Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
Front Physiol ; 13: 1044488, 2022.
Article in English | MEDLINE | ID: mdl-36467705

ABSTRACT

Musculoskeletal diseases are a leading contributor to mobility disability worldwide. Since the majority of patients with musculoskeletal diseases present with associated muscle weakness, treatment approaches typically comprise an element of resistance training to restore physical strength. The health-promoting effects of resistance exercise are mediated via complex, multifarious mechanisms including modulation of systemic and local inflammation. Here we investigated whether targeted inhibition of the chemerin pathway, which largely controls inflammatory processes via chemokine-like receptor 1 (CMKLR1), can improve skeletal muscle function. Using genetically modified mice, we demonstrate that blockade of CMKLR1 transiently increases maximal strength during growth, but lastingly decreases strength endurance. In-depth analyses of the underlying long-term adaptations revealed microscopic alterations in the number of Pax7-positive satellite cells, as well as molecular changes in genes governing myogenesis and calcium handling. Taken together, these data provide evidence of a critical role for CMKLR1 in regulating skeletal muscle function by modulating the regenerative and contractile properties of muscle tissue. CMKLR1 antagonists are increasingly viewed as therapeutic modalities for a variety of diseases (e.g., psoriasis, metabolic disorders, and multiple sclerosis). Our findings thus have implications for the development of novel drug substances that aim at targeting the chemerin pathway for musculoskeletal or other diseases.

2.
Cell Rep ; 29(6): 1539-1554.e7, 2019 Nov 05.
Article in English | MEDLINE | ID: mdl-31693894

ABSTRACT

Age-related loss of skeletal muscle innervation by motor neurons leads to impaired neuromuscular function and is a well-established clinical phenomenon. However, the underlying pathogenesis remains unclear. Studying mice, we find that the number of motor units (MUs) can be maintained by counteracting neurotoxic microglia in the aged spinal cord. We observe that marked innervation changes, detected by motor unit number estimation (MUNE), occur prior to loss of muscle function in aged mice. This coincides with gene expression changes indicative of neuronal remodeling and microglial activation in aged spinal cord. Voluntary exercise prevents loss of MUs and reverses microglia activation. Depleting microglia by CSF1R inhibition also prevents the age-related decline in MUNE and neuromuscular junction disruption, implying a causal link. Our results suggest that age-related changes in spinal cord microglia contribute to neuromuscular decline in aged mice and demonstrate that removal of aged neurotoxic microglia can prevent or reverse MU loss.


Subject(s)
Aging/metabolism , Microglia/metabolism , Motor Neurons/metabolism , Physical Conditioning, Animal/physiology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Aging/pathology , Animals , Cell Line , Databases, Genetic , Humans , Induced Pluripotent Stem Cells , Macrophages , Male , Mice , Mice, Inbred C57BL , Microglia/enzymology , Microglia/physiology , Motor Neurons/cytology , Motor Neurons/pathology , Muscle, Skeletal/metabolism , Muscle, Skeletal/physiopathology , Neuromuscular Junction/metabolism , Neuronal Plasticity/genetics , RNA-Seq , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Spinal Cord/enzymology , Spinal Cord/metabolism , Spinal Cord/physiopathology
3.
Cell Rep ; 29(3): 749-763.e12, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31618641

ABSTRACT

HDAC4, a class IIa histone deacetylase, is upregulated in skeletal muscle in response to denervation-induced atrophy. When HDAC4 is deleted postnatally, mice are partially protected from denervation. Despite the name "histone" deacetylase, HDAC4 demonstrably deacetylates cytosolic and non-histone nuclear proteins. We developed potent and selective class IIa HDAC inhibitors. Using these tools and genetic knockdown, we identified three previously unidentified substrates of HDAC4: myosin heavy chain, peroxisome proliferator-activated receptor gamma co-activator 1alpha (PGC-1α), and heat shock cognate 71 kDa protein (Hsc70). HDAC4 inhibition almost completely prevented denervation-induced loss of myosin heavy chain isoforms and blocked the action of their E3 ligase, MuRF1. PGC-1α directly interacts with class IIa HDACs; selective inhibitors increased PGC-1α protein in muscles. Hsc70 deacetylation by HDAC4 affects its chaperone activity. Through these endogenous HDAC4 substrates, we identified several muscle metabolic pathways that are regulated by class IIa HDACs, opening up new therapeutic options to treat skeletal muscle disorders and potentially other disease where these specific pathways are affected.


