic model developed from a phase I study in healthy adult volunteers (NCT01616277) and following inclusion of phase II data. The refined model was developed using a multiple-step approach comprising structural, random effects, and covariate model development; assessment of model adequacy (goodness-of-fit); and predictive performance. Differences in PKs/PDs between healthy adult volunteers and pediatric patients with DMD were quantitatively accounted for and evaluated by predicting myostatin coverage (the percentage of myostatin bound by domagrozumab). The final model parameter estimates and semimechanistic target-mediated drug disposition structure sufficiently described both domagrozumab and myostatin concentrations in pediatric patients with DMD, and most population parameters were comparable with the prior model (in healthy adult volunteers). Predicted myostatin coverage for phase II patients with DMD was consistently > 90%. Baseline serum myostatin was ~ 65% lower than in healthy adult volunteers. This study provides insights into the regulation of myostatin in healthy adults and pediatric patients with DMD. Clinicaltrials.gov identifiers: NCT01616277 and NCT02310763.


Assuntos
Distrofia Muscular de Duchenne , Humanos , Criança , Adulto , Masculino , Distrofia Muscular de Duchenne/tratamento farmacológico , Miostatina/metabolismo , Miostatina/uso terapêutico , Anticorpos Monoclonais Humanizados/uso terapêutico , Voluntários Saudáveis , Músculo Esquelético/metabolismo
5.
Cell Mol Life Sci ; 79(7): 374, 2022 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-35727341

RESUMO

Myostatin is a negative regulator of skeletal muscle growth secreted by skeletal myocytes. In the past years, myostatin inhibition sparked interest among the scientific community for its potential to enhance muscle growth and to reduce, or even prevent, muscle atrophy. These characteristics make it a promising target for the treatment of muscle atrophy in motor neuron diseases, namely, amyotrophic lateral sclerosis (ALS) and spinal muscular atrophy (SMA), which are rare neurological diseases, whereby the degeneration of motor neurons leads to progressive muscle loss and paralysis. These diseases carry a huge burden of morbidity and mortality but, despite this unfavorable scenario, several therapeutic advancements have been made in the past years. Indeed, a number of different curative therapies for SMA have been approved, leading to a revolution in the life expectancy and outcomes of SMA patients. Similarly, tofersen, an antisense oligonucleotide, is now undergoing clinical trial phase for use in ALS patients carrying the SOD1 mutation. However, these therapies are not able to completely halt or reverse progression of muscle damage. Recently, a trial evaluating apitegromab, a myostatin inhibitor, in SMA patients was started, following positive results from preclinical studies. In this context, myostatin inhibition could represent a useful strategy to tackle motor symptoms in these patients. The aim of this review is to describe the myostatin pathway and its role in motor neuron diseases, and to summarize and critically discuss preclinical and clinical studies of myostatin inhibitors in SMA and ALS. Then, we will highlight promises and pitfalls related to the use of myostatin inhibitors in the human setting, to aid the scientific community in the development of future clinical trials.


Assuntos
Esclerose Lateral Amiotrófica , Atrofia Muscular Espinal , Esclerose Lateral Amiotrófica/tratamento farmacológico , Esclerose Lateral Amiotrófica/genética , Esclerose Lateral Amiotrófica/metabolismo , Animais , Modelos Animais de Doenças , Humanos , Músculo Esquelético/metabolismo , Atrofia Muscular/metabolismo , Atrofia Muscular Espinal/tratamento farmacológico , Atrofia Muscular Espinal/genética , Miostatina/genética , Miostatina/metabolismo , Miostatina/uso terapêutico , Transdução de Sinais
6.
Br J Pharmacol ; 179(6): 1237-1250, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34553378

