transforming-growth-factor-beta has been researched along with Muscular-Dystrophy--Duchenne* in 53 studies
5 review(s) available for transforming-growth-factor-beta and Muscular-Dystrophy--Duchenne
Article | Year |
---|---|
Pre-clinical drug tests in the mdx mouse as a model of dystrophinopathies: an overview.
Duchenne muscular dystrophy is a lethal X-linked muscle disease affecting 1/3500 live male birth. It results from defects in the subsarcolemmal protein dystrophin, a component of the dystrophin-glycoprotein complex (DGC) which links the intracellular cytoskeleton to the extracellular matrix. The absence of dystrophin leads to muscle membrane fragility, muscle necrosis and gradual replacement of skeletal muscle by fat and connective tissue, through a complex and still unclear cascade of interconnecting events. No cure is currently available, with glucocorticoids being the sole drugs in clinical use in spite of their remarkable side effects. A great effort is devoted at performing pre-clinical tests on the mdx mouse, the mostly used homologous animal model for DMD, with the final aim to identify drugs safer than steroids and able to target the pathogenic mechanisms so to delay pathology progression. This review updates the efforts on this topic, focusing on the open issues about the animal model and highlighting the classes of pharmaceuticals that are more promising as disease-modifiers, while awaiting for more corrective therapies. Although caution is necessary in data transfer from mdx model to DMD patients, the implementation of standard operating procedures and the growing understanding of the pathology may allow a more accurate evaluation of therapeutics, alone or in combination, in pre-clinical settings. A continuous cross-talk with clinicians and patients associations are also crucial points for proper translation of data from mouse to bedside. Topics: Anabolic Agents; Animals; Anti-Inflammatory Agents; Antioxidants; Disease Models, Animal; Dystrophin; Immunosuppressive Agents; Mice; Mice, Inbred mdx; Muscular Dystrophy, Duchenne; Signal Transduction; Transforming Growth Factor beta | 2012 |
Therapeutics in duchenne muscular dystrophy.
Duchenne muscular dystrophy (DMD) is a fatal disorder affecting approximately 1 in 3,500 live born males, characterized by progressive muscle weakness. Several different strategies are being investigated in developing a cure for this disorder. Until a cure is found, therapeutic and supportive care is essential in preventing complications and improving the afflicted child's quality of life. Currently, corticosteroids are the only class of drug that has been extensively studied in this condition, with controversy existing over the use of these drugs, especially in light of the multiple side effects that may occur. The use of nutritional supplements has expanded in recent years as researchers improve our abilities to use gene and stem cell therapies, which will hopefully lead to a cure soon. This article discusses the importance of therapeutic interventions in children with DMD, the current debate over the use of corticosteroids to treat this disease, the growing use of natural supplements as a new means of treating these boys and provides an update on the current state of gene and stem cell therapies. Topics: Adrenal Cortex Hormones; Aminoglycosides; Animals; Antibodies; Genetic Therapy; Humans; Mice; Muscular Dystrophy, Duchenne; Nutrition Therapy; Stem Cell Transplantation; Transforming Growth Factor beta | 2006 |
Molecular, cellular, and pharmacological therapies for Duchenne/Becker muscular dystrophies.
Although the molecular defect causing Duchenne/Becker muscular dystrophy (DMD/BMD) was identified nearly 20 years ago, the development of effective therapeutic strategies has nonetheless remained a daunting challenge. Over the years, a variety of different approaches have been explored in an effort to compensate for the lack of the DMD gene product called dystrophin. This review not only presents some of the most promising molecular, cellular, and pharmacological strategies but also highlights some issues that need to be addressed before considering their implementation. Specifically, we describe current strategies being developed to exogenously deliver healthy copies of the dystrophin gene to dystrophic muscles. We present the findings of several studies that have focused on repairing the mutant dystrophin gene using various approaches. We include a discussion of cell-based therapies that capitalize on the use of myoblast or stem cell transfer. Finally, we summarize the results of several studies that may eventually lead to the development of appropriate drug-based therapies. In this context, we review our current knowledge of the mechanisms regulating expression of utrophin, the autosomal homologue of dystrophin. Given the complexity associated with the dystrophic phenotype, it appears likely that a combinatorial approach involving different therapeutic strategies will be necessary for the appropriate management and eventual treatment of this devastating neuromuscular disease. Topics: Animals; Anti-Bacterial Agents; Dystrophin; Gene Expression Regulation; Genetic Therapy; Genetic Vectors; Glucocorticoids; Humans; Muscular Dystrophy, Duchenne; Mutation; Myoblasts; Myostatin; Stem Cell Transplantation; Transforming Growth Factor beta; Utrophin | 2005 |
[Pharmacological treatments for Duchenne and Becker dystrophies].
Duchenne muscular dystrophy (DMD) is a severe X-linked genetic disease affecting 1 boy out of 3500. DMD is due to the lack of a submembranous cytoskeletal protein named dystrophin, leading to the progressive degeneration of skeletal, cardiac and smooth muscle tissue. A milder form of the disease, Becker muscular dystrophy (BMD), is characterised by the presence of a semi-functional truncated dystrophin, or the full-length dystrophin at reduced level. Three different therapeutic approaches are currently under study, gene therapy, cellular therapy and pharmacological therapy. One of the chosen strategies consists of the overexpression of utrophin, a protein 80% homologous with dystrophin, and able to perform similar functions. In this review, we shall consider studies of pharmacological therapy, the aims of which can be classified in three categories: reversal of dystrophic phenotype, dystrophin expression, utrophin overexpression. Topics: Adrenal Cortex Hormones; Adult; Aminoglycosides; Animals; Carnitine; Child; Child, Preschool; Creatine; Dystrophin; Gene Expression; Genetic Therapy; Growth Substances; Humans; Male; Mice; Muscular Dystrophy, Duchenne; Myostatin; RNA, Antisense; Taurine; Transforming Growth Factor beta; Utrophin | 2005 |
Molecular mechanisms involving IGF-1 and myostatin to induce muscle hypertrophy as a therapeutic strategy for Duchenne muscular dystrophy.
Over the past decade, signalling cascades have been characterised that control key features of muscle growth, including the proliferation, differentiation of muscle precursors, the control cell size (hypertrophy) and cell death. In this review we highlight how two differing signalling molecules, Insulin-like Growth Factor-1 (IGF-1) and myostatin, regulate key steps during muscle development. We discuss how IGF-1 and myostatin signalling cascades can be manipulated to stimulate muscle growth. We summarise experimental data from mdx mouse, the animal model for Duchenne muscular dystrophy, that suggest a therapeutic value of these strategies for patients suffering from muscular dystrophy without redressing the primary cause of the lesion. Topics: Animals; Humans; Insulin-Like Growth Factor I; Mice; Mice, Inbred mdx; Muscle Development; Muscle Hypertonia; Muscular Dystrophy, Duchenne; Myostatin; Signal Transduction; Transforming Growth Factor beta | 2005 |
48 other study(ies) available for transforming-growth-factor-beta and Muscular-Dystrophy--Duchenne
Article | Year |
---|---|
Smad8 Is Increased in Duchenne Muscular Dystrophy and Suppresses miR-1, miR-133a, and miR-133b.
Duchenne muscular dystrophy (DMD) is an X-linked recessive disease characterized by skeletal muscle instability, progressive muscle wasting, and fibrosis. A major driver of DMD pathology stems from aberrant upregulation of transforming growth factor β (TGFβ) signaling. In this report, we investigated the major transducers of TGFβ signaling, i.e., receptor Smads (R-Smads), in DMD patient skeletal muscle and observed a 48-fold increase in Topics: Animals; Mice; Mice, Inbred mdx; MicroRNAs; Muscle, Skeletal; Muscular Dystrophy, Duchenne; RNA, Messenger; Smad8 Protein; Transforming Growth Factor beta | 2022 |
Pharmacological inhibition of HDAC6 improves muscle phenotypes in dystrophin-deficient mice by downregulating TGF-β via Smad3 acetylation.
The absence of dystrophin in Duchenne muscular dystrophy disrupts the dystrophin-associated glycoprotein complex resulting in skeletal muscle fiber fragility and atrophy, associated with fibrosis as well as microtubule and neuromuscular junction disorganization. The specific, non-conventional cytoplasmic histone deacetylase 6 (HDAC6) was recently shown to regulate acetylcholine receptor distribution and muscle atrophy. Here, we report that administration of the HDAC6 selective inhibitor tubastatin A to the Duchenne muscular dystrophy, mdx mouse model increases muscle strength, improves microtubule, neuromuscular junction, and dystrophin-associated glycoprotein complex organization, and reduces muscle atrophy and fibrosis. Interestingly, we found that the beneficial effects of HDAC6 inhibition involve the downregulation of transforming growth factor beta signaling. By increasing Smad3 acetylation in the cytoplasm, HDAC6 inhibition reduces Smad2/3 phosphorylation, nuclear translocation, and transcriptional activity. These findings provide in vivo evidence that Smad3 is a new target of HDAC6 and implicate HDAC6 as a potential therapeutic target in Duchenne muscular dystrophy. Topics: Acetylation; Animals; Dystrophin; Fibrosis; Glycoproteins; Histone Deacetylase 6; Histone Deacetylase Inhibitors; Mice; Mice, Inbred mdx; Muscle, Skeletal; Muscular Atrophy; Muscular Dystrophy, Duchenne; Phenotype; Transforming Growth Factor beta | 2022 |
Anti-latent TGFβ binding protein 4 antibody improves muscle function and reduces muscle fibrosis in muscular dystrophy.
Duchenne muscular dystrophy, like other muscular dystrophies, is a progressive disorder hallmarked by muscle degeneration, inflammation, and fibrosis. Latent transforming growth factor β (TGFβ) binding protein 4 (LTBP4) is an extracellular matrix protein found in muscle. LTBP4 sequesters and inhibits a precursor form of TGFβ. LTBP4 was originally identified from a genome-wide search for genetic modifiers of muscular dystrophy in mice, where there are two different alleles. The protective form of LTBP4, which contains an insertion of 12 amino acids in the protein’s hinge region, was linked to increased sequestration of latent TGFβ, enhanced muscle membrane stability, and reduced muscle fibrosis. The deleterious form of LTBP4 protein, lacking 12 amino acids, was more susceptible to proteolysis and promoted release of latent TGF-β, and together, these data underscored the functional role of LTBP4’s hinge. Here, we generated a monoclonal human anti-LTBP4 antibody directed toward LTBP4’s hinge region. In vitro, anti-LTBP4 bound LTBP4 protein and reduced LTBP4 proteolytic cleavage. In isolated myofibers, the LTBP4 antibody stabilized the sarcolemma from injury. In vivo, anti-LTBP4 treatment of dystrophic mice protected muscle against force loss induced by eccentric contraction. Anti-LTBP4 treatment also reduced muscle fibrosis and enhanced muscle force production, including in the diaphragm muscle, where respiratory function was improved. Moreover, the anti-LTBP4 in combination with prednisone, a standard of care for Duchenne muscular dystrophy, further enhanced muscle function and protected against injury in Topics: Carrier Proteins; Fibrosis; Humans; Latent TGF-beta Binding Proteins; Muscle, Skeletal; Muscles; Muscular Dystrophies; Muscular Dystrophy, Duchenne; Transforming Growth Factor beta | 2021 |
Computational Models Provide Insight into In Vivo Studies and Reveal the Complex Role of Fibrosis in mdx Muscle Regeneration.