Subject(s)
HSC70 Heat-Shock Proteins/metabolism , Histone Deacetylases/metabolism , Muscle, Skeletal/metabolism , Myosin Heavy Chains/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Acetylation , Animals , Cells, Cultured , Female , Gene Expression , Histone Deacetylases/chemistry , Histone Deacetylases/genetics , Male , Mice , Mice, Knockout , Muscle Proteins/metabolism , Myoblasts/cytology , Myoblasts/metabolism , Myosin Heavy Chains/genetics , Protein Binding , Protein Isoforms/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Glucocorticoid/metabolism , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism
4.
Biochem Biophys Res Commun ; 503(4): 2415-2420, 2018 09 18.
Article in English | MEDLINE | ID: mdl-29969629

ABSTRACT

The majority of patients with advanced cancer suffer from cachexia, a systemic wasting syndrome, which subsequently impacts the tolerance to anti-cancer treatments, response to therapy, quality of life, and eventually, survival. Despite a high unmet medical need, there is currently no specific remedy available for an effective treatment of cachexia and its sequelae. A key feature of cachexia is the inexorable loss of skeletal muscle mass, which constitutes a main contributor to body weight loss and progressive functional impairments. Therefore, it's crucial to identify early readouts to detect and monitor the loss of muscle mass and function to initiate appropriate treatments timely. Here, we describe experimental cancer models using mouse (syngeneic) or human (xenograft) cancer cell lines with a rapid onset of tumor growth and cachexia. These models are easier to establish, monitor and reproduce compared to the genetically engineered mouse models currently available. Moreover, we establish readouts such as hind limb muscle mass and volume, as well as evoked force and food intake measurements, to allow the evaluation of potential therapeutic agents for the early treatment of cachexia and associated impairments.


Subject(s)
Cachexia/etiology , Cachexia/pathology , Muscle, Skeletal/pathology , Neoplasms/complications , Animals , Body Weight , Cachexia/diagnostic imaging , Cell Line, Tumor , Colonic Neoplasms/complications , Disease Models, Animal , Humans , Lower Extremity/diagnostic imaging , Magnetic Resonance Imaging , Melanoma/complications , Mice , Muscle, Skeletal/diagnostic imaging
5.
Nitric Oxide ; 77: 35-43, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29678764

ABSTRACT

The expression of neuronal NO synthase (nNOS) alpha- and beta-isoforms in skeletal muscle is well documented but only little information is available about their regulation/functions. Using different mouse models, we now assessed whether the expression of nNOS-isoforms in muscle fibers is related to mitochondria content/activity and regulated by peroxisome proliferator-activated receptor gamma coactivator-1alpha (PGC-1alpha). Catalytic histochemistry revealed highest nNOS-concentrations to be present in type-2 oxidative muscle fibers. Differences in mitochondrial density between nNOS-KO-mice and WT-littermates established by morphometry after transmission electron microscopy were significant in the oxidative portion of the tibialis anterior muscle (TA) but not in rectus femoris muscle (RF) indicating an nNOS-dependent mitochondrial pool in TA. Quantitative immunoblotting displayed the nNOS alpha-isoform to preponderate in those striated muscles of C57BL/6-mice that comprise of many type-2 oxidative fibers, e.g. TA, while roughly even levels of the two nNOS-isoforms were expressed in those muscles that mainly consist of type-2 glycolytic fibers, e.g. RF. Differences in citrate synthase-activity in muscle homogenates between nNOS-KO-mice and WT-littermates were positively related to nNOS alpha-isoform levels. In transgenic-mice over-expressing muscular PGC-1alpha compared to WT-littermates, immunoblotting revealed a significant shift in nNOS-expression in favor of the alpha-isoform in six out of eight striated muscles (exceptions: soleus muscle and tongue) without consistent relationship to changes in the expression of mitochondrial markers. In summary, our study demonstrated the nNOS alpha-isoform expression to be related to mitochondrial content/activity and to be up-regulated by up-stream PGC-1alpha in striated muscles, particularly in those enriched with type-2 oxidative fibers implying a functional convergence of the two signaling systems in these fibers.