RESUMO

BACKGROUND AND PURPOSE: Duchenne muscular dystrophy (DMD) is a degenerative muscle disease with no effective drug treatment. This study investigated the positive effects of fenofibrate on dystrophic muscles. EXPERIMENTAL APPROACH: Myostatin expression in serum and muscle tissue from patients with Duchenne muscular dystrophy and mdx mice were tested. Primary myoblasts isolated from mdx mice were challenged with an inflammatory stimulus and treated with fenofibrate. In animal experiments, 6-week-old male mdx mice were treated with fenofibrate (100 mg kg-1 ) administered orally once per day for 6 weeks. Effects of fenofibrate were evaluated by tests of muscle function plus histology and biochemical analyses of serum. Expression of myostatin, MuRF1, and atrogin-1 in skeletal muscle was evaluated by western blotting and real-time PCR. Total and oxidative myosin heavy chain (MHC) were assessed via immunofluorescence. KEY RESULTS: Expression of myostatin protein was increased in dystrophic muscle of patients with Duchenne muscular dystrophy and mdx mice. Fenofibrate enhanced myofibre differentiation by down-regulating the expression of myostatin protein but not mRNA in primary myoblasts of mdx mice. Fenofibrate significantly improved muscle function while ameliorating muscle damage in mdx mice. These benefits were accompanied by an anti-inflammatory effect. Fenofibrate treatment returned myofibre function by inhibiting the expressions of myostatin, MuRF1, and atrogin-1 protein in the gastrocnemius muscle and diaphragm, while leaving the mRNA level of myostatin unaffected. CONCLUSIONS AND IMPLICATIONS: Fenofibrate substantially slows muscle dystrophy by promoting the degradation of myostatin protein, which may indicate a new therapeutic focus for patients with Duchenne muscular dystrophy.


Assuntos
Fenofibrato , Distrofia Muscular de Duchenne , Animais , Fenofibrato/farmacologia , Fenofibrato/uso terapêutico , Humanos , Masculino , Camundongos , Camundongos Endogâmicos mdx , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/tratamento farmacológico , Distrofia Muscular de Duchenne/metabolismo , Miostatina/metabolismo , Miostatina/farmacologia , Miostatina/uso terapêutico
7.
Mol Med Rep ; 22(6): 4947-4955, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33173958

RESUMO

Charcot­Marie­Tooth disease (CMT) is the most common inherited neurological disorder of the peripheral nervous system. The major subtype, CMT type 1A (CMT1A), accounts for ~40% of CMT cases and is characterized by distal muscle atrophy and gait disturbances. Short hairpin (sh) RNA sequences are potentially advantageous therapeutic tools for distal muscle atrophy­induced gait disturbance. Therefore, the current study focused on the effects of an optimal shRNA injection using the myostatin (mstn) gene inhibition system. shLenti­Mstn A demonstrated significant suppression of endogenous mstn gene expression (>40%) via RT­qPCR following direct injection into the gastrocnemius and rectus femoris of the hind limb in C22 mice. The results also reported that shLenti­Mstn A treatment increased muscle mass and size of the hind limbs compared with mock­treated mice via measurement of the mass of injected muscles and magnetic resonance imaging study. Furthermore, electrophysiological measurement using a Nicolet Viking Quest device revealed significantly improved compound muscle action potential (CMAP) in shLenti­Mstn A­treated mice compared with the mock group (P<0.05) whereas nerve conduction velocity (NCV) showed no difference between groups. The shLenti­Mstn A treatment directly affected increased muscle regeneration, including mass and size, but not regeneration of peripheral nerve. Additionally, shLenti­Mstn A treatment significantly enhanced mobility, including locomotor coordination (P<0.01) and grip strength of the hindlimbs (P<0.01). Furthermore, MotoRater analysis using real­time recording with a high­speed camera revealed that shLenti­Mstn­treated mice exhibited an improved walking pattern in terms of step length, base support and duty factor compared with the mock group. It was hypothesized that treatment with shLenti­Mstn A may provide a novel therapeutic strategy for improving gait in patients with CMT1A.