Duchenne muscular dystrophy is a pro-fibrotic, muscle wasting disease. Reducing fibrosis is a potential therapeutic target; however, its effect on muscle regeneration is not fully understood. This study (1) used an agent-based model to predict the effect of increased fibrosis in mdx muscle on regeneration from injury, and (2) experimentally tested the resulting model-derived hypothesis. The model predicted that increasing the area fraction of fibrosis decreased regeneration 28 days post injury due to limited growth factor diffusion and impaired cell migration. WT, mdx, and TGFβ-treated mdx mice were used to test this experimentally. TGFβ injections increased the extracellular matrix (ECM) area fraction; however, the passive stiffness of the treated muscle, which was assumed to correlate with ECM protein density, decreased following injections, suggesting that ECM protein density was lower. Further, there was no cross-sectional area (CSA) difference during recovery between the groups. Additional simulations revealed that decreasing the ECM protein density resulted in no difference in CSA, similar to the experiment. These results suggest that increases in ECM area fraction alone are not sufficient to reduce the regenerative capacity of mdx muscle, and that fibrosis is a complex pathological condition requiring further understanding. Topics: Animals; Disease Models, Animal; Extracellular Matrix; Fibrosis; Male; Mice, Inbred C57BL; Mice, Inbred mdx; Models, Biological; Muscle, Skeletal; Muscular Dystrophy, Duchenne; Regeneration; Transforming Growth Factor beta | 2021 |
Potential cross-talk between muscle and tendon in Duchenne muscular dystrophy.
To describe potential signaling (cross-talk) between dystrophic skeletal muscle and tendon in Duchenne muscular dystrophy.. Review of Duchenne muscular dystrophy and associated literature relevant to muscle-tendon cross-talk.. Duchenne muscular dystrophy results from the absence of the protein dystrophin and the associated dystrophin - glycoprotein complex, which are thought to provide both structural support and signaling functions for the muscle fiber. In addition, there are other potential signal pathways that could represent cross-talk between muscle and tendon, particularly at the myotendinous junction. Duchenne muscular dystrophy is characterized by multiple pathophysiologic mechanisms. Herein, we explore three of these: (1) the extracellular matrix, fibrosis, and fat deposition; (2) satellite cells; and (3) tensegrity. A key signaling protein that emerged in each was transforming growth factor - beta one (TGF-β1).]. Topics: Dystrophin; Humans; Muscle, Skeletal; Muscular Dystrophy, Duchenne; Tendons; Transforming Growth Factor beta | 2021 |
Muscle-specific deletion of SLK/Stk2 enhances p38 activity and myogenesis in mdx mice.
Duchenne's muscular dystrophy (DMD) is a severe muscle wasting disorder characterized by the loss of dystrophin expression, muscle necrosis, inflammation and fibrosis. Ongoing muscle regeneration is impaired by persistent cytokine stress, further decreasing muscle function. Patients with DMD rarely survive beyond their early 20s, with cardiac and respiratory dysfunction being the primary cause of death. Despite an increase in our understanding of disease progression as well as promising preclinical animal models for therapeutic intervention, treatment options for muscular dystrophy remain limited and novel therapeutic targets are required. Many reports suggest that the TGFβ signalling pathway is activated in dystrophic muscle and contributes to the pathology of DMD in part by impairing the differentiation of myoblasts into mature myofibers. Here, we show that in vitro knockdown of the Ste20-like kinase, SLK, can partially restore myoblast differentiation downstream of TGFβ in a Smad2/3 independent manner. In an mdx model, we demonstrate that SLK is expressed at high levels in regenerating myofibers. Muscle-specific deletion of SLK reduced leukocyte infiltration, increased myogenin and utrophin expression and enhanced differentiation. This was accompanied by resistance to eccentric contraction-induced injury in slow fiber type-enriched soleus muscles. Finally, we found that these effects were partially dependent on the upregulation of p38 signalling. Collectively, these results demonstrate that SLK downregulation can restore some aspects of disease progression in DMD. Topics: Animals; Cells, Cultured; Disease Models, Animal; Dogs; Gene Knockout Techniques; MAP Kinase Signaling System; Mice; Mice, Inbred mdx; Mice, Knockout; Muscle Development; Muscle, Skeletal; Muscular Dystrophy, Duchenne; Myoblasts; Myogenin; p38 Mitogen-Activated Protein Kinases; Protein Serine-Threonine Kinases; Transforming Growth Factor beta | 2021 |
Photobiomodulation Therapy for Attenuating the Dystrophic Phenotype of Mdx Mice.
This study analyzed photobiomodulation therapy (PBMT) effects on regenerative, antioxidative, anti-inflammatory and angiogenic markers in the dystrophic skeletal muscle of mdx mice, the experimental model of Duchenne muscular dystrophy (DMD), during the acute phase of dystrophy disease. The following groups were set up: Ctrl (control group of normal wild-type mice; C57BL/10); mdx (untreated mdx mice); mdxPred (mdx mice treated with prednisolone) and mdxLA (mdx mice treated with PBMT). The PBMT was carried out using an Aluminum Gallium Arsenide (AIGaAs; IBRAMED® laserpulse) diode, 830 nm wavelength, applied on the dystrophic quadriceps muscle. The mdxLA group showed a degenerative and regenerative area reduction simultaneously with a MyoD level increase, ROS production and inflammatory marker reduction and up-regulation in the VEGF factor. In addition, PBMT presented similar effects to prednisolone treatment in most of the parameters analyzed. In conclusion, our results indicate that PBMT in the parameters selected attenuated the dystrophic phenotype of mdx mice, improving skeletal muscle regeneration; reducing the oxidative stress and inflammatory process; and up-regulating the angiogenic marker. Topics: Animals; Low-Level Light Therapy; Male; Mice; Mice, Inbred C57BL; Mice, Inbred mdx; Muscular Dystrophy, Animal; Muscular Dystrophy, Duchenne; Phenotype; Transforming Growth Factor beta; Vascular Endothelial Growth Factor A | 2020 |
TGF-β-driven muscle degeneration and failed regeneration underlie disease onset in a DMD mouse model.
Duchenne muscular dystrophy (DMD) is a chronic muscle disease characterized by poor myogenesis and replacement of muscle by extracellular matrix. Despite the shared genetic basis, severity of these deficits varies among patients. One source of these variations is the genetic modifier that leads to increased TGF-β activity. While anti-TGF-β therapies are being developed to target muscle fibrosis, their effect on the myogenic deficit is underexplored. Our analysis of in vivo myogenesis in mild (C57BL/10ScSn-mdx/J and C57BL/6J-mdxΔ52) and severe DBA/2J-mdx (D2-mdx) dystrophic models reveals no defects in developmental myogenesis in these mice. However, muscle damage at the onset of disease pathology, or by experimental injury, drives up TGF-β activity in the severe, but not in the mild, dystrophic models. Increased TGF-β activity is accompanied by increased accumulation of fibroadipogenic progenitors (FAPs) leading to fibro-calcification of muscle, together with failure of regenerative myogenesis. Inhibition of TGF-β signaling reduces muscle degeneration by blocking FAP accumulation without rescuing regenerative myogenesis. These findings provide in vivo evidence of early-stage deficit in regenerative myogenesis in D2-mdx mice and implicates TGF-β as a major component of a pathogenic positive feedback loop in this model, identifying this feedback loop as a therapeutic target. Topics: Animals; Disease Models, Animal; Mice; Mice, Inbred C57BL; Mice, Inbred mdx; Muscle Development; Muscle, Skeletal; Muscular Dystrophy, Duchenne; Regeneration; Transforming Growth Factor beta | 2020 |
Acute conversion of patient-derived Duchenne muscular dystrophy iPSC into myotubes reveals constitutive and inducible over-activation of TGFβ-dependent pro-fibrotic signaling.
In Duchenne muscular dystrophy (DMD), DYSTROPHIN deficiency exposes myofibers to repeated cycles of contraction/degeneration, ultimately leading to muscle loss and replacement by fibrotic tissue. DMD pathology is typically exacerbated by excessive secretion of TGFβ and consequent accumulation of pro-fibrotic components of the extra-cellular matrix (ECM), which in turn impairs compensatory regeneration and complicates the efficacy of therapeutic strategies. It is currently unclear whether DMD skeletal muscle fibers directly contribute to excessive activation of TGFβ. Development of skeletal myofibers from DMD patient-derived induced pluripotent stem cells (iPSC), as an "in dish" model of disease, can be exploited to determine the myofiber contribution to pathogenic TGFβ signaling in DMD and might provide a screening platform for the identification of anti-fibrotic interventions in DMD.. We describe a rapid and efficient method for the generation of contractile human skeletal muscle cells from DMD patient-derived hiPSC, based on the inducible expression of MyoD and BAF60C (encoded by SMARCD3 gene), using an enhanced version of piggyBac (epB) transposone vectors. DMD iPSC-derived myotubes were tested as an "in dish" disease model and exposed to environmental and mechanical cues that recapitulate salient pathological features of DMD.. We show that DMD iPSC-derived myotubes exhibit a constitutive activation of TGFβ-SMAD2/3 signaling. High-content screening (HCS)-based quantification of nuclear phosphorylated SMAD2/3 signal revealed that DMD iPSC-derived myotubes also exhibit increased activation of the TGFβ-SMAD2/3 signaling following exposure to either recombinant TGFβ or electrical pacing-induced contraction.. Acute conversion of DMD patient-derived iPSC into skeletal muscles, by the ectopic expression of MyoD and BAF60C, provides a rapid and reliable protocol for an "in dish" DMD model that recapitulates key pathogenic features of disease pathology, such as the constitutive activation of the TGFβ/SMAD signaling as well as the deregulated response to pathogenic stimuli, e.g., ECM-derived signals or mechanical cues. Thus, this model is suitable for the identification of new therapeutic targets in DMD patient-specific muscles. Topics: Cell Differentiation; Cell Line; Cells, Cultured; Chromosomal Proteins, Non-Histone; Fibrosis; Humans; Induced Pluripotent Stem Cells; Muscle Fibers, Skeletal; Muscular Dystrophy, Duchenne; MyoD Protein; Primary Cell Culture; Signal Transduction; Smad Proteins; Transforming Growth Factor beta | 2020 |
Spp1 (osteopontin) promotes TGFβ processing in fibroblasts of dystrophin-deficient muscles through matrix metalloproteinases.
