Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters











Publication year range
1.
Exp Cell Res ; 433(2): 113820, 2023 12 15.
Article in English | MEDLINE | ID: mdl-37879549

ABSTRACT

The Warburg effect links growth and glycolysis in cancer. A key purpose of the Warburg effect is to generate glycolytic intermediates for anabolic reactions, such as nucleotides → RNA/DNA and amino acids → protein synthesis. The aim of this study was to investigate whether a similar 'glycolysis-for-anabolism' metabolic reprogramming also occurs in hypertrophying skeletal muscle. To interrogate this, we first induced C2C12 myotube hypertrophy with IGF-1. We then added 14C glucose to the differentiation medium and measured radioactivity in isolated protein and RNA to establish whether 14C had entered anabolism. We found that especially protein became radioactive, suggesting a glucose → glycolytic intermediates → non-essential amino acid(s) → protein series of reactions, the rate of which was increased by IGF-1. Next, to investigate the importance of glycolytic flux and non-essential amino acid synthesis for myotube hypertrophy, we exposed C2C12 and primary mouse myotubes to the glycolysis inhibitor 2-Deoxy-d-glucose (2DG). We found that inhibiting glycolysis lowered C2C12 and primary myotube size. Similarly, siRNA silencing of PHGDH, the key enzyme of the serine biosynthesis pathway, decreased C2C12 and primary myotube size; whereas retroviral PHGDH overexpression increased C2C12 myotube size. Together these results suggest that glycolysis is important for hypertrophying myotubes, which reprogram their metabolism to facilitate anabolism, similar to cancer cells.


Subject(s)
Insulin-Like Growth Factor I , Neoplasms , Animals , Mice , Insulin-Like Growth Factor I/metabolism , Phosphoglycerate Dehydrogenase/genetics , Phosphoglycerate Dehydrogenase/metabolism , Phosphoglycerate Dehydrogenase/pharmacology , Muscle Fibers, Skeletal/metabolism , Neoplasms/metabolism , RNA/metabolism , Hypertrophy/metabolism , Glucose/pharmacology , Amino Acids/genetics , Amino Acids/metabolism , Amino Acids/pharmacology
2.
Elife ; 112022 03 24.
Article in English | MEDLINE | ID: mdl-35323108

ABSTRACT

In skeletal muscle, transforming growth factor-ß (TGF-ß) family growth factors, TGF-ß1 and myostatin, are involved in atrophy and muscle wasting disorders. Simultaneous interference with their signalling pathways may improve muscle function; however, little is known about their individual and combined receptor signalling. Here, we show that inhibition of TGF-ß signalling by simultaneous muscle-specific knockout of TGF-ß type I receptors Tgfbr1 and Acvr1b in mice, induces substantial hypertrophy, while such effect does not occur by single receptor knockout. Hypertrophy is induced by increased phosphorylation of Akt and p70S6K and reduced E3 ligases expression, while myonuclear number remains unaltered. Combined knockout of both TGF-ß type I receptors increases the number of satellite cells, macrophages and improves regeneration post cardiotoxin-induced injury by stimulating myogenic differentiation. Extra cellular matrix gene expression is exclusively elevated in muscle with combined receptor knockout. Tgfbr1 and Acvr1b are synergistically involved in regulation of myofibre size, regeneration, and collagen deposition.


Subject(s)
Muscle Development , Muscle, Skeletal , Animals , Hypertrophy , Mice , Muscle, Skeletal/metabolism , Receptor, Transforming Growth Factor-beta Type I/metabolism , Transforming Growth Factor beta/metabolism
3.
Aging (Albany NY) ; 14(1): 28-53, 2022 01 13.
Article in English | MEDLINE | ID: mdl-35023852

ABSTRACT

Aging-associated muscle wasting and impaired regeneration are caused by deficiencies in muscle stem cell (MuSC) number and function. We postulated that aged MuSCs are intrinsically impaired in their responsiveness to omnipresent mechanical cues through alterations in MuSC morphology, mechanical properties, and number of integrins, culminating in impaired proliferative capacity. Here we show that aged MuSCs exhibited significantly lower growth rate and reduced integrin-α7 expression as well as lower number of phospho-paxillin clusters than young MuSCs. Moreover, aged MuSCs were less firmly attached to matrigel-coated glass substrates compared to young MuSCs, as 43% of the cells detached in response to pulsating fluid shear stress (1 Pa). YAP nuclear localization was 59% higher than in young MuSCs, yet YAP target genes Cyr61 and Ctgf were substantially downregulated. When subjected to pulsating fluid shear stress, aged MuSCs exhibited reduced upregulation of proliferation-related genes. Together these results indicate that aged MuSCs exhibit impaired mechanosensitivity and growth potential, accompanied by altered morphology and mechanical properties as well as reduced integrin-α7 expression. Aging-associated impaired muscle regenerative capacity and muscle wasting is likely due to aging-induced intrinsic MuSC alterations and dysfunctional mechanosensitivity.