Assuntos
Doença de Charcot-Marie-Tooth/terapia , Miostatina/genética , RNA Interferente Pequeno/uso terapêutico , Animais , Doença de Charcot-Marie-Tooth/genética , Modelos Animais de Doenças , Marcha/genética , Marcha/fisiologia , Humanos , Masculino , Camundongos , Camundongos Transgênicos , Músculo Esquelético/patologia , Atrofia Muscular/genética , Atrofia Muscular/patologia , Atrofia Muscular/terapia , Miostatina/uso terapêutico , Condução Nervosa , RNA Interferente Pequeno/genética
8.
Circ Res ; 118(7): 1143-50; discussion 1150, 2016 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-27034276

RESUMO

This "Controversies in Cardiovascular Research" article evaluates the evidence for and against the hypothesis that the circulating blood level of growth differentiation factor 11 (GDF11) decreases in old age and that restoring normal GDF11 levels in old animals rejuvenates their skeletal muscle and reverses pathological cardiac hypertrophy and cardiac dysfunction. Studies supporting the original GDF11 hypothesis in skeletal and cardiac muscle have not been validated by several independent groups. These new studies have either found no effects of restoring normal GDF11 levels on cardiac structure and function or have shown that increasing GDF11 or its closely related family member growth differentiation factor 8 actually impairs skeletal muscle repair in old animals. One possible explanation for what seems to be mutually exclusive findings is that the original reagent used to measure GDF11 levels also detected many other molecules so that age-dependent changes in GDF11 are still not well known. The more important issue is whether increasing blood [GDF11] repairs old skeletal muscle and reverses age-related cardiac pathologies. There are substantial new and existing data showing that GDF8/11 can exacerbate rather than rejuvenate skeletal muscle injury in old animals. There is also new evidence disputing the idea that there is pathological hypertrophy in old C57bl6 mice and that GDF11 therapy can reverse cardiac pathologies. Finally, high [GDF11] causes reductions in body and heart weight in both young and old animals, suggestive of a cachexia effect. Our conclusion is that elevating blood levels of GDF11 in the aged might cause more harm than good.


Assuntos
Envelhecimento/patologia , Proteínas Morfogenéticas Ósseas/uso terapêutico , Fatores de Diferenciação de Crescimento/uso terapêutico , Doenças Musculares/tratamento farmacológico , Envelhecimento/sangue , Animais , Proteínas Morfogenéticas Ósseas/sangue , Proteínas Morfogenéticas Ósseas/deficiência , Proteínas Morfogenéticas Ósseas/farmacologia , Proteínas Morfogenéticas Ósseas/toxicidade , Caquexia/induzido quimicamente , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos , Fatores de Diferenciação de Crescimento/sangue , Fatores de Diferenciação de Crescimento/deficiência , Fatores de Diferenciação de Crescimento/farmacologia , Fatores de Diferenciação de Crescimento/toxicidade , Coração/efeitos dos fármacos , Humanos , Hipertrofia , Camundongos Endogâmicos C57BL , Modelos Animais , Músculo Esquelético/lesões , Músculo Esquelético/fisiologia , Músculos/patologia , Doenças Musculares/fisiopatologia , Miocárdio/patologia , Miostatina/fisiologia , Miostatina/uso terapêutico , Miostatina/toxicidade , Parabiose , Proteínas Recombinantes/uso terapêutico , Proteínas Recombinantes/toxicidade , Regeneração/efeitos dos fármacos , Reprodutibilidade dos Testes , Transdução de Sinais , Método Simples-Cego , Proteína Smad2/fisiologia , Proteína Smad3/fisiologia
9.
Biomaterials ; 81: 93-103, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26724457

RESUMO

Duchenne's muscular dystrophy (DMD) is a neuromuscular disorder accompanied with muscle weakness and wasting. Since myostatin was reported to be a key regulator of muscle wasting, myostatin inhibitors have been investigated as therapeutic candidates for the treatment of muscular diseases. Here, we report an antigenic peptide of myostatin fragment (MstnF) conjugated to hyaluronate (HA) with a low molecular weight (MW, 17 kDa) for transdermal immunotherapy of DMD. Facilitating the transdermal delivery, the low MW HA showed a boosting effect on the immunization of MstnF possibly by engaging both toll-like receptors and cluster of differentiation 44 (CD44). In vivo two-photon microscopy clearly visualized the effective transdermal penetration of HA-MstnF conjugates into deep intact skin layers. The transdermal immunization of mdx mice significantly increased antibody titers against myostatin. Furthermore, the mdx mice immunized with HA-MstnF conjugates resulted in statistically significant improvement in the biochemical and pathological status of skeletal musculature as well as functional behaviors.