Duchenne muscular dystrophy (DMD) is caused by mutations in the gene encoding dystrophin. Prior work has shown that DMD progression can vary, depending on the genetic makeup of the patient. Several modifier alleles have been identified including LTBP4 and SPP1. We previously showed that Spp1 exacerbates the DMD phenotype in the mdx mouse model by promoting fibrosis and by skewing macrophage polarization. Here, we studied the mechanisms involved in Spp1's promotion of fibrosis by using both isolated fibroblasts and genetically modified mice. We found that Spp1 upregulates collagen expression in mdx fibroblasts by enhancing TGFβ signaling. Spp1's effects on TGFβ signaling are through induction of MMP9 expression. MMP9 is a protease that can release active TGFβ ligand from its latent complex. In support for activation of this pathway in our model, we showed that treatment of mdx fibroblasts with MMP9 inhibitor led to accumulation of the TGFβ latent complex, decreased levels of active TGFβ and reduced collagen expression. Correspondingly, we found reduced active TGFβ in Spp1-/-mdxB10 and Mmp9-/-mdxB10 muscles in vivo. Taken together with previous observations of reduced fibrosis in both models, these data suggest that Spp1 acts upstream of TGFβ to promote fibrosis in mdx muscles. We found that in the context of constitutively upregulated TGFβ signaling (such as in the mdxD2 model), ablation of Spp1 has very little effect on fibrosis. Finally, we performed proof-of-concept studies showing that postnatal pharmacological inhibition of Spp1 reduces fibrosis and improves muscle function in mdx mice. Topics: Animals; Collagen Type I; Disease Models, Animal; Dystrophin; Extracellular Matrix; Female; Fibroblasts; Fibrosis; Male; Matrix Metalloproteinase 9; Mice; Mice, Inbred mdx; Mice, Knockout; Muscle, Skeletal; Muscular Dystrophy, Duchenne; Osteopontin; Primary Cell Culture; Regeneration; Signal Transduction; Transforming Growth Factor beta | 2019 |
Liposomal steroid nano-drug is superior to steroids as-is in mdx mouse model of Duchenne muscular dystrophy.
Glucocorticosteroids are the most efficacious anti-inflammatory agents and the gold standard treatment in Duchenne muscular dystrophy (DMD). However, their chronic use may lead to severe side effects. We evaluated the use of a novel injectable steroidal nano-drug in mdx mouse model of DMD by comparing the efficacy of nano-liposomes remotely loaded with the steroid prodrug, methylprednisolone hemisuccinate (MPS) with the same steroid as-is, in short (4-weeks) and long-term (58-weeks) treatments. Liposomal-MPS was selectively targeted to the mouse diaphragm, the most dystrophic muscle at early stage of the disease. The bioactivity of the steroidal nano-drug was evidenced by a significant decreased serum TGF-β and reduced diaphragm macrophage infiltration after short-term treatment. In the long-term, the treatment with liposomal-MPS not only demonstrated improved muscle strength and mobility it also induced lower tibia and lumbar vertebrae osteoporosis indicating much lower bone related adverse effects. Topics: Animals; Creatine Kinase; Disease Models, Animal; Immunohistochemistry; Inflammation; Liposomes; Male; Mice; Mice, Inbred mdx; Muscle Strength; Muscular Dystrophy, Duchenne; Steroids; Transforming Growth Factor beta | 2019 |
ERBB3 and NGFR mark a distinct skeletal muscle progenitor cell in human development and hPSCs.
Human pluripotent stem cells (hPSCs) can be directed to differentiate into skeletal muscle progenitor cells (SMPCs). However, the myogenicity of hPSC-SMPCs relative to human fetal or adult satellite cells remains unclear. We observed that hPSC-SMPCs derived by directed differentiation are less functional in vitro and in vivo compared to human satellite cells. Using RNA sequencing, we found that the cell surface receptors ERBB3 and NGFR demarcate myogenic populations, including PAX7 progenitors in human fetal development and hPSC-SMPCs. We demonstrated that hPSC skeletal muscle is immature, but inhibition of transforming growth factor-β signalling during differentiation improved fusion efficiency, ultrastructural organization and the expression of adult myosins. This enrichment and maturation strategy restored dystrophin in hundreds of dystrophin-deficient myofibres after engraftment of CRISPR-Cas9-corrected Duchenne muscular dystrophy human induced pluripotent stem cell-SMPCs. The work provides an in-depth characterization of human myogenesis, and identifies candidates that improve the in vivo myogenic potential of hPSC-SMPCs to levels that are equal to directly isolated human fetal muscle cells. Topics: Adult; Aged; Cell Differentiation; CRISPR-Cas Systems; Dystrophin; Female; Gene Editing; Gene Expression Regulation, Developmental; Humans; Induced Pluripotent Stem Cells; Male; Middle Aged; Muscle Development; Muscle Fibers, Skeletal; Muscular Dystrophy, Duchenne; Myoblasts; Myosins; Nerve Tissue Proteins; PAX7 Transcription Factor; Receptor, ErbB-3; Receptors, Nerve Growth Factor; Signal Transduction; Transforming Growth Factor beta | 2018 |
Identification of plasma interleukins as biomarkers for deflazacort and omega-3 based Duchenne muscular dystrophy therapy.
Duchenne muscular dystrophy (DMD) is a progressive and fatal disease, characterized by the absence of dystrophin, muscle degeneration and cardiorespiratory failure. Creatine kinase is the classic marker to screen for DMD. However, other markers are needed to follow disease progression and to evaluate the response to therapy over longer periods. In the present study, we aim to identify interleukins in the plasma of the mdx mice model of DMD that could serve as biomarkers to monitor dystrophy progression, at distinct stages of the disease (1, 3 and 8 months of age). We used deflazacort and omega-3 therapies to validate the biomarkers studied. Plasma levels of TNF-α and TGF-β were increased in mdx mice in relation to control, at all times studied. Differences in IFN-γ and IL-10 contents, comparing mdx x CTRL, were detected only at the early stage (1 month). IL-6 decreased at 3 and 8 months and IL-13 increased at 8 months in the mdx compared to control. Deflazacort and omega-3 reduced the plasma levels of the pro-inflammatory (TNF-α, INF-γ, IL-6) and pro-fibrotic (IL-13 and TGF-β) interleukins and increased the plasma levels of IL-10. It is suggested that TNF-α and TGF-β in plasma would be the best markers to follow disease progression. IL-6, INF-γ and IL-10 would be suitable markers to the earlier stages of dystrophy and IL-13 a suitable marker to the later stages of dystrophy. Topics: Animals; Anti-Inflammatory Agents; Biomarkers; Disease Progression; Fatty Acids, Omega-3; Interferon-gamma; Interleukins; Mice; Muscular Dystrophy, Duchenne; Pregnenediones; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha | 2018 |
Placenta-derived mesenchymal stromal cells and their exosomes exert therapeutic effects in Duchenne muscular dystrophy.
Duchenne muscular dystrophy (DMD) is a degenerative lethal, X-linked disease of skeletal and cardiac muscles caused by mutations in the dystrophin gene. Cell therapy using different cell types, including mesenchymal stromal cells (MSCs), has been considered as a potential approach for the treatment of DMD. MSCs can be obtained from autologous sources such as bone marrow and adipose tissues or from allogeneic placenta and umbilical cord. The safety and therapeutic impact of these cells has been demonstrated in pre-clinical and clinical studies and their functions are attributed to paracrine effects that are mediated by secreted cytokines and extracellular vesicles. Here, we studied the therapeutic effects of placenta-derived MSCs (PL-MSCs) and their secreted exosomes using mouse and human myoblasts from healthy controls, Duchenne patients and mdx mice. Treatment of myoblasts with conditioned medium or exosomes secreted by PL-MSCs increased the differentiation of these cells and decreased the expression of fibrogenic genes in DMD patient myoblasts. In addition, these treatments also increased the expression of utrophin in these cells. Using a quantitative miR-29c reporter, we demonstrated that the PL-MSC effects were partly mediated by the transfer of exosomal miR-29c. Intramuscular transplantation of PL-MSCs in mdx mice resulted in decreased creatine kinase levels. PL-MSCs significantly decreased the expression of TGF-β and the level of fibrosis in the diaphragm and cardiac muscles, inhibited inflammation and increased utrophin expression. In vivo imaging analyses using MSCs labeled with gold nanoparticles or fluorescent dyes demonstrated localization of the cells in the muscle tissues up to 3 weeks post treatment. Altogether, these results demonstrate that PL-MSCs and their secreted exosomes have important clinical applications in cell therapy of DMD partly via the targeted delivery of exosomal miR-29c. Topics: Adipose Tissue; Animals; Cell Differentiation; Culture Media, Conditioned; Dystrophin; Exosomes; Extracellular Vesicles; Female; Fluorescent Dyes; Gene Expression Regulation; Gold; Humans; Mesenchymal Stem Cell Transplantation; Mesenchymal Stem Cells; Metal Nanoparticles; Mice, Inbred mdx; MicroRNAs; Muscular Dystrophy, Duchenne; Myoblasts; Placenta; Pregnancy; Transfection; Transforming Growth Factor beta; Umbilical Cord; Utrophin | 2018 |
Pharmacological Inhibition of PKCθ Counteracts Muscle Disease in a Mouse Model of Duchenne Muscular Dystrophy.
