Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Rep ; 21(11): 3003-3011, 2017 Dec 12.
Article in English | MEDLINE | ID: mdl-29241530

ABSTRACT

ATP citrate lyase (ACL) plays a key role in regulating mitochondrial function, as well as glucose and lipid metabolism in skeletal muscle. We report here that ACL silencing impairs myoblast and satellite cell (SC) differentiation, and it is accompanied by a decrease in fast myosin heavy chain isoforms and MYOD. Conversely, overexpression of ACL enhances MYOD levels and promotes myogenesis. Myogenesis is dependent on transcriptional but also other mechanisms. We show that ACL regulates the net amount of acetyl groups available, leading to alterations in acetylation of H3(K9/14) and H3(K27) at the MYOD locus, thus increasing MYOD expression. ACL overexpression in murine skeletal muscle leads to improved regeneration after cardiotoxin-mediated damage. Thus, our findings suggest a mechanism for regulating SC differentiation and enhancing regeneration, which might be exploited for devising therapeutic approaches for treating skeletal muscle disease.


Subject(s)
ATP Citrate (pro-S)-Lyase/genetics , Histones/genetics , Muscle, Skeletal/metabolism , MyoD Protein/genetics , Regeneration/genetics , Satellite Cells, Skeletal Muscle/metabolism , ATP Citrate (pro-S)-Lyase/metabolism , Acetylation , Animals , Cardiotoxins/toxicity , Cell Differentiation , Gene Expression Regulation , Histones/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/metabolism , Muscle Development/genetics , Muscle, Skeletal/cytology , Muscle, Skeletal/drug effects , MyoD Protein/metabolism , Primary Cell Culture , Satellite Cells, Skeletal Muscle/cytology , Satellite Cells, Skeletal Muscle/drug effects , Signal Transduction , Transcription, Genetic
2.
Skelet Muscle ; 6: 26, 2016.
Article in English | MEDLINE | ID: mdl-27462398

ABSTRACT

BACKGROUND: Cachexia affects the majority of patients with advanced cancer and is associated with reduced treatment tolerance, response to therapy, quality of life, and life expectancy. Cachectic patients with advanced cancer often receive anti-cancer therapies against their specific cancer type as a standard of care, and whether specific ActRII inhibition is efficacious when combined with anti-cancer agents has not been elucidated yet. METHODS: In this study, we evaluated interactions between ActRII blockade and anti-cancer agents in CT-26 mouse colon cancer-induced cachexia model. CDD866 (murinized version of bimagrumab) is a neutralizing antibody against the activin receptor type II (ActRII) preventing binding of ligands such as myostatin and activin A, which are involved in cancer cachexia. CDD866 was evaluated in association with cisplatin as a standard cytotoxic agent or with everolimus, a molecular-targeted agent against mammalian target of rapamycin (mTOR). In the early studies, the treatment effect on cachexia was investigated, and in the additional studies, the treatment effect on progression of cancer and the associated cachexia was evaluated using body weight loss or tumor volume as interruption criteria. RESULTS: Cisplatin accelerated body weight loss and tended to exacerbate skeletal muscle loss in cachectic animals, likely due to some toxicity of this anti-cancer agent. Administration of CDD866 alone or in combination with cisplatin protected from skeletal muscle weight loss compared to animals receiving only cisplatin, corroborating that ActRII inhibition remains fully efficacious under cisplatin treatment. In contrast, everolimus treatment alone significantly protected the tumor-bearing mice against skeletal muscle weight loss caused by CT-26 tumor. CDD866 not only remains efficacious in the presence of everolimus but also showed a non-significant trend for an additive effect on reversing skeletal muscle weight loss. Importantly, both combination therapies slowed down time-to-progression. CONCLUSIONS: Anti-ActRII blockade is an effective intervention against cancer cachexia providing benefit even in the presence of anti-cancer therapies. Co-treatment comprising chemotherapies and ActRII inhibitors might constitute a promising new approach to alleviate chemotherapy- and cancer-related wasting conditions and extend survival rates in cachectic cancer patients.


Subject(s)
Activin Receptors, Type II/antagonists & inhibitors , Activin Receptors, Type II/metabolism , Antibodies, Blocking/administration & dosage , Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents/administration & dosage , Cachexia/prevention & control , Colonic Neoplasms/complications , Activin Receptors, Type II/immunology , Animals , Antibodies, Monoclonal, Humanized , Body Weight/drug effects , Cachexia/etiology , Cisplatin/administration & dosage , Disease Models, Animal , Disease Progression , Everolimus/administration & dosage , Male , Mice , Mice, Inbred BALB C , TOR Serine-Threonine Kinases/metabolism , Tumor Burden/drug effects
3.
Mol Cell Biol ; 34(4): 619-30, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24298018

ABSTRACT

We have previously shown that activation of Gαi2, an α subunit of the heterotrimeric G protein complex, induces skeletal muscle hypertrophy and myoblast differentiation. To determine whether Gαi2 is required for skeletal muscle growth or regeneration, Gαi2-null mice were analyzed. Gαi2 knockout mice display decreased lean body mass, reduced muscle size, and impaired skeletal muscle regeneration after cardiotoxin-induced injury. Short hairpin RNA (shRNA)-mediated knockdown of Gαi2 in satellite cells (SCs) leads to defective satellite cell proliferation, fusion, and differentiation ex vivo. The impaired differentiation is consistent with the observation that the myogenic regulatory factors MyoD and Myf5 are downregulated upon knockdown of Gαi2. Interestingly, the expression of microRNA 1 (miR-1), miR-27b, and miR-206, three microRNAs that have been shown to regulate SC proliferation and differentiation, is increased by a constitutively active mutant of Gαi2 [Gαi2(Q205L)] and counterregulated by Gαi2 knockdown. As for the mechanism, this study demonstrates that Gαi2(Q205L) regulates satellite cell differentiation into myotubes in a protein kinase C (PKC)- and histone deacetylase (HDAC)-dependent manner.