Assuntos
Adjuvantes Imunológicos/farmacologia , Antígenos/metabolismo , Ácido Hialurônico/farmacologia , Distrofia Muscular Animal/tratamento farmacológico , Peptídeos/uso terapêutico , Administração Cutânea , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Citocinas/metabolismo , Células Dendríticas/citologia , Células Dendríticas/efeitos dos fármacos , Endocitose/efeitos dos fármacos , Imunização , Mediadores da Inflamação/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Microscopia de Fluorescência por Excitação Multifotônica , Peso Molecular , Distrofia Muscular Animal/imunologia , Distrofia Muscular Animal/patologia , Miostatina/uso terapêutico , Peptídeos/farmacologia
10.
Nat Rev Drug Discov ; 14(1): 58-74, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25549588

RESUMO

Atrophy occurs in specific muscles with inactivity (for example, during plaster cast immobilization) or denervation (for example, in patients with spinal cord injuries). Muscle wasting occurs systemically in older people (a condition known as sarcopenia); as a physiological response to fasting or malnutrition; and in many diseases, including chronic obstructive pulmonary disorder, cancer-associated cachexia, diabetes, renal failure, cardiac failure, Cushing syndrome, sepsis, burns and trauma. The rapid loss of muscle mass and strength primarily results from excessive protein breakdown, which is often accompanied by reduced protein synthesis. This loss of muscle function can lead to reduced quality of life, increased morbidity and mortality. Exercise is the only accepted approach to prevent or slow atrophy. However, several promising therapeutic agents are in development, and major advances in our understanding of the cellular mechanisms that regulate the protein balance in muscle include the identification of several cytokines, particularly myostatin, and a common transcriptional programme that promotes muscle wasting. Here, we discuss these new insights and the rationally designed therapies that are emerging to combat muscle wasting.


Assuntos
Atrofia Muscular/diagnóstico , Atrofia Muscular/tratamento farmacológico , Qualidade de Vida , Animais , Humanos , Atrofia Muscular/epidemiologia , Miofibrilas/efeitos dos fármacos , Miofibrilas/patologia , Miostatina/farmacologia , Miostatina/uso terapêutico , Sepse/diagnóstico , Sepse/tratamento farmacológico , Sepse/epidemiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Síndrome de Emaciação/diagnóstico , Síndrome de Emaciação/tratamento farmacológico , Síndrome de Emaciação/epidemiologia
11.
Am J Phys Med Rehabil ; 93(11 Suppl 3): S88-96, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24879554

RESUMO

Skeletal muscle is a fundamental organ in the generation of force and movement, the regulation of whole-body metabolism, and the provision of resiliency. Indeed, physical medicine and rehabilitation is recognized for optimizing skeletal muscle health in the context of aging (sarcopenia) and disease (cachexia). Exercise is, and will remain, the cornerstone of therapies to improve skeletal muscle health. However, there are now a number of promising biologic and small molecule interventions currently under development to rejuvenate skeletal muscle, including myostatin inhibitors, selective androgen receptor modulators, and an activator of the fast skeletal muscle troponin complex. The opportunities for skeletal muscle-based regenerative therapies and a selection of emerging pharmacologic interventions are discussed in this review.