Inflammation plays a considerable role in the progression of Duchenne Muscular Dystrophy (DMD), a severe muscle disease caused by a mutation in the dystrophin gene. We previously showed that genetic ablation of Protein Kinase C θ (PKCθ) in mdx, the mouse model of DMD, improves muscle healing and regeneration, preventing massive inflammation. To establish whether pharmacological targeting of PKCθ in DMD can be proposed as a therapeutic option, in this study we treated young mdx mice with the PKCθ inhibitor Compound 20 (C20). We show that C20 treatment led to a significant reduction in muscle damage associated with reduced immune cells infiltration, reduced inflammatory pathways activation, and maintained muscle regeneration. Importantly, C20 treatment is efficient in recovering muscle performance in mdx mice, by preserving muscle integrity. Together, these results provide proof of principle that pharmacological inhibition of PKCθ in DMD can be considered an attractive strategy to modulate immune response and prevent the progression of the disease.. Duchenne muscular dystrophy (DMD) is a severe muscle disease affecting 1:3500 male births. DMD is caused by a mutation in dystrophin gene, coding for a protein required for skeletal and cardiac muscle integrity. Lack of a functional dystrophin is primarily responsible for the muscle eccentric contraction-induced muscle damage, observed in dystrophic muscle. However, inflammation plays a considerable role in the progression of DMD. Glucocorticoids, which have anti-inflammatory properties, are being used to treat DMD with some success; however, long term treatment with these drugs induces muscle atrophy and wasting, outweighing their benefit. The identification of specific targets for anti-inflammatory therapies is one of the ongoing therapeutic options. Although blunting inflammation would not be a "cure" for the disease, the emerging clue is that multiple strategies, addressing different aspects of the pathology, which may eventually converge, may be successful. In this context, we previously showed that genetic ablation of Protein Kinase C θ (PKCθ), an enzyme known to be involved in immune response, in mdx, the mouse model of DMD, improves muscle healing and regeneration, preventing massive inflammation. To establish whether pharmacological targeting of PKCθ in DMD can be proposed as a therapeutic option, in this study we treated young mdx mice with the PKCθ inhibitor Compound 20 (C20). We show that C20 treatment led to a significant reduction in muscle damage associated with reduced immune cells infiltration, reduced inflammatory pathways activation, and maintained muscle regeneration. Importantly, C20 treatment is efficient in recovering muscle performance in mdx mice, by preserving muscle integrity. Together, these results provide proof of principle that pharmacological inhibition of PKCθ in DMD can be considered an attractive strategy to modulate immune response and prevent the progression of the disease. Topics: Animals; Blotting, Western; Dipeptides; Disease Models, Animal; Gene Expression; Humans; Inflammation; Isoenzymes; Male; Mice, Inbred C57BL; Mice, Inbred mdx; Mice, Knockout; Microscopy, Fluorescence; Motor Activity; Muscle, Skeletal; Muscular Dystrophy, Animal; Muscular Dystrophy, Duchenne; Myocardium; Protein Kinase C; Protein Kinase C-theta; Regeneration; Reverse Transcriptase Polymerase Chain Reaction; Transforming Growth Factor beta | 2017 |
Overexpression of Latent TGFβ Binding Protein 4 in Muscle Ameliorates Muscular Dystrophy through Myostatin and TGFβ.
Latent TGFβ binding proteins (LTBPs) regulate the extracellular availability of latent TGFβ. LTBP4 was identified as a genetic modifier of muscular dystrophy in mice and humans. An in-frame insertion polymorphism in the murine Ltbp4 gene associates with partial protection against muscular dystrophy. In humans, nonsynonymous single nucleotide polymorphisms in LTBP4 associate with prolonged ambulation in Duchenne muscular dystrophy. To better understand LTBP4 and its role in modifying muscular dystrophy, we created transgenic mice overexpressing the protective murine allele of LTBP4 specifically in mature myofibers using the human skeletal actin promoter. Overexpression of LTBP4 protein was associated with increased muscle mass and proportionally increased strength compared to age-matched controls. In order to assess the effects of LTBP4 in muscular dystrophy, LTBP4 overexpressing mice were bred to mdx mice, a model of Duchenne muscular dystrophy. In this model, increased LTBP4 led to greater muscle mass with proportionally increased strength, and decreased fibrosis. The increase in muscle mass and reduction in fibrosis were similar to what occurs when myostatin, a related TGFβ family member and negative regulator of muscle mass, was deleted in mdx mice. Supporting this, we found that myostatin forms a complex with LTBP4 and that overexpression of LTBP4 led to a decrease in myostatin levels. LTBP4 also interacted with TGFβ and GDF11, a protein highly related to myostatin. These data identify LTBP4 as a multi-TGFβ family ligand binding protein with the capacity to modify muscle disease through overexpression. Topics: Animals; Bone Morphogenetic Proteins; Disease Models, Animal; Gene Expression Regulation; Growth Differentiation Factors; Humans; Latent TGF-beta Binding Proteins; Mice; Mice, Inbred mdx; Mice, Transgenic; Muscle, Skeletal; Muscular Dystrophy, Duchenne; Myostatin; Naphthols; Transforming Growth Factor beta; Triazines | 2016 |
Association Study of Exon Variants in the NF-κB and TGFβ Pathways Identifies CD40 as a Modifier of Duchenne Muscular Dystrophy.
The expressivity of Mendelian diseases can be influenced by factors independent from the pathogenic mutation: in Duchenne muscular dystrophy (DMD), for instance, age at loss of ambulation (LoA) varies between individuals whose DMD mutations all abolish dystrophin expression. This suggests the existence of trans-acting variants in modifier genes. Common single nucleotide polymorphisms (SNPs) in candidate genes (SPP1, encoding osteopontin, and LTBP4, encoding latent transforming growth factor β [TGFβ]-binding protein 4) have been established as DMD modifiers. We performed a genome-wide association study of age at LoA in a sub-cohort of European or European American ancestry (n = 109) from the Cooperative International Research Group Duchenne Natural History Study (CINRG-DNHS). We focused on protein-altering variants (Exome Chip) and included glucocorticoid treatment as a covariate. As expected, due to the small population size, no SNPs displayed an exome-wide significant p value (< 1.8 × 10 Topics: Adolescent; Alleles; Case-Control Studies; CD40 Antigens; Child; Dystrophin; Exons; Genes, Modifier; Genome-Wide Association Study; Glucocorticoids; Humans; Latent TGF-beta Binding Proteins; Muscular Dystrophy, Duchenne; Mutation; NF-kappa B; Osteopontin; Polymorphism, Single Nucleotide; Transforming Growth Factor beta; White People | 2016 |
Fibrogenic Cell Plasticity Blunts Tissue Regeneration and Aggravates Muscular Dystrophy.
Preservation of cell identity is necessary for homeostasis of most adult tissues. This process is challenged every time a tissue undergoes regeneration after stress or injury. In the lethal Duchenne muscular dystrophy (DMD), skeletal muscle regenerative capacity declines gradually as fibrosis increases. Using genetically engineered tracing mice, we demonstrate that, in dystrophic muscle, specialized cells of muscular, endothelial, and hematopoietic origins gain plasticity toward a fibrogenic fate via a TGFβ-mediated pathway. This results in loss of cellular identity and normal function, with deleterious consequences for regeneration. Furthermore, this fibrogenic process involves acquisition of a mesenchymal progenitor multipotent status, illustrating a link between fibrogenesis and gain of progenitor cell functions. As this plasticity also was observed in DMD patients, we propose that mesenchymal transitions impair regeneration and worsen diseases with a fibrotic component. Topics: Animals; Antigens, CD; Cell Plasticity; Cells, Cultured; Endothelial Cells; Fibrosis; Integrin alpha Chains; Mice; Mice, Inbred C57BL; Mice, Inbred DBA; Mice, Transgenic; Muscle, Skeletal; Muscular Dystrophy, Duchenne; Myoblasts; Platelet Endothelial Cell Adhesion Molecule-1; Real-Time Polymerase Chain Reaction; Receptor, Platelet-Derived Growth Factor alpha; Regeneration; Satellite Cells, Skeletal Muscle; Signal Transduction; Smad2 Protein; Smad3 Protein; Transforming Growth Factor beta | 2015 |
Chronic effects of losartan on the muscles and the serologic profiles of mdx mice.
Losartan, an angiotensin II type 1 receptor blocker, attenuates transforming growth factor-β (TGF-β) signaling, which inhibits myogenic regeneration. Although many researchers have demonstrated that losartan has anti-fibrotic and protective effects on cardiac and skeletal muscles, for long-term administration to treat dystrophic disorders, it is essential to demonstrate not only the therapeutic effects of losartan on muscles but also its effects on other organs and on blood biochemistry.. Mdx mice, an animal model of Duchenne muscular dystrophy (DMD), were fed losartan dissolved in tap water. After 44weeks, the skeletal (gastrocnemius), cardiac, and diaphragm muscles of mdx mice were removed. Tissue and blood samples were collected from all experimental animals. Effects of losartan on muscle regeneration, fibrosis, and blood enzymatic profiles were evaluated.. In histopathological findings and serum biochemistry analyses, chronic losartan administration showed muscular protective effects and inhibited fibrosis in skeletal (gastrocnemius), cardiac, and diaphragmatic muscles. In addition, losartan had no effects on other solid organs. Interestingly, losartan had beneficial effects on serum HDL ratio.. This study demonstrates the therapeutic effects of losartan on muscles and its effects on other organs and on blood biochemistry. In conclusion, our results provide useful information for consideration of chronic losartan administration be as a treatment of DMD. Topics: Administration, Oral; Angiotensin II Type 1 Receptor Blockers; Animals; Biomarkers; Losartan; Male; Mice; Mice, Inbred C57BL; Mice, Inbred mdx; Muscle, Skeletal; Muscular Dystrophy, Duchenne; Transforming Growth Factor beta; Treatment Outcome | 2015 |
Changes in calsequestrin, TNF-α, TGF-β and MyoD levels during the progression of skeletal muscle dystrophy in mdx mice: a comparative analysis of the quadriceps, diaphragm and intrinsic laryngeal muscles.