Subject(s)
Mitochondria/metabolism , Muscle, Striated/metabolism , Nitric Oxide Synthase Type I/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/biosynthesis , Animals , Isoenzymes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism
6.
Mol Cell Biol ; 37(5)2017 03 01.
Article in English | MEDLINE | ID: mdl-27956698

ABSTRACT

Metallothioneins are proteins that are involved in intracellular zinc storage and transport. Their expression levels have been reported to be elevated in several settings of skeletal muscle atrophy. We therefore investigated the effect of metallothionein blockade on skeletal muscle anabolism in vitro and in vivo We found that concomitant abrogation of metallothioneins 1 and 2 results in activation of the Akt pathway and increases in myotube size, in type IIb fiber hypertrophy, and ultimately in muscle strength. Importantly, the beneficial effects of metallothionein blockade on muscle mass and function was also observed in the setting of glucocorticoid addition, which is a strong atrophy-inducing stimulus. Given the blockade of atrophy and the preservation of strength in atrophy-inducing settings, these results suggest that blockade of metallothioneins 1 and 2 constitutes a promising approach for the treatment of conditions which result in muscle atrophy.


Subject(s)
Metallothionein/metabolism , Muscle Strength/physiology , Muscle, Skeletal/metabolism , Animals , Biomarkers/metabolism , Body Weight , Cell Size , Gene Silencing , Glucocorticoids/adverse effects , Humans , Hypertrophy , Mice , Muscle Development , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle, Skeletal/pathology , Muscle, Skeletal/physiopathology , Muscular Atrophy , Organ Size , Proto-Oncogene Proteins c-akt/metabolism , Rats , Sarcopenia/metabolism , Sarcopenia/pathology , Sarcopenia/physiopathology , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Up-Regulation , Zinc/metabolism
7.
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
8.
Proc Natl Acad Sci U S A ; 110(50): 20314-9, 2013 Dec 10.
Article in English | MEDLINE | ID: mdl-24277823

ABSTRACT

Skeletal muscle mass loss and dysfunction have been linked to many diseases. Conversely, resistance exercise, mainly by activating mammalian target of rapamycin complex 1 (mTORC1), promotes skeletal muscle hypertrophy and exerts several therapeutic effects. Moreover, mTORC1, along with peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α), regulates skeletal muscle metabolism. However, it is unclear whether PGC-1α is required for skeletal muscle adaptations after overload. Here we show that although chronic overload of skeletal muscle via synergist ablation (SA) strongly induces hypertrophy and a switch toward a slow-contractile phenotype, these effects were independent of PGC-1α. In fact, SA down-regulated PGC-1α expression and led to a repression of energy metabolism. Interestingly, however, PGC-1α deletion preserved peak force after SA. Taken together, our data suggest that PGC-1α is not involved in skeletal muscle remodeling induced by SA.


Subject(s)
Energy Metabolism/physiology , Multiprotein Complexes/metabolism , Muscle, Skeletal/metabolism , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Animals , Blotting, Western , DNA Primers/genetics , Hypertrophy/metabolism , Immunohistochemistry , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Transgenic , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Phosphofructokinases/metabolism , Polymerase Chain Reaction , Succinate Dehydrogenase/metabolism
9.
Proc Natl Acad Sci U S A ; 110(21): 8738-43, 2013 May 21.
Article in English | MEDLINE | ID: mdl-23650363

ABSTRACT

The peroxisome proliferator-activated receptor-γ coactivator 1α (PGC-1α) controls metabolic adaptations. We now show that PGC-1α in skeletal muscle drives the expression of lactate dehydrogenase (LDH) B in an estrogen-related receptor-α-dependent manner. Concomitantly, PGC-1α reduces the expression of LDH A and one of its regulators, the transcription factor myelocytomatosis oncogene. PGC-1α thereby coordinately alters the composition of the LDH complex and prevents the increase in blood lactate during exercise. Our results show how PGC-1α actively coordinates lactate homeostasis and provide a unique molecular explanation for PGC-1α-mediated muscle adaptations to training that ultimately enhance exercise performance and improve metabolic health.