Subject(s)
Cell Differentiation/genetics , Cell Proliferation , GTP-Binding Protein alpha Subunit, Gi2/genetics , Muscle Development/genetics , Muscle, Skeletal/metabolism , Regeneration/genetics , Satellite Cells, Skeletal Muscle/metabolism , Animals , Cell Differentiation/physiology , Cells, Cultured , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Development/physiology , Muscle, Skeletal/cytology , Myoblasts/cytology , Myoblasts/metabolism , Satellite Cells, Skeletal Muscle/pathology , Signal Transduction/genetics , Signal Transduction/physiology
4.
Mol Cell Biol ; 34(4): 606-18, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24298022

ABSTRACT

The myostatin/activin type II receptor (ActRII) pathway has been identified to be critical in regulating skeletal muscle size. Several other ligands, including GDF11 and the activins, signal through this pathway, suggesting that the ActRII receptors are major regulatory nodes in the regulation of muscle mass. We have developed a novel, human anti-ActRII antibody (bimagrumab, or BYM338) to prevent binding of ligands to the receptors and thus inhibit downstream signaling. BYM338 enhances differentiation of primary human skeletal myoblasts and counteracts the inhibition of differentiation induced by myostatin or activin A. BYM338 prevents myostatin- or activin A-induced atrophy through inhibition of Smad2/3 phosphorylation, thus sparing the myosin heavy chain from degradation. BYM338 dramatically increases skeletal muscle mass in mice, beyond sole inhibition of myostatin, detected by comparing the antibody with a myostatin inhibitor. A mouse version of the antibody induces enhanced muscle hypertrophy in myostatin mutant mice, further confirming a beneficial effect on muscle growth beyond myostatin inhibition alone through blockade of ActRII ligands. BYM338 protects muscles from glucocorticoid-induced atrophy and weakness via prevention of muscle and tetanic force losses. These data highlight the compelling therapeutic potential of BYM338 for the treatment of skeletal muscle atrophy and weakness in multiple settings.


Subject(s)
Activin Receptors, Type II/immunology , Activins/metabolism , Antibodies, Blocking/pharmacology , Antibodies, Monoclonal/pharmacology , Hypertrophy/metabolism , Myoblasts, Skeletal/metabolism , Activin Receptors, Type II/metabolism , Animals , Antibodies, Blocking/metabolism , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal, Humanized , Atrophy/immunology , Atrophy/metabolism , Cell Differentiation/physiology , Humans , Hypertrophy/pathology , Mice , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myoblasts, Skeletal/immunology , Signal Transduction/physiology , Smad2 Protein/metabolism , Smad3 Protein/metabolism
5.
Sci Signal ; 4(201): ra80, 2011 Nov 29.
Article in English | MEDLINE | ID: mdl-22126963

ABSTRACT

Skeletal muscle atrophy results in loss of strength and an increased risk of mortality. We found that lysophosphatidic acid, which activates a G protein (heterotrimeric guanine nucleotide-binding protein)-coupled receptor, stimulated skeletal muscle hypertrophy through activation of Gα(i2). Expression of a constitutively active mutant of Gα(i2) stimulated myotube growth and differentiation, effects that required the transcription factor NFAT (nuclear factor of activated T cells) and protein kinase C. In addition, expression of the constitutively active Gα(i2) mutant inhibited atrophy caused by the cachectic cytokine TNFα (tumor necrosis factor-α) by blocking an increase in the abundance of the mRNA encoding the E3 ubiquitin ligase MuRF1 (muscle ring finger 1). Gα(i2) activation also enhanced muscle regeneration and caused a switch to oxidative fibers. Our study thus identifies a pathway that promotes skeletal muscle hypertrophy and differentiation and demonstrates that Gα(i2)-induced signaling can act as a counterbalance to MuRF1-mediated atrophy, indicating that receptors that act through Gα(i2) might represent potential targets for preventing skeletal muscle wasting.


Subject(s)
Cell Differentiation , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Myoblasts, Skeletal/enzymology , Regeneration , Signal Transduction , Animals , Enzyme Activation/genetics , GTP-Binding Protein alpha Subunits, G12-G13/genetics , HEK293 Cells , Humans , Hypertrophy/enzymology , Hypertrophy/genetics , Hypertrophy/pathology , Mice , Mice, Transgenic , Muscle Proteins/genetics , Muscle Proteins/metabolism , Muscular Atrophy/enzymology , Muscular Atrophy/genetics , Muscular Atrophy/pathology , Mutation , Myoblasts, Skeletal/pathology , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Protein Kinase C/genetics , Protein Kinase C/metabolism , Tripartite Motif Proteins , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...