In Duchenne muscular dystrophy (DMD), the search for new biomarkers to follow the evolution of the disease is of fundamental importance in the light of the evolving gene and pharmacological therapies. In addition to the lack of dystrophin, secondary events including changes in calcium levels, inflammation and fibrosis greatly contribute to DMD progression and the molecules involved in these events may represent potential biomarkers. In this study, we performed a comparative evaluation of the progression of dystrophy within muscles that are differently affected by dystrophy (diaphragm; DIA and quadriceps; QDR) or spared (intrinsic laryngeal muscles) using the mdx mice model of DMD. We assessed muscle levels of calsequestrin (calcium-related protein), tumour necrosis factor (TNF-α; pro-inflammatory cytokine), tumour growth factor (TGF-β; pro-fibrotic factor) and MyoD (muscle proliferation) vs. histopathology at early (1 and 4 months of age) and late (9 months of age) stages of dystrophy. Fibrosis was the primary feature in the DIA of mdx mice (9 months: 32% fibrosis), which was greater than in the QDR (9 months: 0.6% fibrosis). Muscle regeneration was the primary feature in the QDR (9 months: 90% of centrally nucleated fibres areas vs. 33% in the DIA). The QDR expressed higher levels of calsequestrin than the DIA. Laryngeal muscles showed normal levels of TNF-α, TGF-β and MyoD. A positive correlation between histopathology and cytokine levels was observed only in the diaphragm, suggesting that TNF-α and TGF-β serve as markers of dystrophy primarily for the diaphragm. Topics: Animals; Biomarkers; Blotting, Western; Calsequestrin; Diaphragm; Disease Models, Animal; Disease Progression; Female; Fluorescent Antibody Technique; Laryngeal Muscles; Male; Mice; Mice, Inbred mdx; Muscular Dystrophy, Duchenne; MyoD Protein; Quadriceps Muscle; Transforming Growth Factor beta; Tumor Necrosis Factor-alpha | 2015 |
Restoration of muscle strength in dystrophic muscle by angiotensin-1-7 through inhibition of TGF-β signalling.
Duchenne muscular dystrophy (DMD) is the most common inherited neuromuscular disease, and is characterized by the lack of dystrophin, muscle wasting, increased transforming growth factor (TGF)-β Smad-dependent signalling and fibrosis. Acting via the Mas receptor, angiotensin-1-7 [Ang-(1-7)], is part of the renin-angiotensin system, with the opposite effect to that of angiotensin II. We hypothesized that the Ang-(1-7)/Mas receptor axis might protect chronically damaged tissues as in skeletal muscle of the DMD mouse model mdx. Infusion or oral administration of Ang-(1-7) in mdx mice normalized skeletal muscle architecture, decreased local fibrosis and improved muscle function in vitro and in vivo. These positive effects were mediated by the inhibition of TGF-β Smad signalling, which in turn led to reduction of the pro-fibrotic microRNA miR-21 concomitant with a reduction in the number of TCF4 expressing fibroblasts. Mdx mice infused with Mas antagonist (A-779) and mdx deficient for the Mas receptor showed highly deteriorated muscular architecture, increased fibrosis and TGF-β signalling with diminished muscle strength. These results suggest that this novel compound Ang-(1-7) might be used to improve quality of life and delay death in individuals with DMD and this drug should be investigated in further pre-clinical trials. Topics: Angiotensin I; Animals; Disease Models, Animal; Extracellular Matrix; Fibroblasts; Fibrosis; Humans; Male; Mice; Mice, Inbred mdx; Mice, Knockout; MicroRNAs; Muscle Strength; Muscle, Skeletal; Muscular Dystrophy, Duchenne; Peptide Fragments; Receptors, Cell Surface; Signal Transduction; Transforming Growth Factor beta | 2014 |
Asynchronous remodeling is a driver of failed regeneration in Duchenne muscular dystrophy.
We sought to determine the mechanisms underlying failure of muscle regeneration that is observed in dystrophic muscle through hypothesis generation using muscle profiling data (human dystrophy and murine regeneration). We found that transforming growth factor β-centered networks strongly associated with pathological fibrosis and failed regeneration were also induced during normal regeneration but at distinct time points. We hypothesized that asynchronously regenerating microenvironments are an underlying driver of fibrosis and failed regeneration. We validated this hypothesis using an experimental model of focal asynchronous bouts of muscle regeneration in wild-type (WT) mice. A chronic inflammatory state and reduced mitochondrial oxidative capacity are observed in bouts separated by 4 d, whereas a chronic profibrotic state was seen in bouts separated by 10 d. Treatment of asynchronously remodeling WT muscle with either prednisone or VBP15 mitigated the molecular phenotype. Our asynchronous regeneration model for pathological fibrosis and muscle wasting in the muscular dystrophies is likely generalizable to tissue failure in chronic inflammatory states in other regenerative tissues. Topics: Animals; Anti-Inflammatory Agents; Cell Differentiation; Cells, Cultured; Dystrophin; Fibrosis; Humans; Inflammation; Mice; Mice, Transgenic; Mitochondria; Muscle, Skeletal; Muscular Dystrophies, Limb-Girdle; Muscular Dystrophy, Animal; Muscular Dystrophy, Duchenne; Muscular Dystrophy, Emery-Dreifuss; Prednisone; Pregnadienediols; Protein Interaction Mapping; Protein Interaction Maps; Protein Structure, Tertiary; Regeneration; RNA, Messenger; Stem Cells; Transforming Growth Factor beta | 2014 |
Suramin attenuates dystrophin-deficient cardiomyopathy in the mdx mouse model of duchenne muscular dystrophy.
The purpose of this study was to determine the effects of suramin, an antifibrotic agent, on cardiac function and remodeling in mdx mice.. mdx mice (8 months old) received intraperitoneal injections of suramin twice a week for 3 months. Control mdx mice (8 months old) were injected with saline.. Suramin improved the electrocardiography profile with the main corrections seen in S- to R-wave ratio, PR interval, and Q amplitude, and a significant decrease in the cardiomyopathy index. Suramin decreased myocardial fibrosis, inflammation, and myonecrosis.. These findings suggest that suramin may be a new adjunctive therapy to help improve cardiomyopathy in DMD. Topics: Age Factors; Analysis of Variance; Animals; Antineoplastic Agents; Cardiomyopathies; Creatine Kinase; Disease Models, Animal; Dystrophin; Electrocardiography; Electroencephalography; Female; Male; Mice; Mice, Inbred C57BL; Mice, Inbred mdx; Muscle Fibers, Skeletal; Muscular Dystrophy, Duchenne; Suramin; Transforming Growth Factor beta | 2013 |
TGFBR2 but not SPP1 genotype modulates osteopontin expression in Duchenne muscular dystrophy muscle.
A polymorphism (rs28357094) in the promoter region of the SPP1 gene coding for osteopontin (OPN) is a strong determinant of disease severity in Duchenne muscular dystrophy (DMD). The rare G allele of rs28357094 alters gene promoter function and reduces mRNA expression in transfected HeLa cells. To dissect the molecular mechanisms of increased disease severity associated with the G allele, we characterized SPP1 mRNA and protein in DMD muscle biopsies of patients with defined rs28357094 genotype. We did not find significant differences in osteopontin mRNA or protein expression between patients carrying the T (ancestral allele) or TG/GG genotypes at rs28357094. The G allele was significantly associated with reduced CD4(+) and CD68(+) cells on patient muscle biopsy. We also quantified transforming growth factor-β (TGFB) and TGFB receptor-2 (TGFBR2) mRNA in DMD muscle biopsies, given the ability of TGFB and TGFBR2 to activate SPP1 promoter region and their role in DMD pathogenesis. The amount of TGFB and TGFBR2 mRNA did not predict the amount of SPP1 mRNA or protein, while a polymorphism in the TGFBR2 gene (rs4522809) was found to be a strong predictor of SPP1 mRNA level. Our findings suggest that OPN mediates inflammatory changes in DMD and that TGFB signalling has a role in the complex regulation of osteopontin expression. Topics: Blotting, Western; Child; Child, Preschool; Cohort Studies; Female; Gene Expression Regulation; Genetic Predisposition to Disease; Genotype; Humans; Infant; Male; Muscle, Skeletal; Muscular Dystrophy, Duchenne; Osteopontin; Polymorphism, Genetic; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; RNA, Messenger; Signal Transduction; Transforming Growth Factor beta | 2012 |
Loss of miR-29 in myoblasts contributes to dystrophic muscle pathogenesis.
microRNAs (miRNAs) are noncoding RNAs that regulate gene expression in post-transcriptional fashion, and emerging studies support their importance in a multitude of physiological and pathological processes. Here, we describe the regulation and function of miR-29 in Duchenne muscular dystrophy (DMD) and its potential use as therapeutic target. Our results demonstrate that miR-29 expression is downregulated in dystrophic muscles of mdx mice, a model of DMD. Restoration of its expression by intramuscular and intravenous injection improved dystrophy pathology by both promoting regeneration and inhibiting fibrogenesis. Mechanistic studies revealed that loss of miR-29 in muscle precursor cells (myoblasts) promotes their transdifferentiation into myofibroblasts through targeting extracellular molecules including collagens and microfibrillar-associated protein 5 (Mfap5). We further demonstrated that miR-29 is under negative regulation by transforming growth factor-β (TGF-β) signaling. Together, these results not only identify TGF-β-miR-29 as a novel regulatory axis during myoblasts conversion into myofibroblasts which constitutes a novel contributing route to muscle fibrogenesis of DMD but also implicate miR-29 replacement therapy as a promising treatment approach for DMD. Topics: Animals; Cell Differentiation; Cell Transdifferentiation; Cells, Cultured; Down-Regulation; Extracellular Matrix; Fibrosis; Gene Expression Regulation; Mice; Mice, Inbred mdx; MicroRNAs; Models, Biological; Muscle, Skeletal; Muscular Dystrophy, Duchenne; Myoblasts; Myofibroblasts; Regeneration; Signal Transduction; Transforming Growth Factor beta | 2012 |
Losartan decreases cardiac muscle fibrosis and improves cardiac function in dystrophin-deficient mdx mice.
Recent studies showed that chronic administration of losartan, an angiotensin II type I receptor antagonist, improved skeletal muscle function in dystrophin-deficient mdx mice. In this study, C57BL/10ScSn-Dmd(mdx)/J female mice were either untreated or treated with losartan (n = 15) in the drinking water at a dose of 600 mg/L over a 6-month period. Cardiac function was assessed via in vivo high frequency echocardiography and skeletal muscle function was assessed using grip strength testing, Digiscan monitoring, Rotarod timing, and in vitro force testing. Fibrosis was assessed using picrosirius red staining and Image J analysis. Gene expression was evaluated using real-time polymerized chain reaction (RT-PCR). Percentage shortening fraction was significantly decreased in untreated (26.9% ± 3.5%) mice compared to losartan-treated (32.2% ± 4.2%; P < .01) mice. Systolic blood pressure was significantly reduced in losartan-treated mice (56 ± 6 vs 69 ± 7 mm Hg; P < .0005). Percentage cardiac fibrosis was significantly reduced in losartan-treated hearts (P < .05) along with diaphragm (P < .01), extensor digitorum longus (P < .05), and gastrocnemius (P < .05) muscles compared to untreated mdx mice. There were no significant differences in skeletal muscle function between treated and untreated groups. Chronic treatment with losartan decreases cardiac and skeletal muscle fibrosis and improves cardiac systolic function in dystrophin-deficient mdx mice. Topics: Angiotensin II Type 1 Receptor Blockers; Animals; Blood Pressure; Cardiomyopathies; Cell Adhesion Molecules; Dystrophin; Female; Fibrosis; Gene Expression Regulation; Heart; Losartan; Mice; Mice, Inbred mdx; Muscle Weakness; Muscle, Skeletal; Muscular Dystrophy, Duchenne; Myocardium; RNA, Messenger; Thrombospondin 1; Transforming Growth Factor beta | 2011 |
Timeline of cardiac dystrophy in 3-18-month-old MDX mice.