Subject(s)
Cell Proliferation/physiology , Cellular Senescence/physiology , Mechanotransduction, Cellular/physiology , Muscle Fibers, Skeletal/physiology , Stem Cells/physiology , Aging , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Cell Adhesion/physiology , Integrin alpha Chains/genetics , Integrin alpha Chains/metabolism , Mice , Nitric Oxide/metabolism , Shear Strength
4.
Cells ; 9(2)2020 02 06.
Article in English | MEDLINE | ID: mdl-32041253

ABSTRACT

Transforming Growth Factor ß (TGF-ß) is involved in fibrosis as well as the regulation of muscle mass, and contributes to the progressive pathology of muscle wasting disorders. However, little is known regarding the time-dependent signalling of TGF-ß in myoblasts and myotubes, as well as how TGF-ß affects collagen type I expression and the phenotypes of these cells. Here, we assessed effects of TGF-ß on gene expression in C2C12 myoblasts and myotubes after 1, 3, 9, 24 and 48 h treatment. In myoblasts, various myogenic genes were repressed after 9, 24 and 48 h, while in myotubes only a reduction in Myh3 expression was observed. In both myoblasts and myotubes, TGF-ß acutely induced the expression of a subset of genes involved in fibrosis, such as Ctgf and Fgf-2, which was subsequently followed by increased expression of Col1a1. Knockdown of Ctgf and Fgf-2 resulted in a lower Col1a1 expression level. Furthermore, the effects of TGF-ß on myogenic and fibrotic gene expression were more pronounced than those of myostatin, and knockdown of TGF-ß type I receptor Tgfbr1, but not receptor Acvr1b, resulted in a reduction in Ctgf and Col1a1 expression. These results indicate that, during muscle regeneration, TGF-ß induces fibrosis via Tgfbr1 by stimulating the autocrine signalling of Ctgf and Fgf-2.


Subject(s)
Collagen Type I/metabolism , Connective Tissue Growth Factor/metabolism , Fibroblast Growth Factor 2/metabolism , Muscle Fibers, Skeletal/metabolism , Myoblasts/metabolism , Transforming Growth Factor beta/pharmacology , Animals , Cell Differentiation/drug effects , Cell Size/drug effects , Cells, Cultured , Fibrosis , Gene Expression Regulation/drug effects , Mice, Inbred C57BL , Models, Biological , Muscle Development/drug effects , Muscle Development/genetics , Muscle Fibers, Skeletal/drug effects , Myoblasts/drug effects , Myostatin/pharmacology , Receptor, Transforming Growth Factor-beta Type I/metabolism , Time Factors
5.
Adv Exp Med Biol ; 1088: 109-137, 2018.
Article in English | MEDLINE | ID: mdl-30390250

ABSTRACT

Insulin-like growth factor 1 (IGF-1) is a key anabolic growth factor stimulating phosphatidylinositol 3-kinase (PI3K)/Akt signaling which is well known for regulating muscle hypertrophy. However, the role of IGF-1 in muscle atrophy is less clear. This review provides an overview of the mechanisms via which IGF-1 signaling is implicated in several conditions of muscle atrophy and via which mechanisms protein turnover is altered. IGF-1/PI3K/Akt signaling stimulates the rate of protein synthesis via p70S6Kinase and p90 ribosomal S6 kinase and negatively regulates protein degradation, predominantly by its inhibiting effect on proteasomal and lysosomal protein degradation. Caspase-dependent protein degradation is also attenuated by IGF/PI3K/Akt signaling, whereas evidence for an effect on calpain-dependent protein degradation is inconclusive. IGF-1/PI3K/Akt signaling reduces during denervation-, unloading-, and joint immobilization-induced muscle atrophy, whereas IGF-1/PI3K/Akt signaling seems unaltered during aging-associated muscle atrophy. During denervation and aging, IGF-1 overexpression or injection counteracts denervation- and aging-associated muscle atrophy, despite enhanced anabolic resistance with regard to IGF-1 signaling with aging. It remains unclear whether pharmacological stimulation of IGF-1/PI3K/Akt signaling attenuates immobilization- or unloading-induced muscle atrophy. Exploration of the possibilities to interfere with IGF-1/PI3K/Akt signaling reveals that microRNAs targeting IGF-1 signaling components are promising targets to counterbalance muscle atrophy. Overall, the findings summarized in this review show that in disuse conditions, but not with aging, IGF-1/PI3K/Akt signaling is attenuated and that in some conditions stimulation of this pathway may alleviate skeletal muscle atrophy.


