Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
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.
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
3.
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
4.
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
5.
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
6.
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
7.
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
8.
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
9.
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
10.
Neurobiol Dis ; 41(2): 353-60, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20940052

ABSTRACT

Duchenne Muscular Dystrophy (DMD) is an X-linked lethal muscle wasting disease characterized by muscle fiber degeneration and necrosis. The progressive pathology of DMD can be explained by an insufficient regenerative response resulting in fibrosis and adipose tissue formation. BMPs are known to inhibit myogenic differentiation and in a previous study we found an increased expression of a BMP family member BMP4 in DMD myoblasts. The aim of the current study was therefore to investigate whether inhibition of BMP signaling could be beneficial for myoblast differentiation and muscle regeneration processes in a DMD context. All tested BMP inhibitors, Noggin, dorsomorphin and LDN-193189, were able to accelerate and enhance myogenic differentiation. However, dorsomorphin repressed both BMP and TGFß signaling and was found to be toxic to primary myoblast cell cultures. In contrast, Noggin was found to be a potent and selective BMP inhibitor and was therefore tested in vivo in a DMD mouse model. Local adenoviral-mediated overexpression of Noggin in muscle resulted in an increased expression of the myogenic regulatory genes Myog and Myod1 and improved muscle histology. In conclusion, our results suggest that repression of BMP signaling may constitute an attractive adjunctive therapy for DMD patients.


Subject(s)
Bone Morphogenetic Proteins/antagonists & inhibitors , Cell Differentiation/drug effects , Disease Models, Animal , Muscle, Skeletal/drug effects , Muscular Dystrophy, Duchenne/drug therapy , Muscular Dystrophy, Duchenne/genetics , Myoblasts/pathology , Phenotype , Animals , Bone Morphogenetic Proteins/metabolism , Carrier Proteins/genetics , Carrier Proteins/pharmacology , Carrier Proteins/therapeutic use , Cell Differentiation/genetics , Cell Line , Humans , Male , Mice , Mice, Inbred mdx , Mice, Knockout , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Dystrophy, Duchenne/pathology , Myoblasts/drug effects , Myoblasts/metabolism
11.
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
12.
Circ Res ; 100(3): 354-62, 2007 Feb 16.
Article in English | MEDLINE | ID: mdl-17234970

ABSTRACT

The sinoatrial node, which resides at the junction of the right atrium and the superior caval vein, contains specialized myocardial cells that initiate the heart beat. Despite this fundamental role in heart function, the embryonic origin and mechanisms of localized formation of the sinoatrial node have not been defined. Here we show that subsequent to the formation of the Nkx2-5-positive heart tube, cells bordering the inflow tract of the heart tube give rise to the Nkx2-5-negative myocardial cells of the sinoatrial node and the sinus horns. Using genetic models, we show that as the myocardium of the heart tube matures, Nkx2-5 suppresses pacemaker channel gene Hcn4 and T-box transcription factor gene Tbx3, thereby enforcing a progressive confinement of their expression to the forming Nkx2-5-negative sinoatrial node and sinus horns. Thus, Nkx2-5 is essential for establishing a gene expression border between the atrium and sinoatrial node. Tbx3 was found to suppress chamber differentiation, providing an additional mechanism by which the Tbx3-positive sinoatrial node is shielded from differentiating into atrial myocardium. Pitx2c-deficient fetuses form sinoatrial nodes with indistinguishable molecular signatures at both the right and left sinuatrial junction, indicating that Pitx2c functions within the left/right pathway to suppress a default program for sinuatrial node formation on the left. Our molecular pathway provides a mechanism for how pacemaker activity becomes progressively relegated to the most recently added components of the venous pole of the heart and, ultimately, to the junction of the right atrium and superior caval vein.


Subject(s)
Body Patterning/physiology , Gene Expression Regulation, Developmental/physiology , Heart Atria/embryology , Heart Ventricles/embryology , Homeodomain Proteins/physiology , Ion Channels/biosynthesis , Sinoatrial Node/embryology , T-Box Domain Proteins/physiology , Transcription Factors/physiology , Animals , Atrial Natriuretic Factor , Biomarkers , Body Patterning/genetics , Cardiac Myosins/biosynthesis , Cardiac Myosins/genetics , Connexins/biosynthesis , Connexins/genetics , Cyclic Nucleotide-Gated Cation Channels , Gene Expression Regulation, Developmental/genetics , Genes, Reporter , Homeobox Protein Nkx-2.5 , Homeodomain Proteins/genetics , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels , Imaging, Three-Dimensional , In Situ Hybridization , Ion Channels/genetics , Mice , Mice, Knockout , Mice, Transgenic , Myocardium/metabolism , Myosin Light Chains/biosynthesis , Myosin Light Chains/genetics , Natriuretic Peptide, C-Type/biosynthesis , Natriuretic Peptide, C-Type/genetics , Protein Precursors/biosynthesis , Protein Precursors/genetics , Recombinant Fusion Proteins/physiology , Sinoatrial Node/cytology , T-Box Domain Proteins/biosynthesis , T-Box Domain Proteins/genetics , Transcription Factors/deficiency , Transcription Factors/genetics , Troponin I/biosynthesis , Troponin I/genetics , Gap Junction alpha-5 Protein , Homeobox Protein PITX2
13.
Cardiovasc Res ; 62(3): 489-99, 2004 Jun 01.
Article in English | MEDLINE | ID: mdl-15158141