Assuntos
Envelhecimento/fisiologia , Exercício Físico/fisiologia , Terapia de Reposição Hormonal/métodos , Força Muscular/efeitos dos fármacos , Miostatina/antagonistas & inibidores , Sarcopenia/reabilitação , Idoso , Idoso de 80 Anos ou mais , Caquexia/fisiopatologia , Caquexia/terapia , Terapia Combinada , Estrogênios/uso terapêutico , Feminino , Idoso Fragilizado , Humanos , Masculino , Contração Muscular/efeitos dos fármacos , Contração Muscular/fisiologia , Força Muscular/fisiologia , Debilidade Muscular/fisiopatologia , Debilidade Muscular/reabilitação , Miostatina/uso terapêutico , Prognóstico , Ensaios Clínicos Controlados Aleatórios como Assunto , Sarcopenia/fisiopatologia , Índice de Gravidade de Doença , Testosterona/uso terapêutico , Resultado do Tratamento
12.
Curr Opin Support Palliat Care ; 7(4): 352-60, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24157714

RESUMO

PURPOSE OF REVIEW: This review summarizes recent progress in the development of myostatin inhibitors for the treatment of muscle wasting disorders. It also focuses on findings in myostatin biology that may have implications for the development of antimyostatin therapies. RECENT FINDINGS: There has been progress in evaluating antimyostatin therapies in animal models of muscle wasting disorders. Some programs have progressed into clinical development with initial results showing positive impact on muscle volume.In normal mice myostatin deficiency results in enlarged muscles with increased total force but decreased specific force (total force/total mass). An increase in myofibrillar protein synthesis without concomitant satellite cell proliferation and fusion leads to muscle hypertrophy with unchanged myonuclear number. A specific force reduction is not observed when atrophied muscle, the predominant therapeutic target of myostatin inhibitor therapy, is made myostatindeficient.Myostatin has been shown to be expressed by a number of tumor cell lines in mice and man. SUMMARY: Myostatin inhibition remains a promising therapeutic strategy for a range of muscle wasting disorders.


Assuntos
Caquexia/tratamento farmacológico , Atrofia Muscular/tratamento farmacológico , Miostatina/antagonistas & inibidores , Neoplasias/complicações , Animais , Caquexia/complicações , Caquexia/etiologia , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/patologia , Atrofia Muscular/etiologia , Miostatina/uso terapêutico , Neoplasias/tratamento farmacológico
13.
Exp Gerontol ; 48(9): 898-904, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23832079

RESUMO

Loss of muscle and bone mass with age are significant contributors to falls and fractures among the elderly. Myostatin deficiency is associated with increased muscle mass in mice, dogs, cows, sheep and humans, and mice lacking myostatin have been observed to show increased bone density in the limb, spine, and jaw. Transgenic overexpression of myostatin propeptide, which binds to and inhibits the active myostatin ligand, also increases muscle mass and bone density in mice. We therefore sought to test the hypothesis that in vivo inhibition of myostatin using an injectable myostatin propeptide (GDF8 propeptide-Fc) would increase both muscle mass and bone density in aged (24 mo) mice. Male mice were injected weekly (20 mg/kg body weight) with recombinant myostatin propeptide-Fc (PRO) or vehicle (VEH; saline) for four weeks. There was no difference in body weight between the two groups at the end of the treatment period, but PRO treatment significantly increased mass of the tibialis anterior muscle (+ 7%) and increased muscle fiber diameter of the extensor digitorum longus (+ 16%) and soleus (+ 6%) muscles compared to VEH treatment. Bone volume relative to total volume (BV/TV) of the femur calculated by microCT did not differ significantly between PRO- and VEH-treated mice, and ultimate force (Fu), stiffness (S), toughness (U) measured from three-point bending tests also did not differ significantly between groups. Histomorphometric assays also revealed no differences in bone formation or resorption in response to PRO treatment. These data suggest that while developmental perturbation of myostatin signaling through either gene knockout or transgenic inhibition may alter both muscle and bone mass in mice, pharmacological inhibition of myostatin in aged mice has a more pronounced effect on skeletal muscle than on bone.