Subject(s)
Insulin-Like Growth Factor I/metabolism , Muscular Atrophy/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Humans , Muscle, Skeletal/physiopathology , Muscular Atrophy/physiopathology
6.
Adv Exp Med Biol ; 1088: 153-206, 2018.
Article in English | MEDLINE | ID: mdl-30390252

ABSTRACT

Myostatin was identified more than 20 years ago as a negative regulator of muscle mass in mice and cattle. Since then, a wealth of studies have uncovered the potential involvement of myostatin in muscle atrophy and sparked interest in myostatin as a promising therapeutic target to counteract decline of muscle mass in patients afflicted with different muscle-wasting conditions. Insight in the molecular mechanism of myostatin signaling and regulation of myostatin activity has resulted in the identification of specific treatments to inhibit myostatin signaling and related signaling pathways. Currently, several treatments that target myostatin and related proteins have been evaluated in preclinical animal models of muscle wasting, and some potential therapies have progressed to clinical trials. However, studies also revealed potential downsides of myostatin targeting in skeletal muscle and other tissues, which raises the question if myostatin is indeed a valuable target to counteract muscle atrophy. In this review we provide an updated overview of the molecular mechanisms of myostatin signaling, the preclinical evidence supporting a role for myostatin and related proteins in muscle atrophy, and the potential issues that arise when targeting myostatin. In addition, we evaluate the current clinical status of different treatments aimed at inhibiting myostatin and discuss future perspectives of targeting myostatin to counteract muscle atrophy.


Subject(s)
Molecular Targeted Therapy , Muscle, Skeletal/pathology , Muscular Atrophy/drug therapy , Myostatin/antagonists & inhibitors , Signal Transduction , Animals , Cattle , Humans , Mice , Muscular Atrophy/physiopathology
7.
FASEB J ; 31(1): 238-255, 2017 01.
Article in English | MEDLINE | ID: mdl-27733450

ABSTRACT

Skeletal muscle fibrosis and impaired muscle regeneration are major contributors to muscle wasting in Duchenne muscular dystrophy (DMD). Muscle growth is negatively regulated by myostatin (MSTN) and activins. Blockage of these pathways may improve muscle quality and function in DMD. Antisense oligonucleotides (AONs) were designed specifically to block the function of ALK4, a key receptor for the MSTN/activin pathway in skeletal muscle. AON-induced exon skipping resulted in specific Alk4 down-regulation, inhibition of MSTN activity, and increased myoblast differentiation in vitro Unexpectedly, a marked decrease in muscle mass (10%) was found after Alk4 AON treatment in mdx mice. In line with in vitro results, muscle regeneration was stimulated, and muscle fiber size decreased markedly. Notably, when Alk4 was down-regulated in adult wild-type mice, muscle mass decreased even more. RNAseq analysis revealed dysregulated metabolic functions and signs of muscle atrophy. We conclude that ALK4 inhibition increases myogenesis but also regulates the tight balance of protein synthesis and degradation. Therefore, caution must be used when developing therapies that interfere with MSTN/activin pathways.-Pasteuning-Vuhman, S., Boertje-van der Meulen, J. W., van Putten, M., Overzier, M., ten Dijke, P., Kielbasa, S. M., Arindrarto, W., Wolterbeek, R., Lezhnina, K. V., Ozerov, I. V., Aliper, A. M., Hoogaars, W. M., Aartsma-Rus, A., Loomans, C. J. M. New function of the myostatin/activin type I receptor (ALK4) as a mediator of muscle atrophy and muscle regeneration.


Subject(s)
Activin Receptors, Type I/metabolism , Muscle, Skeletal/physiology , Myoblasts/physiology , Regeneration/physiology , Activin Receptors, Type I/genetics , Animals , Base Sequence , Cell Line , DNA Damage , Down-Regulation , Gene Expression Regulation/physiology , Mice , Mice, Inbred mdx , Muscle Development/physiology , Oligonucleotides, Antisense/pharmacology , RNA/genetics , RNA/metabolism , Regeneration/genetics , Signal Transduction
8.
J Am Soc Nephrol ; 27(12): 3589-3599, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27020852