ABSTRACT

OBJECTIVE: The molecular mechanisms that regulate the formation of the conduction system are poorly understood. We studied the developmental expression pattern and functional aspects of the T-box transcription factor Tbx3, a novel marker for the murine central conduction system (CCS). METHODS: The patterns of expression of Tbx3, and of Cx40, Cx43, and Nppa, which are markers for atrial and ventricular chamber-type myocardium in the developing heart, were analyzed in mice by in situ hybridization and three-dimensional reconstruction analysis. The function of Tbx3 in regulating Nppa and Cx40 promoter activity was studied in vitro. RESULTS: In the formed heart, Tbx3 is expressed in the sinoatrial node (SAN), atrioventricular node (AVN), bundle and proximal bundle branches (BBs), as well as the internodal regions and the atrioventricular region. Throughout cardiac development, Tbx3 is expressed in an uninterrupted myocardial domain that extends from the sinoatrial node to the atrioventricular region. This expression domain is present in the looping heart tube from E8.5 onwards. Expression of the chamber-type myocardial markers is specifically absent from the Tbx3 expression domain. Tbx3 is able to repress Nppa and Cx40 promoter activity and abolish the synergistic activation of the Nppa promoter by Tbx5 and Nkx2.5. CONCLUSION: We identified the T-box transcription factor Tbx3 as a novel and accurate marker for the central conduction system. Our analysis implicates a role for Tbx3 in repressing a chamber-specific program of gene expression in regions from which the components of the central conduction system are subsequently formed.


Subject(s)
Gene Expression Regulation, Developmental , Heart Conduction System/embryology , T-Box Domain Proteins/genetics , Animals , Atrial Natriuretic Factor , COS Cells , Cell Line , Connexins/genetics , Gene Expression , Genetic Markers , Gestational Age , Heart Conduction System/chemistry , Image Processing, Computer-Assisted , In Situ Hybridization , Mice , Mice, Inbred Strains , Myocardium/chemistry , Natriuretic Peptide, C-Type , Promoter Regions, Genetic , Protein Precursors , T-Box Domain Proteins/analysis , Gap Junction alpha-5 Protein
14.
Fundam Clin Pharmacol ; 18(1): 45-50, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14748753

ABSTRACT

To investigate the involvement of the mitogen-activated protein kinase (MAPK) family of extracellular signal-regulated kinase (ERK) 1 and 2 (MAPKerk1/2) in the vasopressin-mediated vasoconstriction in the rat aorta. Vasopressin-induced vasoconstriction was measured in isolated rat thoracic aortae in the presence or absence of MAPKerk1/2 kinase (MKKmek1/2) inhibitors. Thereafter the MAPKerk1/2 phosphorylation in the rat aorta was quantified using Western blot analysis. Vasopressin (1-300 nm) induced a concentration-dependent vasoconstriction, which could be inhibited concentration dependently by the selective MKKmek1/2 inhibitors, PD 98059 (10 and 100 microm) and U 0126 (10 and 100 microm). Western blot analysis revealed a 2.7 +/- 0.6-fold increase in the MAPKerk1/2 phosphorylation induced by vasopressin (300 nm). This phosphorylation could be dose dependently prevented by both PD 98059 (100 microm) and U 0126 (10 and 100 microm). These results indicate that vasoconstriction induced by vasopressin is partly regulated by the MAPKerk1/2 pathway.


Subject(s)
Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Vasoconstrictor Agents/pharmacology , Vasopressins/pharmacology , Animals , Aorta, Thoracic/drug effects , Blotting, Western , Butadienes/pharmacology , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , In Vitro Techniques , Male , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Muscle Contraction/drug effects , Muscle, Smooth, Vascular/drug effects , Myocardial Contraction/drug effects , Nitriles/pharmacology , Phosphorylation , Potassium/pharmacology , Rats , Rats, Wistar , Signal Transduction/drug effects , Signal Transduction/physiology
15.
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
16.
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
17.
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
18.
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
19.
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
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