Assuntos
Densidade Óssea/efeitos dos fármacos , Músculo Esquelético/efeitos dos fármacos , Miostatina/uso terapêutico , Osteoporose/tratamento farmacológico , Sarcopenia/tratamento farmacológico , Envelhecimento/patologia , Envelhecimento/fisiologia , Animais , Peso Corporal/efeitos dos fármacos , Densidade Óssea/fisiologia , Avaliação Pré-Clínica de Medicamentos/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/patologia , Músculo Esquelético/patologia , Miostatina/antagonistas & inibidores , Miostatina/deficiência , Miostatina/farmacologia , Osteogênese/efeitos dos fármacos , Osteogênese/fisiologia , Osteoporose/patologia , Osteoporose/fisiopatologia , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Sarcopenia/patologia , Sarcopenia/fisiopatologia , Estresse Mecânico , Tíbia/efeitos dos fármacos , Tíbia/fisiopatologia , Microtomografia por Raio-X/métodos
14.
Cell Death Dis ; 4: e494, 2013 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-23412387

RESUMO

Myostatin, a member of the transforming growth factor-ß superfamily, regulates the glucose metabolism of muscle cells, while dysregulated myostatin activity is associated with a number of metabolic disorders, including muscle cachexia, obesity and type II diabetes. We observed that myostatin induced significant mitochondrial metabolic alterations and prolonged exposure of myostatin induced mitochondria-dependent apoptosis in cancer cells addicted to glycolysis. To address the underlying mechanism, we found that the protein levels of Hexokinase II (HKII) and voltage-dependent anion channel 1 (VDAC1), two key regulators of glucose metabolisms as well as metabolic stress-induced apoptosis, were negatively correlated. In particular, VDAC1 was dramatically upregulated in cells that are sensitive to myostatin treatment whereas HKII was downregulated and dissociated from mitochondria. Myostatin promoted the translocation of Bax from cytosol to mitochondria, and knockdown of VDAC1 inhibited myostatin-induced Bax translocation and apoptosis. These apoptotic changes can be partially rescued by repletion of ATP, or by ectopic expression of HKII, suggesting that perturbation of mitochondrial metabolism is causally linked with subsequent apoptosis. Our findings reveal novel function of myostatin in regulating mitochondrial metabolism and apoptosis in cancer cells.


Assuntos
Apoptose/efeitos dos fármacos , Mitocôndrias/metabolismo , Miostatina/toxicidade , Trifosfato de Adenosina , Animais , Linhagem Celular Tumoral , Regulação para Baixo , Glucose/metabolismo , Células HeLa , Hexoquinase/metabolismo , Humanos , Células MCF-7 , Melanoma/tratamento farmacológico , Melanoma/metabolismo , Melanoma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Miostatina/uso terapêutico , Fosforilação Oxidativa/efeitos dos fármacos , Transplante Heterólogo , Regulação para Cima , Canal de Ânion 1 Dependente de Voltagem/metabolismo , Proteína X Associada a bcl-2/metabolismo
15.
Int J Urol ; 20(5): 522-9, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23050675

RESUMO

OBJECTIVES: Myostatin, a member of the transforming growth factor-ß superfamily, is a negative regulator of myogenesis in skeletal muscle. We examined the effect of myostatin and myostatin inhibition by an antagonistic agent, follistatin, on growth of human urethral rhabdosphincter satellite cells (muscle stem cells) to develop a new strategy for treatment of stress urinary incontinence. METHODS: Rhabdosphincter satellite cells were cultured and selected by magnetic affinity cell sorting using an anti-neural cell adhesion molecule antibody. The cells were transfected with simian virus-40 antigen to extend their lifespan. A cell proliferation assay, a cell cycle analysis and an investigation of signal transduction were carried out. The autocrine action of endogenous myostatin by western blotting, real-time reverse transcription polymerase chain reaction and immunoneutralization using an anti-myostatin antibody was also evaluated. RESULTS: Selectively cultured cells expressed markers of striated muscles and successfully differentiated into myotubes. Myostatin inhibited proliferation of these cells through Smad2 phosphorylation and cell cycle arrest. Inhibitory effects of myostatin were reversed by addition of follistatin. However, rhabdosphincter satellite cells did not appear to use autocrine secretion of myostatin to regulate their proliferation. CONCLUSIONS: Inhibition of myostatin function might be a useful pathway in the development of novel strategies for stimulating rhabdosphincter cells regeneration to treat stress urinary incontinence.