Subject(s)
Estrogen Receptor alpha/metabolism , Gene Expression Regulation, Enzymologic/physiology , Homeostasis/physiology , L-Lactate Dehydrogenase/biosynthesis , Lactic Acid/blood , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Trans-Activators/metabolism , Animals , Cell Line , Estrogen Receptor alpha/genetics , Isoenzymes/biosynthesis , Isoenzymes/genetics , L-Lactate Dehydrogenase/genetics , Lactate Dehydrogenase 5 , Mice , Mice, Knockout , Muscle Proteins/genetics , Muscle, Skeletal/cytology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Physical Conditioning, Animal , Trans-Activators/genetics , Transcription Factors
10.
Skelet Muscle ; 3(1): 6, 2013 Mar 06.
Article in English | MEDLINE | ID: mdl-23497627

ABSTRACT

BACKGROUND: Skeletal muscle mass is determined by the balance between protein synthesis and degradation. Mammalian target of rapamycin complex 1 (mTORC1) is a master regulator of protein translation and has been implicated in the control of muscle mass. Inactivation of mTORC1 by skeletal muscle-specific deletion of its obligatory component raptor results in smaller muscles and a lethal dystrophy. Moreover, raptor-deficient muscles are less oxidative through changes in the expression PGC-1α, a critical determinant of mitochondrial biogenesis. These results suggest that activation of mTORC1 might be beneficial to skeletal muscle by providing resistance to muscle atrophy and increasing oxidative function. Here, we tested this hypothesis by deletion of the mTORC1 inhibitor tuberous sclerosis complex (TSC) in muscle fibers. METHOD: Skeletal muscles of mice with an acute or a permanent deletion of raptor or TSC1 were examined using histological, biochemical and molecular biological methods. Response of the muscles to changes in mechanical load and nerve input was investigated by ablation of synergistic muscles or by denervation . RESULTS: Genetic deletion or knockdown of raptor, causing inactivation of mTORC1, was sufficient to prevent muscle growth and enhance muscle atrophy. Conversely, short-term activation of mTORC1 by knockdown of TSC induced muscle fiber hypertrophy and atrophy-resistance upon denervation, in both fast tibialis anterior (TA) and slow soleus muscles. Surprisingly, however, sustained activation of mTORC1 by genetic deletion of Tsc1 caused muscle atrophy in all but soleus muscles. In contrast, oxidative capacity was increased in all muscles examined. Consistently, TSC1-deficient soleus muscle was atrophy-resistant whereas TA underwent normal atrophy upon denervation. Moreover, upon overloading, plantaris muscle did not display enhanced hypertrophy compared to controls. Biochemical analysis indicated that the atrophy response of muscles was based on the suppressed phosphorylation of PKB/Akt via feedback inhibition by mTORC1 and subsequent increased expression of the E3 ubiquitin ligases MuRF1 and atrogin-1/MAFbx. In contrast, expression of both E3 ligases was not increased in soleus muscle suggesting the presence of compensatory mechanisms in this muscle. CONCLUSIONS: Our study shows that the mTORC1- and the PKB/Akt-FoxO pathways are tightly interconnected and differentially regulated depending on the muscle type. These results indicate that long-term activation of the mTORC1 signaling axis is not a therapeutic option to promote muscle growth because of its strong feedback induction of the E3 ubiquitin ligases involved in protein degradation.

11.
Diabetes ; 62(1): 85-95, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23086035

ABSTRACT

Metabolic disorders are a major burden for public health systems globally. Regular exercise improves metabolic health. Pharmacological targeting of exercise mediators might facilitate physical activity or amplify the effects of exercise. The peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) largely mediates musculoskeletal adaptations to exercise, including lipid refueling, and thus constitutes such a putative target. Paradoxically, forced expression of PGC-1α in muscle promotes diet-induced insulin resistance in sedentary animals. We show that elevated PGC-1α in combination with exercise preferentially improves glucose homeostasis, increases Krebs cycle activity, and reduces the levels of acylcarnitines and sphingosine. Moreover, patterns of lipid partitioning are altered in favor of enhanced insulin sensitivity in response to combined PGC-1α and exercise. Our findings reveal how physical activity improves glucose homeostasis. Furthermore, our data suggest that the combination of elevated muscle PGC-1α and exercise constitutes a promising approach for the treatment of metabolic disorders.