The dystrophin-deficient (mdx) mouse remains the most commonly used model for Duchenne muscular dystrophy (DMD). Mdx mice show a predominantly covert cardiomyopathy, the hallmark of which is fibrosis. We compared mdx and normal mice at six ages (3, 6, 9, 12, 15, and 18 months) using in vivo assessment of cardiac function, selective collagen staining, and measures of TGF-β mRNA, Evans blue dye infiltration, macrophage infiltration, and aortic wall thickness. Clear temporal progression was demonstrated, including early fragility of cardiomyocyte membranes, which has an unrelated impact on cardiac function but is associated with macrophage infiltration and fibrosis. Aortic wall thickness is less in older mdx mice. Mdx mice display impaired responses to inotropic challenge from a young age; this is indicative of altered adrenoreceptor function. We draw attention to the paradox of ongoing fibrosis in mdx hearts without a strong molecular signature (in the form of TGF-β mRNA expression). Topics: Aging; Animals; Aorta, Thoracic; Cardiomyopathies; Coloring Agents; Evans Blue; Fibrosis; Macrophages; Male; Mice; Mice, Inbred C57BL; Mice, Inbred mdx; Muscular Dystrophy, Duchenne; Myocardium; Phosphorylation; RNA, Messenger; Smad2 Protein; Time Factors; Transforming Growth Factor beta | 2010 |
Activin IIB receptor blockade attenuates dystrophic pathology in a mouse model of Duchenne muscular dystrophy.
Modulation of transforming growth factor-β (TGF-β) signaling to promote muscle growth holds tremendous promise for the muscular dystrophies and other disorders involving the loss of functional muscle mass. Previous studies have focused on the TGF-β family member myostatin and demonstrated that inhibition of myostatin leads to muscle growth in normal and dystrophic mice. We describe a unique method of systemic inhibition of activin IIB receptor signaling via adeno-associated virus (AAV)-mediated gene transfer of a soluble form of the extracellular domain of the activin IIB receptor to the liver. Treatment of mdx mice with activin IIB receptor blockade led to increased skeletal muscle mass, increased force production in the extensor digitorum longus (EDL), and reduced serum creatine kinase. No effect on heart mass or function was observed. Our results indicate that activin IIB receptor blockade represents a novel and effective therapeutic strategy for the muscular dystrophies. Topics: Activin Receptors, Type II; Animals; Blotting, Western; Cloning, Molecular; Creatine; Creatine Kinase; Dependovirus; Echocardiography; Heart Function Tests; Mice; Mice, Inbred mdx; Mice, Transgenic; Muscle Contraction; Muscle, Skeletal; Muscular Dystrophy, Duchenne; Myostatin; Organ Size; Signal Transduction; Transforming Growth Factor beta | 2010 |
Intramuscular renin-angiotensin system is activated in human muscular dystrophy.
To investigate the role of the muscular renin-angiotensin system (RAS) in human muscular dystrophy, we used immunohistochemistry and Western blotting to examine the cellular localization of angiotensin-converting enzyme (ACE), the angiotensin II type 1 receptor (AT1) and the angiotensin II type 2 receptor (AT2) in muscle biopsies from patients with Duchenne muscular dystrophy (DMD), Becker muscular dystrophy (BMD), and congenital muscular dystrophy (CMD). In normal muscle, ACE was expressed in vascular endothelial cells and neuromuscular junctions (NMJs), whereas AT1 was immunolocalized to the smooth muscle cells of blood vessels and intramuscular nerve twigs. AT2 was immunolocalized in the smooth muscle cells of blood vessels. These findings suggest that the RAS has a functional role in peripheral nerves and NMJs. ACE and AT1, but AT2 immunoreactivity were increased markedly in dystrophic muscle as compared to controls. ACE and the AT1 were strongly expressed in the cytoplasm and nuclei of regenerating muscle fibers, fibroblasts, and in macrophages infiltrating necrotic fibers. Double immunolabeling revealed that activated fibroblasts in the endomysium and perimysium of DMD and CMD muscle were positive for ACE and AT1. Triple immunolabeling demonstrated that transforming growth factor-beta1 (TGF-beta1) and ACE were colocalized on the cytoplasm of activated fibroblasts in dystrophic muscle. Furthermore, Western blotting showed increases in the expression of AT1 and TGF-beta1 protein in dystrophic muscle, which coincided with our immunohistochemical results. The overexpression of ACE and AT1 in dystrophic muscle would likely result in the increased production of Ang II, which may act on these cells in an autocrine manner via AT1. The activation of AT1 may induce fibrous tissue formation through overexpression of TGF-beta1, which potently activates fibrogenesis and suppresses regeneration. In conclusion, our results imply that the intramuscular RAS-TGF-beta1 pathway is activated in human muscular dystrophy and plays a role at least partly in the pathophysiology of this disease. Topics: Blood Vessels; Child; Child, Preschool; Endothelial Cells; Female; Fibroblasts; Humans; Infant; Macrophages; Male; Muscles; Muscular Dystrophies; Muscular Dystrophy, Duchenne; Neuromuscular Junction; Peptidyl-Dipeptidase A; Peripheral Nerves; Receptor, Angiotensin, Type 1; Receptor, Angiotensin, Type 2; Regeneration; Signal Transduction; Transforming Growth Factor beta | 2009 |
AAV-dependent targeting of myostatin function: follistatin strikes back at muscular dystrophy.
Topics: Animals; Dependovirus; Follistatin; Genetic Therapy; Genetic Vectors; Humans; Mice; Mice, Inbred mdx; Muscular Dystrophy, Duchenne; Myostatin; Protein Isoforms; Transduction, Genetic; Transforming Growth Factor beta | 2008 |
Fibrinogen drives dystrophic muscle fibrosis via a TGFbeta/alternative macrophage activation pathway.
In the fatal degenerative Duchenne muscular dystrophy (DMD), skeletal muscle is progressively replaced by fibrotic tissue. Here, we show that fibrinogen accumulates in dystrophic muscles of DMD patients and mdx mice. Genetic loss or pharmacological depletion of fibrinogen in these mice reduced fibrosis and dystrophy progression. Our results demonstrate that fibrinogen-Mac-1 receptor binding, through induction of IL-1beta, drives the synthesis of transforming growth factor-beta (TGFbeta) by mdx macrophages, which in turn induces collagen production in mdx fibroblasts. Fibrinogen-produced TGFbeta further amplifies collagen accumulation through activation of profibrotic alternatively activated macrophages. Fibrinogen, by engaging its alphavbeta3 receptor on fibroblasts, also directly promotes collagen synthesis. These data unveil a profibrotic role of fibrinogen deposition in muscle dystrophy. Topics: Animals; Cells, Cultured; Child; Child, Preschool; Collagen; Fibrinogen; Fibroblasts; Fibrosis; Humans; Integrin alphaVbeta3; Interleukin-1beta; Macrophage Activation; Macrophage-1 Antigen; Macrophages; Mice; Mice, Inbred mdx; Muscle, Skeletal; Muscular Dystrophy, Animal; Muscular Dystrophy, Duchenne; Protein Binding; Transforming Growth Factor beta | 2008 |
Wide ranges of serum myostatin concentrations in Duchenne muscular dystrophy patients.
Topics: Adolescent; Adult; Child; Child, Preschool; Dystrophin; Humans; Infant; Muscular Dystrophy, Duchenne; Myostatin; Transforming Growth Factor beta | 2008 |
Angiotensin II type 1 receptor blockade attenuates TGF-beta-induced failure of muscle regeneration in multiple myopathic states.
Skeletal muscle has the ability to achieve rapid repair in response to injury or disease. Many individuals with Marfan syndrome (MFS), caused by a deficiency of extracellular fibrillin-1, exhibit myopathy and often are unable to increase muscle mass despite physical exercise. Evidence suggests that selected manifestations of MFS reflect excessive signaling by transforming growth factor (TGF)-beta (refs. 2,3). TGF-beta is a known inhibitor of terminal differentiation of cultured myoblasts; however, the functional contribution of TGF-beta signaling to disease pathogenesis in various inherited myopathic states in vivo remains unknown. Here we show that increased TGF-beta activity leads to failed muscle regeneration in fibrillin-1-deficient mice. Systemic antagonism of TGF-beta through administration of TGF-beta-neutralizing antibody or the angiotensin II type 1 receptor blocker losartan normalizes muscle architecture, repair and function in vivo. Moreover, we show TGF-beta-induced failure of muscle regeneration and a similar therapeutic response in a dystrophin-deficient mouse model of Duchenne muscular dystrophy. Topics: Analysis of Variance; Angiotensin II Type 1 Receptor Blockers; Animals; Antibodies; Fibrillin-1; Fibrillins; Fluorescent Antibody Technique; Histocytochemistry; Losartan; Marfan Syndrome; Mice; Microfilament Proteins; Muscle, Skeletal; Muscular Dystrophy, Duchenne; Mutation; Regeneration; Signal Transduction; Transforming Growth Factor beta | 2007 |
ACE inhibitor bulks up muscle.
Topics: Angiotensin II Type 1 Receptor Blockers; Animals; Losartan; Marfan Syndrome; Mice; Models, Biological; Muscle, Skeletal; Muscular Dystrophy, Duchenne; Regeneration; Signal Transduction; Transforming Growth Factor beta | 2007 |
Two novel missense mutations in the myostatin gene identified in Japanese patients with Duchenne muscular dystrophy.
Myostatin is a negative regulator of skeletal muscle growth. Truncating mutations in the myostatin gene have been reported to result in gross muscle hypertrophy. Duchenne muscular dystrophy (DMD), the most common lethal muscle wasting disease, is a result of an absence of muscle dystrophin. Although this disorder causes a rather uniform pattern of muscle wasting, afflicted patients display phenotypic variability. We hypothesized that genetic variation in myostatin is a modifier of the DMD phenotype.. We analyzed 102 Japanese DMD patients for mutations in the myostatin gene.. Two polymorphisms that are commonly observed in Western countries, p.55A>T and p.153K>R, were not observed in these Japanese patients. An uncommon polymorphism of p.164E>K was uncovered in four cases; each patient was found to be heterozygous for this polymorphism, which had the highest frequency of the polymorphism observed in the Japanese patients. Remarkably, two patients were found to be heterozygous for one of two novel missense mutations (p.95D>H and p.156L>I). One DMD patient carrying a novel missense mutation of p.95D>H was not phenotypically different from the non-carriers. The other DMD patient was found to carry both a novel mutation (p.156L>I) and a known polymorphism (p.164E>K) in one allele, although his phenotype was not significantly modified. Any nucleotide change creating a target site for micro RNAs was not disclosed in the 3' untranslated region.. Our results indicate that heterozygous missense mutations including two novel mutations did not produce an apparent increase in muscle strength in Japanese DMD cases, even in a patient carrying two missense mutations. Topics: Adolescent; Adult; Child; Child, Preschool; Humans; Infant; Japan; Male; Muscular Dystrophy, Duchenne; Mutation, Missense; Myostatin; Polymerase Chain Reaction; Sequence Analysis, DNA; Transforming Growth Factor beta | 2007 |
Studies point way to new therapeutic prospects for muscular dystrophy.
Topics: Animals; Codon, Nonsense; Dystrophin; Gene Expression; Humans; Losartan; Marfan Syndrome; Mice; Muscular Dystrophy, Duchenne; Oxadiazoles; PPAR alpha; Transforming Growth Factor beta | 2007 |
Pentoxifylline fails to attenuate fibrosis in dystrophic (mdx) diaphragm muscle.
Fibrosis is a common pathological feature observed in muscle from patients with Duchenne muscular dystrophy and in mdx diaphragm. The purpose of this study was to determine whether pentoxifylline (PTX) treatment for 4 weeks (16 mg/kg/day) could significantly attenuate the process of fibrosis in diaphragm muscle from mdx mice. PTX treatment had no impact on in vitro diaphragm muscle contractile function. In addition, diaphragm muscle hydroxyproline concentration and the level of type I and III collagen and TGF-beta1 mRNA were unaffected by PTX treatment. These findings do not support the use of PTX as an antifibrotic drug for the treatment of muscular dystrophy. Topics: Animals; Collagen Type I; Collagen Type III; Diaphragm; Disease Models, Animal; Enzyme Inhibitors; Fibrosis; Hydroxyproline; Male; Mice; Mice, Inbred C57BL; Mice, Inbred mdx; Muscle Contraction; Muscular Dystrophy, Animal; Muscular Dystrophy, Duchenne; Pentoxifylline; RNA, Messenger; Transforming Growth Factor beta; Transforming Growth Factor beta1; Treatment Failure | 2006 |
Long-term administration of pirfenidone improves cardiac function in mdx mice.
Duchenne muscular dystrophy, an X-linked recessive neuromuscular disorder due to lack of the protein dystrophin, manifests as progressive muscle degeneration and cardiomyopathy with increased fibrosis. The exact mechanisms involved in fibrosis are unknown, but a cytokine, transforming growth factor-beta (TGF-beta), is a likely mediator. This study tested whether the TGF-beta antagonist, pirfenidone, could reduce cardiac fibrosis. Eight-month-old mdx mice were treated for 7 months with 0.4%, 0.8%, and 1.2% pirfenidone in drinking water; untreated water was given to control mdx and C57 mice. Mice treated with 0.8% and 1.2% pirfendone had lowered cardiac TGF-beta mRNA and improved in vitro cardiac contractility (P < 0.05) to levels consistent with C57 mice, yet without a change in cardiac stiffness or fibrosis. These results show that the TGF-beta antagonist, pirfenidone, can improve cardiac function in mdx mice, potentially providing a new avenue for developing cardiac therapies for patients with Duchenne muscular dystrophy. Topics: Animals; Anti-Inflammatory Agents, Non-Steroidal; Cardiomyopathies; Disease Models, Animal; Fibrosis; Male; Mice; Mice, Inbred C57BL; Mice, Inbred mdx; Muscular Dystrophy, Animal; Muscular Dystrophy, Duchenne; Myocardium; Pyridones; RNA, Messenger; Transforming Growth Factor beta; Ventricular Dysfunction, Left | 2006 |
Muscle-bone interactions in dystrophin-deficient and myostatin-deficient mice.
We have investigated muscle-bone interactions using two mouse mutants that are known to differ from normal mice in skeletal muscle growth and development: mice lacking myostatin (GDF8) and mice lacking dystrophin (mdx). Myostatin-deficient mice show increased muscle size and strength compared to normal mice, whereas the mdx mouse is a well-established animal model for Duchenne muscular dystrophy. The mdx mice have significantly larger hindlimb muscles than controls, and histological sections of the quadriceps muscles show dystrophic changes with extensive fibrosis. Femoral bone mineral density (BMD) and fracture strength (Fu) are significantly greater in mdx mice than controls, and these variables are more strongly correlated with quadriceps muscle mass than with body mass. In contrast, mdx mice do not shower high bone mineral density in the spine relative to controls, whereas myostatin-deficient mice have significantly increased BMD in the lumbar spine compared to normal mice. Both mdx mice and myostatin-deficient mice have expanded femoral trochanters for attachment of large hindlimb muscles, and both mutant strains show increased cross-sectional area moments of inertia mediolaterally (Iyy) but not anteroposteriorly (Ixx) compared to normal mice. These data suggest that lean (muscle) mass is a significant determinant of bone mineral density and strength in the limb skeleton, even when accompanied by a dystrophic phenotype. Likewise, increased muscle mass produces a marked increase in the external dimensions of muscle attachment sites, even when increased muscle size is accompanied by extensive fibrosis and muscle weakness. Topics: Animals; Bone Density; Disease Models, Animal; Dystrophin; Female; Femur; Lumbar Vertebrae; Mice; Mice, Inbred C57BL; Mice, Inbred mdx; Mice, Mutant Strains; Muscle Fibers, Skeletal; Muscle Weakness; Muscle, Skeletal; Muscular Dystrophy, Animal; Muscular Dystrophy, Duchenne; Myostatin; Transforming Growth Factor beta | 2005 |
Early onset of inflammation and later involvement of TGFbeta in Duchenne muscular dystrophy.
To identify stage-specific induction of molecular pathology pathways in Duchenne muscular dystrophy (DMD).. We performed mRNA profiling using muscles from fetopsies, infants (aged 8 to 10 months), and symptomatic patients (aged 5 to 12 years) with DMD, and age- and sex-matched controls. We performed immunohistochemistry to determine changes at the protein level and protein localization.. Activated tissue dendritic cells, expression of toll-like receptor 7, and strong induction of nuclear factor-kappaB pathways occurred soon after birth in DMD muscle. Two muscle wasting pathways, atrogin-1 and myostatin, were not induced at any stage of the disease. Normal muscle showed accumulation of glycolytic and oxidative metabolism capacity with increased age, but this accumulation failed in DMD. The transforming growth factor (TGF)-beta pathway was strongly induced in symptomatic patients, with expression of TGFbeta type II receptor and apoptosis signal-regulating kinase 1 proteins on subsets of mature DMD myofibers.. Our data show stage-specific remodeling of human dystrophin-deficient muscle, with inflammatory pathways predominating in the presymptomatic stages and acute activation of TGFbeta and failure of metabolic pathways later in the disease. Topics: Child; Child, Preschool; Dendritic Cells; Disease Progression; Energy Metabolism; Female; Humans; Immunohistochemistry; Infant; Inflammation; Male; MAP Kinase Kinase Kinase 5; Muscle Fibers, Skeletal; Muscle, Skeletal; Muscular Dystrophy, Duchenne; NF-kappa B; Protein Serine-Threonine Kinases; Receptor, Transforming Growth Factor-beta Type II; Receptors, Transforming Growth Factor beta; RNA, Messenger; Signal Transduction; Toll-Like Receptor 7; Transforming Growth Factor beta | 2005 |
Decorin and biglycan expression is differentially altered in several muscular dystrophies.
Biglycan and decorin are small extracellular proteoglycans that interact with cytokines, whose activity they may modulate, and with matrix proteins, particularly collagens. To better understand their role in muscle fibrosis, we investigated expression of decorin and biglycan transcripts and protein in muscle of several forms of muscular dystrophy, and also expression of perlecan, an extracellular proteoglycan unrelated to collagen deposition. In Duchenne muscular dystrophy (DMD) and LAMA2-mutated congenital muscular dystrophy (MDC1A) we also quantitated transcript levels of the profibrotic cytokine TGF-beta1. We examined muscle biopsies from nine DMD patients, aged 2-8 years; 14 BMD (Becker muscular dystrophy) patients (nine aged 1-5 years; five aged 30-37 years); four MDC1A patients (aged 2-7 years); six dysferlin-deficient patients (aged 19-53 years) with mutation ascertained in two, and normal expression of proteins related to limb girdle muscular dystrophies in the others; 10 sarcoglycan-deficient patients: seven with alpha-sarcoglycan mutation, two with beta-sarcoglycan mutation and one with gamma-sarcoglycan mutation (five aged 8-15 years; five aged 26-43 years); and nine children (aged 1-6 years) and 12 adults (aged 16-61 years) suspected of neuromuscular disease, but who had normal muscle on biopsy. Biglycan mRNA levels varied in DMD and MDC1A depending on the quantitation method, but were upregulated in BMD, sarcoglycanopathies and dysferlinopathy. Decorin mRNA was significantly downregulated in DMD and MDC1A, whereas TGF-beta1 was significantly upregulated. Decorin mRNA was normal in paediatric BMD, but upregulated in adult BMD, sarcoglycanopathies and dysferlinopathy. Perlecan transcript levels were similar to those of age-matched controls in all disease groups. By immunohistochemistry, decorin and biglycan were mainly localized in muscle connective tissue; their presence increased in relation to increased fibrosis in all dystrophic muscle. By visual inspection, decorin bands on immunoblot did not differ from those of age-matched controls in all patient groups. However, when the intensity of the bands was quantitated against vimentin and normalized against sarcomeric actin, in DMD and MDC1A the ratio of band intensities was significantly lower than in age-matched controls. Variations in the transcript and protein levels of these proteoglycans in different muscular dystrophies probably reflect the variable disruption of extracellular matrix orga Topics: Adolescent; Adult; Biglycan; Child; Child, Preschool; Collagen Type VI; Decorin; Extracellular Matrix Proteins; Gene Expression; Heparan Sulfate Proteoglycans; Humans; Infant; Male; Middle Aged; Muscle, Skeletal; Muscular Dystrophies; Muscular Dystrophy, Duchenne; Polymerase Chain Reaction; Proteoglycans; RNA, Messenger; Transforming Growth Factor beta; Transforming Growth Factor beta1 | 2005 |
[Altered expression of myostatin gene in the progressive muscular dystrophy patients].