ABSTRACT

Autosomal dominant polycystic kidney disease (ADPKD), characterized by the formation of numerous kidney cysts, is caused by PKD1 or PKD2 mutations and affects 0.1% of the population. Although recent clinical studies indicate that reduction of cAMP levels slows progression of PKD, this finding has not led to an established safe and effective therapy for patients, indicating the need to find new therapeutic targets. The role of TGF-ß in PKD is not clearly understood, but nuclear accumulation of phosphorylated SMAD2/3 in cyst-lining cells suggests the involvement of TGF-ß signaling in this disease. In this study, we ablated the TGF-ß type 1 receptor (also termed activin receptor-like kinase 5) in renal epithelial cells of PKD mice, which had little to no effect on the expression of SMAD2/3 target genes or the progression of PKD. Therefore, we investigated whether alternative TGF-ß superfamily ligands account for SMAD2/3 activation in cystic epithelial cells. Activins are members of the TGF-ß superfamily and drive SMAD2/3 phosphorylation via activin receptors, but activins have not been studied in the context of PKD. Mice with PKD had increased expression of activin ligands, even at early stages of disease. In addition, treatment with a soluble activin receptor IIB fusion (sActRIIB-Fc) protein, which acts as a soluble trap to sequester activin ligands, effectively inhibited cyst formation in three distinct mouse models of PKD. These data point to activin signaling as a key pathway in PKD and a promising target for therapy.


Subject(s)
Activins/antagonists & inhibitors , Polycystic Kidney Diseases/prevention & control , Signal Transduction , Animals , Disease Progression , Epithelial Cells , Female , Kidney/cytology , Male , Mice , Polycystic Kidney Diseases/etiology , Recombinant Fusion Proteins/pharmacology , Smad2 Protein/physiology , Smad3 Protein/physiology , Time Factors
9.
Mol Ther Nucleic Acids ; 3: e156, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24691207

ABSTRACT

Duchenne muscular dystrophy (DMD) is caused by lack of functional dystrophin and results in progressive myofiber damage and degeneration. In addition, impaired muscle regeneration and fibrosis contribute to the progressive pathology of DMD. Importantly, transforming growth factor-ß (TGF-ß) is implicated in DMD pathology and is known to stimulate fibrosis and inhibit muscle regeneration. In this study, we present a new strategy to target TGF-ß signaling cascades by specifically inhibiting the expression of TGF-ß type I receptor TGFBR1 (ALK5). Antisense oligonucleotides (AONs) were designed to specifically induce exon skipping of mouse ALK5 transcripts. AON-induced exon skipping of ALK5 resulted in specific downregulation of full-length receptor transcripts in vitro in different cell types, repression of TGF-ß activity, and enhanced C2C12 myoblast differentiation. To determine the effect of these AONs in dystrophic muscles, we performed intramuscular injections of ALK5 AONs in mdx mice, which resulted in a decrease in expression of fibrosis-related genes and upregulation of Myog expression compared to control AON-injected muscles. In summary, our study presents a novel method to target TGF-ß signaling cascades with potential beneficial effects for DMD.

10.
PLoS One ; 8(7): e69096, 2013.
Article in English | MEDLINE | ID: mdl-23861958

ABSTRACT

Fibrodysplasia ossificans progressiva (FOP) is a rare heritable disease characterized by progressive heterotopic ossification of connective tissues, for which there is presently no definite treatment. A recurrent activating mutation (c.617G→A; R206H) of activin receptor-like kinase 2 (ACVR1/ALK2), a BMP type I receptor, has been shown as the main cause of FOP. This mutation constitutively activates the BMP signaling pathway and initiates the formation of heterotopic bone. In this study, we have designed antisense oligonucleotides (AONs) to knockdown mouse ALK2 expression by means of exon skipping. The ALK2 AON could induce exon skipping in cells, which was accompanied by decreased ALK2 mRNA levels and impaired BMP signaling. In addition, the ALK2 AON potentiated muscle differentiation and repressed BMP6-induced osteoblast differentiation. Our results therefore provide a potential therapeutic approach for the treatment of FOP disease by reducing the excessive ALK2 activity in FOP patients.


Subject(s)
Activin Receptors, Type I/antagonists & inhibitors , Exons , Muscle Cells/cytology , Myoblasts/cytology , Myositis Ossificans/therapy , Oligonucleotides, Antisense/genetics , Activin Receptors, Type I/genetics , Activin Receptors, Type I/metabolism , Animals , Bone Morphogenetic Protein 6/genetics , Bone Morphogenetic Protein 6/metabolism , Cell Differentiation , Gene Expression Regulation , Gene Knockdown Techniques , Genetic Therapy/methods , Humans , Mice , Muscle Cells/metabolism , Mutation , Myoblasts/metabolism , Myositis Ossificans/genetics , Myositis Ossificans/metabolism , Myositis Ossificans/pathology , Oligonucleotides, Antisense/chemical synthesis , Osteoblasts/metabolism , Osteoblasts/pathology , Osteogenesis/genetics , Signal Transduction
11.
Am J Physiol Endocrinol Metab ; 305(2): E171-82, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23695214