Subject(s)
Glucose/metabolism , Homeostasis , Muscle, Skeletal/metabolism , Trans-Activators/physiology , Animals , Carnitine/analogs & derivatives , Carnitine/metabolism , Cells, Cultured , Citric Acid Cycle , Fatty Acid Synthases/physiology , Lipogenesis , Male , Mice , Oxidative Phosphorylation , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Physical Conditioning, Animal , Sphingosine/pharmacology , Transcription Factors
12.
Diabetes ; 62(2): 362-72, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22961086

ABSTRACT

Catch-up growth, a risk factor for type 2 diabetes, is characterized by hyperinsulinemia and accelerated body fat recovery. Using a rat model of semistarvation-refeeding that exhibits catch-up fat, we previously reported that during refeeding on a low-fat diet, glucose tolerance is normal but insulin-dependent glucose utilization is decreased in skeletal muscle and increased in adipose tissue, where de novo lipogenic capacity is concomitantly enhanced. Here we report that isocaloric refeeding on a high-fat (HF) diet blunts the enhanced in vivo insulin-dependent glucose utilization for de novo lipogenesis (DNL) in adipose tissue. These are shown to be early events of catch-up growth that are independent of hyperphagia and precede the development of overt adipocyte hypertrophy, adipose tissue inflammation, or defective insulin signaling. These results suggest a role for enhanced DNL as a glucose sink in regulating glycemia during catch-up growth, which is blunted by exposure to an HF diet, thereby contributing, together with skeletal muscle insulin resistance, to the development of glucose intolerance. Our findings are presented as an extension of the Randle cycle hypothesis, whereby the suppression of DNL constitutes a mechanism by which dietary lipids antagonize glucose utilization for storage as triglycerides in adipose tissue, thereby impairing glucose homeostasis during catch-up growth.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Glucose/metabolism , Homeostasis/physiology , Lipogenesis/physiology , Refeeding Syndrome/metabolism , Adipocytes/pathology , Adipose Tissue/pathology , Animals , Diet, High-Fat , Hyperinsulinism/metabolism , Hyperphagia/metabolism , Hypertrophy/metabolism , Hypertrophy/pathology , Hypoglycemic Agents/pharmacology , Insulin/pharmacology , Male , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology
13.
Mol Cell Biol ; 32(24): 4913-24, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23028049

ABSTRACT

Skeletal muscle exhibits a high plasticity and accordingly can quickly adapt to different physiological and pathological stimuli by changing its phenotype largely through diverse epigenetic mechanisms. The nuclear receptor corepressor 1 (NCoR1) has the ability to mediate gene repression; however, its role in regulating biological programs in skeletal muscle is still poorly understood. We therefore studied the mechanistic and functional aspects of NCoR1 function in this tissue. NCoR1 muscle-specific knockout mice exhibited a 7.2% higher peak oxygen consumption (VO(2peak)), a 11% reduction in maximal isometric force, and increased ex vivo fatigue resistance during maximal stimulation. Interestingly, global gene expression analysis revealed a high overlap between the effects of NCoR1 deletion and peroxisome proliferator-activated receptor gamma (PPARγ) coactivator 1α (PGC-1α) overexpression on oxidative metabolism in muscle. Importantly, PPARß/δ and estrogen-related receptor α (ERRα) were identified as common targets of NCoR1 and PGC-1α with opposing effects on the transcriptional activity of these nuclear receptors. In fact, the repressive effect of NCoR1 on oxidative phosphorylation gene expression specifically antagonizes PGC-1α-mediated coactivation of ERRα. We therefore delineated the molecular mechanism by which a transcriptional network controlled by corepressor and coactivator proteins determines the metabolic properties of skeletal muscle, thus representing a potential therapeutic target for metabolic diseases.