Assuntos
Proliferação de Células/efeitos dos fármacos , Miostatina/farmacologia , Uretra/efeitos dos fármacos , Incontinência Urinária por Estresse/tratamento farmacológico , Comunicação Autócrina , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos , Humanos , Miostatina/uso terapêutico , Transdução de Sinais/efeitos dos fármacos
16.
Acta Myol ; 31(1): 4-8, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22655510

RESUMO

Duchenne muscular dystrophy (DMD) is a disease linked to the X-chromosome which affects 1 in 3,600-6,000 newborn males. It is manifested by the absence of the dystrophin protein in muscle fibres, which causes progressive damage leading to death in the third decade of life. The only medication so far shown to be effective in delaying the progression of this illness are corticosteroids, which have been shown to increase muscle strength in randomised controlled studies; long-term studies have demonstrated that they prolong walking time and retard the progression of respiratory dysfunction, dilated cardiomyopathy and scoliosis. Several potential drugs are now being investigated. Genetic therapy, involving the insertion of a dystrophin gene through a vector, has proven effective in animals but not humans. Currently under clinical study is Ataluren, a molecule that binds with ribosomes and may allow the insertion of an aminoacid in the premature termination codon, and exon-skipping, which binds with RNA and excludes specific sites of RNA splicing, producing a dystrophin that is smaller but functional. There are also studies attempting to modulate other muscular proteins, such as myostatin and utrophin, to reduce symptoms. This paper does not address cardiomyopathy treatment in DMD patients.


Assuntos
Glucocorticoides/uso terapêutico , Imunossupressores/uso terapêutico , Distrofia Muscular de Duchenne/tratamento farmacológico , Aminoglicosídeos/uso terapêutico , Terapia Genética , Humanos , Miostatina/uso terapêutico , Oxidiazóis/uso terapêutico , Prednisona/uso terapêutico , Pregnenodionas/uso terapêutico , Utrofina/uso terapêutico
17.
J Histochem Cytochem ; 60(1): 22-30, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22205678

RESUMO

The time course and cellular localization of myostatin expression following musculoskeletal injury are not well understood; therefore, the authors evaluated the temporal and spatial localization of myostatin during muscle and bone repair following deep penetrant injury in a mouse model. They then used hydrogel delivery of exogenous myostatin in the same injury model to determine the effects of myostatin exposure on muscle and bone healing. Results showed that a "pool" of intense myostatin staining was observed among injured skeletal muscle fibers 12-24 hr postsurgery and that myostatin was also expressed in the soft callus chondrocytes 4 days following osteotomy. Hydrogel delivery of 10 or 100 µg/ml recombinant myostatin decreased fracture callus cartilage area relative to total callus area in a dose-dependent manner by 41% and 80% (p<0.05), respectively, compared to vehicle treatment. Myostatin treatment also decreased fracture callus total bone volume by 30.6% and 38.8% (p<0.05), with the higher dose of recombinant myostatin yielding the greatest decrease in callus bone volume. Finally, exogenous myostatin treatment caused a significant dose-dependent increase in fibrous tissue formation in skeletal muscle. Together, these findings suggest that early pharmacological inhibition of myostatin is likely to improve the regenerative potential of both muscle and bone following deep penetrant musculoskeletal injury.


Assuntos
Osso e Ossos/lesões , Músculo Esquelético/lesões , Miostatina/metabolismo , Cicatrização/efeitos dos fármacos , Ferimentos Penetrantes/metabolismo , Animais , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/metabolismo , Calo Ósseo/efeitos dos fármacos , Calo Ósseo/metabolismo , Calo Ósseo/patologia , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Condrócitos/patologia , Fíbula/efeitos dos fármacos , Fíbula/lesões , Fíbula/metabolismo , Consolidação da Fratura/efeitos dos fármacos , Hidrogéis , Imuno-Histoquímica , Camundongos , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Miostatina/farmacologia , Miostatina/uso terapêutico , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Regeneração/efeitos dos fármacos , Ferimentos Penetrantes/tratamento farmacológico
18.
J Trauma ; 69(3): 579-83, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20173658