ABSTRACT

The importance of adequate levels of muscle size and function and physical activity is widely recognized. Myostatin/activin blocking increases skeletal muscle mass but may decrease muscle oxidative capacity and can thus be hypothesized to affect voluntary physical activity. Soluble activin receptor IIB (sActRIIB-Fc) was produced to block myostatin/activins. Modestly dystrophic mdx mice were injected with sActRIIB-Fc or PBS with or without voluntary wheel running exercise for 7 wk. Healthy mice served as controls. Running for 7 wk attenuated the sActRIIB-Fc-induced increase in body mass by decreasing fat mass. Running also enhanced/restored the markers of muscle oxidative capacity and autophagy in mdx mice to or above the levels of healthy mice. Voluntary running activity was decreased by sActRIIB-Fc during the first 3-4 wk correlating with increased body mass. Home cage physical activity of mice, quantified from the force plate signal, was decreased by sActRIIB-Fc the whole 7-wk treatment in sedentary mice. To understand what happens during the first weeks after sActRIIB-Fc administration, when mice are less active, healthy mice were injected with sActRIIB-Fc or PBS for 2 wk. During the sActRIIB-Fc-induced rapid 2-wk muscle growth period, oxidative capacity and autophagy were reduced, which may possibly explain the decreased running activity. These results show that increased muscle size and decreased markers of oxidative capacity and autophagy during the first weeks of myostatin/activin blocking are associated with decreased voluntary activity levels. Voluntary exercise in dystrophic mice enhances the markers of oxidative capacity and autophagy to or above the levels of healthy mice.


Subject(s)
Activin Receptors, Type II/pharmacology , Activins/antagonists & inhibitors , Autophagy/physiology , Motor Activity/physiology , Myostatin/antagonists & inhibitors , Physical Conditioning, Animal/physiology , Activin Receptors, Type II/biosynthesis , Activins/physiology , Adiposity/genetics , Adiposity/physiology , Animals , Blotting, Western , Body Weight/physiology , Citrate (si)-Synthase/metabolism , Creatine Kinase/blood , DNA/biosynthesis , DNA/isolation & purification , Eating/physiology , Hematocrit , Hemoglobins/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiology , Myostatin/physiology , Oxidation-Reduction , Tumor Necrosis Factor-alpha/metabolism
12.
Am J Physiol Endocrinol Metab ; 304(1): E41-50, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23115080

ABSTRACT

Loss of muscle mass and function occurs in various diseases. Myostatin blocking can attenuate muscle loss, but downstream signaling is not well known. Therefore, to elucidate associated signaling pathways, we used the soluble activin receptor IIb (sActRIIB-Fc) to block myostatin and activins in mice. Within 2 wk, the treatment rapidly increased muscle size as expected but decreased capillary density per area. sActRIIB-Fc increased muscle protein synthesis 1-2 days after the treatment correlating with enhanced mTORC1 signaling (phosphorylated rpS6 and S6K1, r = 0.8). Concurrently, increased REDD1 and eIF2Bε protein contents and phosphorylation of 4E-BP1 and AMPK was observed. In contrast, proangiogenic MAPK signaling and VEGF-A protein decreased. Hippo signaling has been characterized recently as a regulator of organ size and an important regulator of myogenesis in vitro. The phosphorylation of YAP (Yes-associated protein), a readout of activated Hippo signaling, increased after short- and longer-term myostatin and activin blocking and in exercised muscle. Moreover, dystrophic mdx mice had elevated phosphorylated and especially total YAP protein content. These results show that the blocking of myostatin and activins induce rapid skeletal muscle growth. This is associated with increased protein synthesis and mTORC1 signaling but decreased capillary density and proangiogenic signaling. It is also shown for the first time that Hippo signaling is activated in skeletal muscle after myostatin blocking and exercise and also in dystrophic muscle. This suggests that Hippo signaling may have a role in skeletal muscle in various circumstances.


Subject(s)
Capillaries/drug effects , Extracellular Signal-Regulated MAP Kinases/physiology , Intercellular Signaling Peptides and Proteins/pharmacology , Multiprotein Complexes/physiology , Muscle Proteins/biosynthesis , Protein Biosynthesis/drug effects , Protein Serine-Threonine Kinases/physiology , TOR Serine-Threonine Kinases/physiology , Activins/antagonists & inhibitors , Animals , Capillaries/cytology , Cell Count , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Hippo Signaling Pathway , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Inbred C57BL , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Myostatin/antagonists & inhibitors , Protein Biosynthesis/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
13.
Cell Mol Life Sci ; 70(3): 407-23, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22752156