Subject(s)
Muscle, Skeletal/physiology , Nuclear Receptor Co-Repressor 1/metabolism , Receptors, Estrogen/metabolism , Trans-Activators/metabolism , Animals , Male , Mice , Mice, Knockout , Mice, Transgenic , Models, Biological , Muscle Contraction/genetics , Muscle Contraction/physiology , Muscle, Skeletal/metabolism , Nuclear Receptor Co-Repressor 1/deficiency , Nuclear Receptor Co-Repressor 1/genetics , Oxidative Phosphorylation , Oxygen Consumption , PPAR delta/metabolism , PPAR-beta/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/genetics , Trans-Activators/antagonists & inhibitors , Trans-Activators/genetics , Transcription Factors , ERRalpha Estrogen-Related Receptor
14.
Am J Physiol Cell Physiol ; 302(1): C88-99, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21918181

ABSTRACT

Regular endurance exercise remodels skeletal muscle, largely through the peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α). PGC-1α promotes fiber type switching and resistance to fatigue. Intracellular calcium levels might play a role in both adaptive phenomena, yet a role for PGC-1α in the adaptation of calcium handling in skeletal muscle remains unknown. Using mice with transgenic overexpression of PGC-1α, we now investigated the effect of PGC-1α on calcium handling in skeletal muscle. We demonstrate that PGC-1α induces a quantitative reduction in calcium release from the sarcoplasmic reticulum by diminishing the expression of calcium-releasing molecules. Concomitantly, maximal muscle force is reduced in vivo and ex vivo. In addition, PGC-1α overexpression delays calcium clearance from the myoplasm by interfering with multiple mechanisms involved in calcium removal, leading to higher myoplasmic calcium levels following contraction. During prolonged muscle activity, the delayed calcium clearance might facilitate force production in mice overexpressing PGC-1α. Our results reveal a novel role of PGC-1α in altering the contractile properties of skeletal muscle by modulating calcium handling. Importantly, our findings indicate PGC-1α to be both down- as well as upstream of calcium signaling in this tissue. Overall, our findings suggest that in the adaptation to chronic exercise, PGC-1α reduces maximal force, increases resistance to fatigue, and drives fiber type switching partly through remodeling of calcium transients, in addition to promoting slow-type myofibrillar protein expression and adequate energy supply.


Subject(s)
Calcium/metabolism , Muscle Contraction/physiology , Muscle Fatigue/physiology , Muscle Fibers, Skeletal/physiology , Trans-Activators/physiology , Animals , Calcium/physiology , Mice , Mice, Transgenic , Muscle Fibers, Fast-Twitch/physiology , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Slow-Twitch/physiology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Random Allocation , Transcription Factors
15.
Biochem Biophys Res Commun ; 408(1): 180-5, 2011 Apr 29.
Article in English | MEDLINE | ID: mdl-21501593

ABSTRACT

The peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) enhances oxidative metabolism in skeletal muscle. Excessive lipid oxidation and electron transport chain activity can, however, lead to the accumulation of harmful metabolites and impair glucose homeostasis. Here, we investigated the effect of over-expression of PGC-1α on metabolic control and generation of insulin desensitizing agents in extensor digitorum longus (EDL), a muscle that exhibits low levels of PGC-1α in the untrained state and minimally relies on oxidative metabolism. We demonstrate that PGC-1α induces a strictly balanced substrate oxidation in EDL by concomitantly promoting the transcription of activators and inhibitors of lipid oxidation. Moreover, we show that PGC-1α enhances the potential to uncouple oxidative phosphorylation. Thereby, PGC-1α boosts elevated, yet tightly regulated oxidative metabolism devoid of side products that are detrimental for glucose homeostasis. Accordingly, PI3K activity, an early phase marker for insulin resistance, is preserved in EDL muscle. Our findings suggest that PGC-1α coordinately coactivates the simultaneous transcription of gene clusters implicated in the positive and negative regulation of oxidative metabolism and thereby increases metabolic flexibility. Thus, in mice fed a normal chow diet, over-expression of PGC-1α does not alter insulin sensitivity and the metabolic adaptations elicited by PGC-1α mimic the beneficial effects of endurance training on muscle metabolism in this context.