Progressive muscular dystrophy is a group of inherited disorders characterized by progressive skeletal muscle wasting and weakness, which is not of neurogenic origin. Myostatin, a new member of the TGF-beta super-family, is a negative regulator of skeletal muscle growth. To investigate the possible involvement of myostatin in the development of progressive muscular dystrophy, we cloned and sequenced myostatin cDNAs from the progressive muscular dystrophy patients by RT-PCR. Levels of myostatin mRNA and protein in the patients were analyzed by semi-quantitative RT-PCR and Western blot,respectively. We did not find any mutations in the myostatin cDNA sequences from the progressive muscular dystrophy patients in this study. However, we found that the levels of myostatin transcripts were reduced in some patients and the processing and maturation of myostatin protein were inhibited in some patients. Our data demonstrated that the pathogenesis of some types or subtypes of progressive muscular dystrophy is probably associated with the altered myostatin expression and the processing inhibition of myostatin protein. Topics: Blotting, Western; DNA, Complementary; Humans; Muscle, Skeletal; Muscular Dystrophies, Limb-Girdle; Muscular Dystrophy, Duchenne; Myostatin; Promoter Regions, Genetic; Reverse Transcriptase Polymerase Chain Reaction; RNA, Messenger; Sequence Analysis, DNA; Transcription, Genetic; Transforming Growth Factor beta | 2005 |
Fourth round table conference in Monaco on 15 January 2005: regulation of muscle growth, a therapeutic issue for Duchenne muscular dystrophy?
Topics: Animals; Calpain; Humans; Insulin-Like Growth Factor I; Muscle Development; Muscular Dystrophy, Duchenne; Myostatin; Transforming Growth Factor beta | 2005 |
Localization and early time course of TGF-beta 1 mRNA expression in dystrophic muscle.
Fibrosis is a common pathological feature observed in muscle from patients with Duchenne muscular dystrophy (DMD). In the dystrophic (mdx) mouse model of DMD, the diaphragm is more severely affected than other skeletal muscles. The level of transforming growth factor-beta1 (TGF-beta1), an inflammatory cytokine, is significantly elevated in mdx diaphragm. However, little is known about the onset of TGF-beta1 messenger ribonucleic acid (mRNA) expression, or which cells express the mRNA. In this study, we characterized the location and time course of expression of TGF-beta1 mRNA in diaphragm from mdx mice. TGF-beta1 mRNA was significantly elevated in mdx diaphragm at 6 and 9 but not 12 weeks of age, and these changes corresponded with changes in type I collagen mRNA and hydroxyproline concentration. Mononucleated cells localized to areas of fiber necrosis highly expressed the TGF-beta1 transcript in mdx diaphragm. Neutralization of TGF-beta1 by decorin administration resulted in a 40% reduction in the level of diaphragm muscle type I collagen mRNA. These findings support a role for TGF-beta1 during the early stages of fibrogenesis in dystrophic diaphragm muscle. Therapeutic interventions aimed at neutralizing this cytokine may be beneficial in slowing the development of fibrosis in DMD. Topics: Animals; Diaphragm; Gene Expression Regulation; Male; Mice; Mice, Inbred C57BL; Mice, Inbred mdx; Muscular Dystrophy, Duchenne; RNA, Messenger; Time Factors; Transforming Growth Factor beta; Transforming Growth Factor beta1 | 2004 |
Fibrogenic cytokines and extent of fibrosis in muscle of dogs with X-linked golden retriever muscular dystrophy.
Fibrosis in Duchenne muscular dystrophy patients' muscle seems to be mediated by fibrogenic cytokines, particularly transforming growth factor-beta1. Golden retriever muscular dystrophy is a model of Duchenne dystrophy characterised by severe myopathy and muscle fibrosis. We evaluated mRNA levels and protein distribution of transforming growth factor-beta1, connective tissue growth factor and collagens in muscle of golden retriever muscular dystrophy dogs at different ages. Fibrosis occurs early in golden retriever muscular dystrophy dogs and transforming growth factor-beta1 levels tend to be high up to 60 days of age (P=0.019 at 30 days), suggesting that transforming growth factor-beta1 is involved in the early stages of fibrosis. We also found greater expression of connective tissue growth factor in golden retriever muscular dystrophy than control muscle (P=0.0065 at 30 days), suggesting involvement of this molecule in fibrosis progression. Our findings sustain the hypothesis that cytokines are actively involved in fibrosis in golden retriever muscular dystrophy, as it seems to be in humans, and confirm the utility of this model for investigating new therapeutic approaches for Duchenne dystrophy. Topics: Age Factors; Animals; Base Sequence; Collagen; Connective Tissue Growth Factor; Cytokines; Disease Models, Animal; Dogs; Female; Fibrosis; Genetic Diseases, X-Linked; Immediate-Early Proteins; Immunohistochemistry; Intercellular Signaling Peptides and Proteins; Male; Muscular Dystrophy, Duchenne; Polymerase Chain Reaction; RNA, Messenger; Transforming Growth Factor beta; Transforming Growth Factor beta1 | 2002 |
Functional improvement of dystrophic muscle by myostatin blockade.
Mice and cattle with mutations in the myostatin (GDF8) gene show a marked increase in body weight and muscle mass, indicating that this new member of the TGF-beta superfamily is a negative regulator of skeletal muscle growth. Inhibition of the myostatin gene product is predicted to increase muscle mass and improve the disease phenotype in a variety of primary and secondary myopathies. We tested the ability of inhibition of myostatin in vivo to ameliorate the dystrophic phenotype in the mdx mouse model of Duchenne muscular dystrophy (DMD). Blockade of endogenous myostatin by using intraperitoneal injections of blocking antibodies for three months resulted in an increase in body weight, muscle mass, muscle size and absolute muscle strength in mdx mouse muscle along with a significant decrease in muscle degeneration and concentrations of serum creatine kinase. The functional improvement of dystrophic muscle by myostatin blockade provides a novel, pharmacological strategy for treatment of diseases associated with muscle wasting such as DMD, and circumvents the major problems associated with conventional gene therapy in these disorders. Topics: Animals; Antibodies; Body Weight; Creatine Kinase; Male; Mice; Mice, Inbred mdx; Muscle Fibers, Skeletal; Muscle, Skeletal; Muscular Dystrophy, Animal; Muscular Dystrophy, Duchenne; Myostatin; Organ Size; Transforming Growth Factor beta | 2002 |
Growth factor-dependent proliferation and invasion of muscle satellite cells require the cell-associated fibrinolytic system.
The process of muscle regeneration in normal and dystrophic muscle depends on locally produced cytokines and growth factors and requires the activity of the urokinase plasminogen activator/urokinase plasminogen activator receptor/plasminogen activator inhibitor-1 system. In this study we tested the effect of basic fibroblast growth factor (bFGF), hepatocyte growth factor (HGF) and transforming growth factor-beta (TGFbeta) on the fibrinolytic pattern of normal and dystrophic satellite cells, their mitogenic and motogenic activities and the dependence of such activities on the cell-associated fibrinolytic system. We have observed that the urokinase plasminogen activator (u-PA) receptor is weakly upregulated by bFGF in normal satellite cells, while it is strongly up-regulated by TGFbeta, mainly in dystrophic myoblasts. bFGF up-regulated u-PA in both normal and dystrophic myoblasts grown in primary culture, while a striking down-regulation was observed with TGFbeta. TGFbeta was the only growth factor able to exceptionally up-regulate plasminogen activator inhibitor-1 (PAI-1), mainly in dystrophic satellite cells. HGF did not show any activity on the fibrinolytic system. Proliferation and invasion into Matrigel matrices of normal and dystrophic cells occurred regardless of the growth factor-dependent regulation of the fibrinolytic system. Nevertheless, each growth factor required the efficiency of the constitutive cell-associated fibrinolytic system to operate, as shown by impairment of growth factor activity with antagonists of u-PA and of its receptor. Noteworthy, TGFbeta induced a dose-dependent increase of Matrigel invasion only in dystrophic myoblasts. Since TGFbeta-challenged dystrophic myoblasts undergo an exceptional up-regulation of the receptor and of PAI-1, we propose the possibility that the TGFbeta-induced fibrinolytic pattern (low urokinase plasminogen activator, high receptor and high PAI-1) may be exploited to promote survival and spreading of transplanted engineered myoblasts in Duchenne muscular dystrophy. Topics: Case-Control Studies; Cell Division; Cell Movement; Child; Child, Preschool; Fibrinolytic Agents; Fibroblast Growth Factor 2; Growth Substances; Hepatocyte Growth Factor; Humans; Infant; Muscle, Skeletal; Muscular Dystrophy, Duchenne; Myeloid Progenitor Cells; Plasminogen Activator Inhibitor 1; Plasminogen Activators; Receptors, Cell Surface; Receptors, Urokinase Plasminogen Activator; Regeneration; Transforming Growth Factor beta; Urokinase-Type Plasminogen Activator | 2002 |
Defective growth in vitro of Duchenne Muscular Dystrophy myoblasts: the molecular and biochemical basis.
As the molecular basis of Duchenne Muscular Dystrophy (DMD) was being discovered, increasing focus was placed on the mechanisms of progressive failure of myoregeneration. In this study, we propose a pathogenesis model for DMD, where an autocrine growth factor release of TGF-beta1-from necrotic myofibers-could contribute to the increasing loss of muscle regeneration. In fact, we report evidence that DMD myoblasts reduce their proliferation rate, in time and later cultures; in connection with this, we observed TGF-beta1 increase in conditioned media of DMD myoblasts, able to control the myoblast growth by reducing fusion and differentiation of DMD satellite cells. Topics: Cell Differentiation; Cell Division; Cell Fusion; Child, Preschool; Culture Media, Conditioned; Humans; Muscle, Skeletal; Muscular Dystrophy, Duchenne; Regeneration; Transforming Growth Factor beta | 1999 |