ABSTRACT

Bone morphogenetic proteins (BMPs) are important extracellular cytokines that play critical roles in embryogenesis and tissue homeostasis. BMPs signal via transmembrane type I and type II serine/threonine kinase receptors and intracellular Smad effector proteins. BMP signaling is precisely regulated and perturbation of BMP signaling is connected to multiple diseases, including musculoskeletal diseases. In this review, we will summarize the recent progress in elucidation of BMP signal transduction, how overactive BMP signaling is involved in the pathogenesis of heterotopic ossification and Duchenne muscular dystrophy, and discuss possible therapeutic strategies for treatment of these diseases.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Muscular Dystrophy, Duchenne/metabolism , Ossification, Heterotopic/metabolism , Bone Morphogenetic Protein Receptors, Type I/metabolism , Bone Morphogenetic Protein Receptors, Type II/metabolism , Bone Morphogenetic Proteins/antagonists & inhibitors , Humans , Muscular Dystrophy, Duchenne/pathology , Ossification, Heterotopic/pathology , Signal Transduction
14.
Hum Gene Ther ; 23(12): 1269-79, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22894762

ABSTRACT

Adeno-associated virus (AAV)-U7-mediated skipping of dystrophin-exon-23 restores dystrophin expression and muscle function in the mdx mouse model of Duchenne muscular dystrophy. Soluble activin receptor IIB (sActRIIB-Fc) inhibits signaling of myostatin and homologous molecules and increases muscle mass and function of wild-type and mdx mice. We hypothesized that combined treatment with AAV-U7 and sActRIIB-Fc may synergistically improve mdx muscle function. Bioactivity of sActRIIB-Fc on skeletal muscle was first demonstrated in wild-type mice. In mdx mice we show that AAV-U7-mediated dystrophin restoration improved specific muscle force and resistance to eccentric contractions when applied alone. Treatment of mdx mice with sActRIIB-Fc increased body weight, muscle mass and myofiber size, but had little effect on muscle function. Combined treatment stimulated muscle growth comparable to the effect of sActRIIB-Fc alone and dystrophin rescue was similar to AAV-U7 alone. Moreover, combined treatment improved maximal tetanic force and the resistance to eccentric contraction to similar extent as AAV-U7 alone. In conclusion, combination of dystrophin exon skipping with sActRIIB-Fc brings together benefits of each treatment; however, we failed to evidence a clear synergistic effect on mdx muscle function.


Subject(s)
Activin Receptors, Type II/metabolism , Dependovirus/genetics , Dystrophin/genetics , Genetic Therapy/methods , Muscular Dystrophy, Animal/therapy , Activin Receptors, Type II/genetics , Animals , Body Weight , Dystrophin/metabolism , Exons , Genetic Vectors/genetics , Genetic Vectors/pharmacology , Mice , Mice, Inbred C57BL , Mice, Inbred mdx , Muscle Contraction/genetics , Muscle, Skeletal/growth & development , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Animal/physiopathology
15.
Curr Gene Ther ; 12(3): 245-59, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22554312

ABSTRACT

Since the discovery of the myostatin/ActRIIB signaling pathway 15 years ago, numerous strategies were developed to block its inhibitory function during skeletal muscle growth. Accumulating evidence demonstrates that abrogation of myostatin/ActRIIB signaling ameliorates pathology and function of dystrophic muscle in animal models for Duchenne muscular dystrophy (DMD). Therapeutic trials in healthy man and muscular dystrophy patients suggest feasibility of blockade strategies for potential clinical use. However, many key questions on the effect of myostatin/ActRIIB blockade remain unresolved; such as the underlying molecular mechanism that triggers muscle growth, the effect on muscle regeneration and adult muscle stem cell regulation and whether it causes long term metabolic alterations. Current therapeutic strategies aim to systemically abrogate myostatin/ActRIIB signaling. Although this ensures widespread effect on musculature, it also interferes with ActRIIB signaling in other tissues than skeletal muscle, thereby risking adverse effects. This review discusses current knowledge on myostatin/ActRIIB signaling and its potential value as a therapeutic target for DMD.


Subject(s)
Activin Receptors, Type II , Molecular Targeted Therapy , Muscular Dystrophy, Duchenne , Myostatin/antagonists & inhibitors , Activin Receptors, Type II/antagonists & inhibitors , Activin Receptors, Type II/genetics , Activin Receptors, Type II/metabolism , Animals , Clinical Trials, Phase II as Topic , Humans , Mice , Mice, Inbred mdx , Muscle, Skeletal/growth & development , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Muscular Dystrophy, Duchenne/genetics , Muscular Dystrophy, Duchenne/metabolism , Muscular Dystrophy, Duchenne/therapy , Myostatin/genetics , Myostatin/metabolism , Signal Transduction
16.
Cardiovasc Res ; 94(3): 439-49, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22419669