Subject(s)
Gene Expression Regulation , Genes, Mitochondrial , Insulin Resistance , Insulin/physiology , Mitochondria, Muscle/genetics , Muscle, Skeletal/metabolism , Trans-Activators/metabolism , Animals , Citric Acid Cycle/genetics , DNA, Mitochondrial/metabolism , Glucose/metabolism , Insulin/pharmacology , Lipid Metabolism/genetics , Mice , Mice, Transgenic , Muscle, Skeletal/drug effects , Oxidation-Reduction , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Reactive Oxygen Species/metabolism , Trans-Activators/genetics , Transcription Factors , Transcription, Genetic
16.
J Biol Chem ; 285(43): 32793-32800, 2010 Oct 22.
Article in English | MEDLINE | ID: mdl-20716531

ABSTRACT

Exercise induces a pleiotropic adaptive response in skeletal muscle, largely through peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α). PGC-1α enhances lipid oxidation and thereby provides energy for sustained muscle contraction. Its potential implication in promoting muscle refueling remains unresolved, however. Here, we investigated a possible role of elevated PGC-1α levels in skeletal muscle lipogenesis in vivo and the molecular mechanisms that underlie PGC-1α-mediated de novo lipogenesis. To this end, we studied transgenic mice with physiological overexpression of PGC-1α and human muscle biopsies pre- and post-exercise. We demonstrate that PGC-1α enhances lipogenesis in skeletal muscle through liver X receptor α-dependent activation of the fatty acid synthase (FAS) promoter and by increasing FAS activity. Using chromatin immunoprecipitation, we establish a direct interaction between PGC-1α and the liver X receptor-responsive element in the FAS promoter. Moreover, we show for the first time that increased glucose uptake and activation of the pentose phosphate pathway provide substrates for RNA synthesis and cofactors for de novo lipogenesis. Similarly, we observed increased lipogenesis and lipid levels in human muscle biopsies that were obtained post-exercise. Our findings suggest that PGC-1α coordinates lipogenesis, intramyocellular lipid accumulation, and substrate oxidation in exercised skeletal muscle in vivo.


Subject(s)
Heat-Shock Proteins/metabolism , Lipids/biosynthesis , Muscle, Skeletal/metabolism , Pentose Phosphate Pathway/physiology , Trans-Activators/metabolism , Transcription Factors/metabolism , Animals , Fatty Acid Synthase, Type I/biosynthesis , Fatty Acid Synthase, Type I/genetics , Heat-Shock Proteins/genetics , Humans , Lipids/genetics , Liver X Receptors , Mice , Mice, Transgenic , Muscle, Skeletal/cytology , Orphan Nuclear Receptors/genetics , Orphan Nuclear Receptors/metabolism , Oxidation-Reduction , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Response Elements/physiology , Trans-Activators/genetics , Transcription Factors/genetics
17.
PLoS One ; 5(6): e10970, 2010 Jun 04.
Article in English | MEDLINE | ID: mdl-20532042

ABSTRACT

Adequate levels of physical activity are at the center of a healthy lifestyle. However, the molecular mechanisms that mediate the beneficial effects of exercise remain enigmatic. This gap in knowledge is caused by the lack of an amenable experimental model system. Therefore, we optimized electric pulse stimulation of muscle cells to closely recapitulate the plastic changes in gene expression observed in a trained skeletal muscle. The exact experimental conditions were established using the peroxisome proliferator-activated receptor gamma coactivator 1alpha (PGC-1alpha) as a marker for an endurance-trained muscle fiber. We subsequently compared the changes in the relative expression of metabolic and myofibrillar genes in the muscle cell system with those observed in mouse muscle in vivo following either an acute or repeated bouts of treadmill exercise. Importantly, in electrically stimulated C2C12 mouse muscle cells, the qualitative transcriptional adaptations were almost identical to those in trained muscle, but differ from the acute effects of exercise on muscle gene expression. In addition, significant alterations in the expression of myofibrillar proteins indicate that this stimulation could be used to modulate the fiber-type of muscle cells in culture. Our data thus describe an experimental cell culture model for the study of at least some of the transcriptional aspects of skeletal muscle adaptation to physical activity. This system will be useful for the study of the molecular mechanisms that regulate exercise adaptation in muscle.


Subject(s)
Electric Stimulation , Gene Expression Regulation , Muscle, Skeletal/physiology , Physical Conditioning, Animal , Animals , Cell Line , Mice , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Trans-Activators/metabolism , Transcription Factors , Transcription, Genetic
19.
Diabetes ; 58(10): 2228-37, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19602538

ABSTRACT

OBJECTIVE: Catch-up growth, a risk factor for later type 2 diabetes, is characterized by hyperinsulinemia, accelerated body-fat recovery (catch-up fat), and enhanced glucose utilization in adipose tissue. Our objective was to characterize the determinants of enhanced glucose utilization in adipose tissue during catch-up fat. RESEARCH DESIGN AND METHODS: White adipose tissue morphometry, lipogenic capacity, fatty acid composition, insulin signaling, in vivo glucose homeostasis, and insulinemic response to glucose were assessed in a rat model of semistarvation-refeeding. This model is characterized by glucose redistribution from skeletal muscle to adipose tissue during catch-up fat that results solely from suppressed thermogenesis (i.e., without hyperphagia). RESULTS: Adipose tissue recovery during the dynamic phase of catch-up fat is accompanied by increased adipocyte number with smaller diameter, increased expression of genes for adipogenesis and de novo lipogenesis, increased fatty acid synthase activity, increased proportion of saturated fatty acids in triglyceride (storage) fraction but not in phospholipid (membrane) fraction, and no impairment in insulin signaling. Furthermore, it is shown that hyperinsulinemia and enhanced adipose tissue de novo lipogenesis occur concomitantly and are very early events in catch-up fat. CONCLUSIONS: These findings suggest that increased adipose tissue insulin stimulation and consequential increase in intracellular glucose flux play an important role in initiating catch-up fat. Once activated, the machinery for lipogenesis and adipogenesis contribute to sustain an increased insulin-stimulated glucose flux toward fat storage. Such adipose tissue plasticity could play an active role in the thrifty metabolism that underlies glucose redistribution from skeletal muscle to adipose tissue.


Subject(s)
Adipose Tissue/growth & development , Adipose Tissue/metabolism , Dietary Fats/metabolism , Glucose/metabolism , Growth Disorders/metabolism , Lipids/physiology , Muscle, Skeletal/growth & development , Adipocytes/metabolism , Adipocytes/pathology , Animals , Body Composition , Diabetes Mellitus, Type 2/epidemiology , Energy Metabolism , Glucose Tolerance Test , Growth Disorders/complications , Humans , Hyperinsulinism/metabolism , Male , Muscle, Skeletal/metabolism , Rats , Rats, Sprague-Dawley , Thermogenesis
20.
FASEB J ; 20(10): 1751-3, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16809433

ABSTRACT

An enhanced metabolic efficiency for accelerating the recovery of fat mass (or catch-up fat) is a characteristic feature of body weight regulation after weight loss or growth retardation and is the outcome of an "adipose-specific" suppression of thermogenesis, i.e., a feedback control system in which signals from the depleted adipose tissue fat stores exert a suppressive effect on thermogenesis. Using a previously described rat model of semistarvation-refeeding in which catch-up fat results from suppressed thermogenesis per se, we report here that the gene expression of stearoyl-coenzyme A desaturase 1 (SCD1) is elevated in skeletal muscle after 2 wk of semistarvation and remains elevated in parallel to the phase of suppressed thermogenesis favoring catch-up fat during refeeding. These elevations in the SCD1 transcript are skeletal muscle specific and are associated with elevations in microsomal Delta9 desaturase enzyme activity, in the Delta9 desaturation index, and in the relative content of SCD1-derived monounsaturates in several lipid fractions extracted from skeletal muscle. An elevated skeletal muscle SCD1, by desaturating the products of de novo lipogenesis and diverting them away from mitochondrial oxidation, would inhibit substrate cycling between de novo lipogenesis and lipid oxidation, thereby leading to a state of suppressed thermogenesis that regulates the body's fat stores.


Subject(s)
Muscle, Skeletal/enzymology , Stearoyl-CoA Desaturase/physiology , Thermogenesis , Animals , Fatty Acid Desaturases/genetics , Feedback, Physiological , Gene Expression Regulation/physiology , Lipids/analysis , Lipogenesis , Muscle, Skeletal/physiology , Rats , Starvation , Stearoyl-CoA Desaturase/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...