RESUMO

BACKGROUND: Myostatin (GDF-8) is known as a potent inhibitor of muscle growth and development, and myostatin is also expressed early in the fracture healing process. The purpose of this study was to test the hypothesis that a new myostatin inhibitor, a recombinant myostatin propeptide, can enhance the repair and regeneration of both muscle and bone in cases of deep penetrant injury. METHODS: We used a fibula osteotomy model with associated damage to lateral compartment muscles (fibularis longus and brevis) in mice to test the hypothesis that blocking active myostatin with systemic injections of a recombinant myostatin propeptide would improve muscle and bone repair. Mice were assigned to two treatment groups after undergoing a fibula osteotomy: those receiving either vehicle (saline) or recombinant myostatin propeptide (20 mg/kg). Mice received one injection on the day of surgery, another injection 5 days after surgery, and a third injection 10 days after surgery. Mice were killed 15 days after the osteotomy procedure. Bone repair was assessed using microcomputed tomography (micro-CT) and histologic evaluation of the fracture callus. Muscle healing was assessed using Masson trichrome staining of the injury site, and image analysis was used to quantify the degree of fibrosis and muscle regeneration. RESULTS: Three propeptide injections over a period of 15 days increased body mass by 7% and increased muscle mass by almost 20% (p < 0.001). Micro-CT analysis of the osteotomy site shows that by 15 days postosteotomy, bony callus tissue was observed bridging the osteotomy gap in 80% of the propeptide-treated mice but only 40% of the control (vehicle)-treated mice (p < 0.01). Micro-CT quantification shows that bone volume of the fracture callus was increased by ∼ 30% (p < 0.05) with propeptide treatment, and the increase in bone volume was accompanied by a significant increase in cartilage area (p = 0.01). Propeptide treatment significantly decreased the fraction of fibrous tissue in the wound site and increased the fraction of muscle relative to fibrous tissue by 20% (p < 0.01). CONCLUSIONS: Blocking myostatin signaling in the injured limb improves fracture healing and enhances muscle regeneration. These data suggest that myostatin inhibitors may be effective for improving wound repair in cases of orthopaedic trauma and extremity injury.


Assuntos
Osso e Ossos/lesões , Músculos/lesões , Miostatina/uso terapêutico , Cicatrização/efeitos dos fármacos , Ferimentos Penetrantes/tratamento farmacológico , Animais , Regeneração Óssea/efeitos dos fármacos , Osso e Ossos/efeitos dos fármacos , Modelos Animais de Doenças , Fraturas Ósseas/tratamento farmacológico , Masculino , Camundongos , Músculos/efeitos dos fármacos , Miostatina/antagonistas & inibidores , Osteotomia , Proteínas Recombinantes/uso terapêutico , Microtomografia por Raio-X
19.
Curr Pharm Des ; 16(8): 978-87, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20041828

RESUMO

Muscle degeneration and regeneration are two of the most evident pathological events characterizing muscular diseases and in particular muscular dystrophies. Muscular dystrophies are an heterogeneous group of hereditary diseases affecting both children and adults, and are characterized by muscle wasting and weakness. Until now at least 30 different genes have been associated with muscular dystrophies. They have been divided into several subgroups depending on the distribution of the muscle weakness. Thus, the histopathological markers of all these forms are dystrophic changes at the muscle biopsy characterized by fiber size variability, fibres necrosis, regeneration, inflammation and connective tissues deposition. As for now, no effective therapy is available for these diseases but new inside has now been expanded in regenerative therapy such as cell therapy and gene therapy. This review is focused on muscular dystrophies and new acknowledgments in regenerative therapy.


Assuntos
Proteínas Musculares/genética , Músculo Esquelético/cirurgia , Distrofias Musculares/terapia , Terapia Genética/métodos , Humanos , Modelos Biológicos , Proteínas Musculares/metabolismo , Distrofias Musculares/genética , Distrofias Musculares/patologia , Mutação , Miostatina/uso terapêutico , Transplante de Células-Tronco/métodos , Fator de Crescimento Transformador beta1/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...