ABSTRACT

AIM: Treatment of disorders of the sinus node or the atrioventricular node requires insights into the molecular mechanisms of development and homoeostasis of these pacemaker tissues. In the developing heart, transcription factor TBX3 is required for pacemaker and conduction system development. Here, we explore the role of TBX3 in the adult heart and investigate whether TBX3 is able to reprogramme terminally differentiated working cardiomyocytes into pacemaker cells. METHODS AND RESULTS: TBX3 expression was ectopically induced in cardiomyocytes of adult transgenic mice using tamoxifen. Expression analysis revealed an efficient switch from the working myocardial expression profile to that of the pacemaker myocardium. This included suppression of genes encoding gap junction subunits (Cx40, Cx43), the cardiac Na(+) channel (Na(V)1.5; I(Na)), and inwardly rectifying K(+) ion channels (K(ir) genes; I(K1)). Concordantly, we observed conduction slowing in these hearts and reductions in I(Na) and I(K1) in cardiomyocytes isolated from these hearts. The reduction in I(K1) resulted in a more depolarized maximum diastolic potential, thus enabling spontaneous diastolic depolarization. Neither ectopic pacemaker activity nor pacemaker current I(f) was observed. Lentiviral expression of TBX3 in ventricular cardiomyocytes resulted in conduction slowing and development of heterogeneous phenotypes, including depolarized and spontaneously active cardiomyocytes. CONCLUSIONS: TBX3 reprogrammes terminally differentiated working cardiomyocytes and induces important pacemaker properties. The ability of TBX3 to reduce intercellular coupling to overcome current-to-load mismatch and the ability to reduce I(K1) density to enable diastolic depolarization are promising TBX3 characteristics that may facilitate biological pacemaker formation strategies.


Subject(s)
Biological Clocks/genetics , Cell Differentiation , Gene Expression Regulation, Developmental , Myocytes, Cardiac/metabolism , T-Box Domain Proteins/metabolism , Animals , Ion Channels/genetics , Ion Channels/metabolism , Mice , Mice, Transgenic , Myocytes, Cardiac/cytology , Sinoatrial Node/metabolism , T-Box Domain Proteins/genetics
17.
Neuromuscul Disord ; 22(5): 406-17, 2012 May.
Article in English | MEDLINE | ID: mdl-22284942

ABSTRACT

The genetic defect of mdx mice resembles that of Duchenne muscular dystrophy, although their functional performance and life expectancy is nearly normal. By contrast, mice lacking utrophin and dystrophin (mdx/utrn -/-) are severely affected and die prematurely. Mice with one utrophin allele (mdx/utrn +/-) are more severely affected than mdx mice, but outlive mdx/utrn -/- mice. We subjected mdx/utrn +/+, +/-, -/- and wild type males to a 12week functional test regime of four different functional tests. Mdx/utrn +/+ and +/- mice completed the regime, while mdx/utrn -/- mice died prematurely. Mdx/utrn +/- mice performed significantly worse compared to mdx/utrn +/+ mice in functional tests. Creatine kinase levels, percentage of fibrotic/necrotic tissue, morphology of neuromuscular synapses and expression of biomarker genes were comparable, whereas mdx/utrn +/- and -/- mice had increased levels of regenerating fibers. This makes mdx/utrn +/- mice valuable for testing the benefit of potential therapies on muscle function parameters.


Subject(s)
Dystrophin/metabolism , Motor Activity , Muscle, Skeletal/pathology , Muscular Dystrophy, Animal/genetics , Muscular Dystrophy, Duchenne/genetics , Utrophin/metabolism , Animals , Disease Models, Animal , Dystrophin/deficiency , Mice , Mice, Inbred mdx , Mice, Knockout , Muscle, Skeletal/metabolism , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Animal/physiopathology , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/physiopathology , Survival Analysis , Utrophin/deficiency
18.
FASEB J ; 26(4): 1462-72, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22202673

ABSTRACT

The transforming growth factor (TGF)-ß family member myostatin is an important regulator of myoblast, adipocyte, and fibroblast growth and differentiation, but the signaling mechanisms remain to be established. We therefore determined the contribution of myostatin type I receptors activin receptor-like kinase-4 (ALK4) and -5 (ALK5) and different coreceptors in C2C12 myoblasts, C3H10T1/2 mesenchymal stem cells, and 3T3-L1 fibroblasts, as well as in primary myoblast and fibroblasts. We performed siRNA-mediated knockdown of each receptor and measured signaling activity using Smad3-dependent luciferase and Smad2 phosphorylation assays with nontargeting siRNA as control. We find that myostatin utilizes ALK4 in myoblasts, whereas it has a preference for ALK5 in nonmyogenic cells. Notably, our results show that coreceptor Cripto is expressed in myoblasts but not in the nonmyogenic cells and that it regulates myostatin activity. More specifically, myostatin requires Cripto in myoblasts, whereas Cripto represses activin activity and TGF-ß signaling is Cripto independent. Cripto-mediated myostatin signaling is dependent on both epidermal growth factor (EGF)-like and Cripto-FRL1-cryptic (CFC) domains, whereas activin signaling is solely conferred by the CFC domain. Furthermore, Cripto down-regulation enhances myoblast differentiation, showing its importance in myostatin signaling. Together, our results identify a molecular mechanism that explains the cell-type specific aspects of signaling by myostatin and other TGF-ß family members.


Subject(s)
Activin Receptors, Type I/metabolism , Fibroblasts/physiology , Mesenchymal Stem Cells/physiology , Myoblasts/physiology , Myostatin/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/physiology , Activin Receptors/metabolism , Activin Receptors, Type I/genetics , Activins/metabolism , Animals , Cell Differentiation/physiology , Cell Line , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , Fibroblasts/cytology , Humans , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Mesenchymal Stem Cells/cytology , Mice , Mice, Inbred C57BL , Myoblasts/cytology , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
19.
Cell Mol Life Sci ; 69(8): 1377-89, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22130515

ABSTRACT

A key step in heart development is the coordinated development of the atrioventricular canal (AVC), the constriction between the atria and ventricles that electrically and physically separates the chambers, and the development of the atrioventricular valves that ensure unidirectional blood flow. Using knock-out and inducible overexpression mouse models, we provide evidence that the developmentally important T-box factors Tbx2 and Tbx3, in a functionally redundant manner, maintain the AVC myocardium phenotype during the process of chamber differentiation. Expression profiling and ChIP-sequencing analysis of Tbx3 revealed that it directly interacts with and represses chamber myocardial genes, and induces the atrioventricular pacemaker-like phenotype by activating relevant genes. Moreover, mutant mice lacking 3 or 4 functional alleles of Tbx2 and Tbx3 failed to form atrioventricular cushions, precursors of the valves and septa. Tbx2 and Tbx3 trigger development of the cushions through a regulatory feed-forward loop with Bmp2, thus providing a mechanism for the co-localization and coordination of these important processes in heart development.


Subject(s)
Endocardial Cushions/embryology , Gene Expression Regulation, Developmental , T-Box Domain Proteins/metabolism , Animals , Base Sequence , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Endocardial Cushions/metabolism , Humans , Mice , Mice, Knockout , Molecular Sequence Data , Myocardium/metabolism , Rats , T-Box Domain Proteins/genetics , Up-Regulation
20.
BMC Med Genomics ; 4: 36, 2011 Apr 20.
Article in English | MEDLINE | ID: mdl-21507246

ABSTRACT

BACKGROUND: Myostatin is a potent muscle growth inhibitor that belongs to the Transforming Growth Factor-ß (TGF-ß) family. Mutations leading to non functional myostatin have been associated with hypermuscularity in several organisms. By contrast, Duchenne muscular dystrophy (DMD) is characterized by a loss of muscle fibers and impaired regeneration. In this study, we aim to knockdown myostatin by means of exon skipping, a technique which has been successfully applied to reframe the genetic defect of dystrophin gene in DMD patients. METHODS: We targeted myostatin exon 2 using antisense oligonucleotides (AON) in healthy and DMD-derived myotubes cultures. We assessed the exon skipping level, transcriptional expression of myostatin and its target genes, and combined myostatin and several dystrophin AONs. These AONs were also applied in the mdx mice models via intramuscular injections. RESULTS: Myostatin AON induced exon 2 skipping in cell cultures and to a lower extent in the mdx mice. It was accompanied by decrease in myostatin mRNA and enhanced MYOG and MYF5 expression. Furthermore, combination of myostatin and dystrophin AONs induced simultaneous skipping of both genes. CONCLUSIONS: We conclude that two AONs can be used to target two different genes, MSTN and DMD, in a straightforward manner. Targeting multiple ligands of TGF-beta family will be more promising as adjuvant therapies for DMD.


Subject(s)
Dystrophin/genetics , Exons/genetics , Muscular Dystrophy, Duchenne/genetics , Myostatin/genetics , Animals , Cell Differentiation/drug effects , Cells, Cultured , Gene Expression Regulation/drug effects , Humans , Mice , Mice, Inbred mdx , Muscle Development/drug effects , Muscle Development/genetics , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscular Dystrophy, Animal/genetics , Myoblasts/drug effects , Myoblasts/metabolism , Myoblasts/pathology , Myostatin/metabolism , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL