Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
1.
Biochimie ; 179: 65-68, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32946989

ABSTRACT

Our understanding of cancer-specific metabolic changes is currently unclear. In recent years, the fruit fly Drosophila melanogaster with its powerful genetic tools has become an attractive model for studying both tumor autonomous and the systemic processes resulting from the tumor growth. Here we investigated the effect of tumorigenesis on the modulation of lipid droplets (LDs) in the larval fat bodies (mammalian equivalent of adipose tissue). We have overexpressed Notch signaling alone or in combination with the developmental regulator Myocyte enhancer factor 2 (Mef2) using wing-specific and eye-specific drivers, quantified the size of LDs in the fat body of the different tumor bearing larvae, and estimated the expression of genes associated with lipolysis and lipogenesis. We have found that hyperplastic and neoplastic tumor induced by overexpression of Notch and co-expression of Notch and Mef2 respectively triggers impaired lipid metabolism marked by increased size of fat body LDs. The impaired lipid metabolism in tumor carrying larvae is linked to the altered expression of genes that participate in lipolysis and lipogenesis. These findings reveal modulation of LDs as one of the host's specific response upon tumor initiation. This information could potentially uncover mechanisms for designing innovative approaches to modulate cancer growth.


Subject(s)
Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Epithelium/chemistry , Epithelium/metabolism , Fat Body/metabolism , Imaginal Discs/metabolism , Lipid Droplets/metabolism , Animals , Drosophila Proteins/biosynthesis , Eye/growth & development , Eye/pathology , Fat Body/pathology , Gene Expression Regulation, Neoplastic , Hyperplasia/genetics , Hyperplasia/metabolism , Larva/metabolism , Lipogenesis/genetics , Lipolysis/genetics , Myogenic Regulatory Factors/biosynthesis , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Receptors, Notch/biosynthesis , Wings, Animal/growth & development , Wings, Animal/pathology
2.
J Appl Physiol (1985) ; 127(5): 1419-1426, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31513447

ABSTRACT

Skeletal muscle satellite cells (SC) play an important role in muscle repair following injury. The regulation of SC activity is governed by myogenic regulatory factors (MRF), including MyoD, Myf5, myogenin, and MRF4. The mRNA expression of these MRF in humans following muscle damage has been predominately measured in whole muscle homogenates. Whether the temporal expression of MRF in a whole muscle homogenate reflects SC-specific expression of MRF remains largely unknown. Sixteen young men (23.1 ± 1.0 yr) performed 300 unilateral eccentric contractions (180°/s) of the knee extensors. Percutaneous muscle biopsies from the vastus lateralis were taken before (Pre) and 48 h postexercise. Fluorescence-activated cell sorting analysis was utilized to purify NCAM+ muscle SC from the whole muscle homogenate. Forty-eight hours post-eccentric exercise, MyoD, Myf5, and myogenin mRNA expression were increased in the whole muscle homogenate (~1.4-, ~4.0-, ~1.7-fold, respectively, P < 0.05) and in isolated SC (~19.3-, ~17.5-, ~58.9-fold, respectively, P < 0.05). MRF4 mRNA expression was not increased 48 h postexercise in the whole muscle homogenate (P > 0.05) or in isolated SC (P > 0.05). In conclusion, our results suggest that the directional changes in mRNA expression of the MRF in a whole muscle homogenate in response to acute eccentric exercise reflects that observed in isolated muscle SC.NEW & NOTEWORTHY The myogenic program is controlled via transcription factors referred to as myogenic regulatory factors (MRF). Previous studies have derived MRF expression from whole muscle homogenates, but little work has examined whether the mRNA expression of these transcripts reflects the pattern of expression in the actual population of satellite cells (SC). We report that MRF expression from an enriched SC population reflects the directional pattern of expression from skeletal muscle biopsy samples following eccentric contractions.


Subject(s)
Exercise/physiology , Muscle Contraction/physiology , Muscle, Skeletal/metabolism , Myogenic Regulatory Factors/biosynthesis , Satellite Cells, Skeletal Muscle/metabolism , Gene Expression , Humans , Male , Myogenic Regulatory Factors/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Young Adult
3.
PLoS One ; 12(6): e0179194, 2017.
Article in English | MEDLINE | ID: mdl-28617826

ABSTRACT

CF2 and Mef2 influence a variety of developmental muscle processes at distinct stages of development. Nevertheless, the exact nature of the CF2-Mef2 relationship and its effects on muscle building remain yet to be resolved. Here, we explored the regulatory role of CF2 in the Drosophila embryo muscle formation. To address this question and not having proper null CF2 mutants we exploited loss or gain of function strategies to study the contribution of CF2 to Mef2 transcription regulation and to muscle formation. Our data point to CF2 as a factor involved in the regulation of muscle final size and/or the number of nuclei present in each muscle. This function is independent of its role as a Mef2 collaborative factor in the transcriptional regulation of muscle-structural genes. Although Mef2 expression patterns do not change, reductions or increases in parallel in CF2 and Mef2 transcript abundance were observed in interfered and overexpressed CF2 embryos. Since CF2 expression variations yield altered Mef2 expression levels but with correct spatio-temporal Mef2 expression patterns, it can be concluded that only the mechanism controlling expression levels is de-regulated. Here, it is proposed that CF2 regulates Mef2 expression through a Feedforward Loop circuit.


Subject(s)
Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Drosophila Proteins/biosynthesis , Drosophila Proteins/metabolism , Embryo, Nonmammalian/embryology , Muscle Development/physiology , Muscles/embryology , Myogenic Regulatory Factors/biosynthesis , RNA, Messenger/biosynthesis , Transcription Factors/metabolism , Animals , Body Patterning/physiology , Cell Nucleus/genetics , DNA-Binding Proteins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster , Gene Expression Regulation, Developmental/physiology , Myogenic Regulatory Factors/genetics , RNA, Messenger/genetics , Transcription Factors/genetics
4.
PLoS One ; 10(12): e0142699, 2015.
Article in English | MEDLINE | ID: mdl-26691660

ABSTRACT

Despite surgical innovation, the sensory and motor outcome after a peripheral nerve injury remains incomplete. One contributing factor to the poor outcome is prolonged denervation of the target organ, leading to apoptosis of both mature myofibres and satellite cells with subsequent replacement of the muscle tissue with fibrotic scar and adipose tissue. In this study, we investigated the expression of myogenic transcription factors, muscle specific microRNAs and muscle-specific E3 ubiquitin ligases at several time points following denervation in two different muscles, the gastrocnemius (containing predominantly fast type fibres) and soleus (slow type) muscles, since these molecules may influence the degree of atrophy following denervation. Both muscles exhibited significant atrophy (compared with the contra-lateral sides) at 7 days following either a nerve transection or crush injury. In the crush model, the soleus muscle showed significantly increased muscle weights at days 14 and 28 which was not the case for the gastrocnemius muscle which continued to atrophy. There was a significantly more pronounced up-regulation of MyoD expression in the denervated soleus muscle compared with the gastrocnemius muscle. Conversely, myogenin was more markedly elevated in the gastrocnemius versus soleus muscles. The muscles also showed significantly contrasting transcriptional regulation of the microRNAs miR-1 and miR-206. MuRF1 and Atrogin-1 showed the highest levels of expression in the denervated gastrocnemius muscle. This study provides further insights regarding the intracellular regulatory molecules that generate and maintain distinct patterns of gene expression in different fibre types following peripheral nerve injury.


Subject(s)
Gene Expression Regulation , MicroRNAs/biosynthesis , Muscle Proteins/biosynthesis , Muscle, Skeletal/metabolism , Myogenic Regulatory Factors/biosynthesis , Peripheral Nerve Injuries/metabolism , SKP Cullin F-Box Protein Ligases/biosynthesis , Ubiquitin-Protein Ligases/biosynthesis , Animals , Female , Muscle, Skeletal/pathology , Peripheral Nerve Injuries/pathology , Rats , Rats, Sprague-Dawley , Tripartite Motif Proteins
5.
Exp Cell Res ; 336(2): 287-97, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-26101156

ABSTRACT

Single-nucleotide polymorphisms associated with type 2 diabetes (T2D) have been identified in Jazf1, which is also involved in the oncogenesis of endometrial stromal tumors. To understand how Jazf1 variants confer a risk of tumorigenesis and T2D, we explored the functional roles of JAZF1 and searched for JAZF1 target genes in myogenic C2C12 cells. Consistent with an increase of Jazf1 transcripts during myoblast proliferation and their decrease during myogenic differentiation in regenerating skeletal muscle, JAZF1 overexpression promoted cell proliferation, whereas it retarded myogenic differentiation. Examination of myogenic genes revealed that JAZF1 overexpression transcriptionally repressed MEF2C and MRF4 and their downstream genes. AMP deaminase1 (AMPD1) was identified as a candidate for JAZF1 target by gene array analysis. However, promoter assays of Ampd1 demonstrated that mutation of the putative binding site for the TR4/JAZF1 complex did not alleviate the repressive effects of JAZF1 on promoter activity. Instead, JAZF1-mediated repression of Ampd1 occurred through the MEF2-binding site and E-box within the Ampd1 proximal regulatory elements. Consistently, MEF2C and MRF4 expression enhanced Ampd1 promoter activity. AMPD1 overexpression and JAZF1 downregulation impaired AMPK phosphorylation, while JAZF1 overexpression also reduced it. Collectively, these results suggest that aberrant JAZF1 expression contributes to the oncogenesis and T2D pathogenesis.


Subject(s)
AMP Deaminase/genetics , Carrier Proteins/genetics , Cell Transformation, Neoplastic/genetics , Diabetes Mellitus, Type 2/genetics , Muscle Development/genetics , Nuclear Proteins/genetics , AMP Deaminase/biosynthesis , Animals , Binding Sites/genetics , Carrier Proteins/biosynthesis , Cell Differentiation/genetics , Cell Line , Cell Proliferation/genetics , Co-Repressor Proteins , DNA-Binding Proteins , Diabetes Mellitus, Type 2/pathology , Gene Expression Regulation/genetics , MEF2 Transcription Factors/biosynthesis , MEF2 Transcription Factors/genetics , Mice , Muscle Fibers, Skeletal/cytology , Myogenic Regulatory Factors/biosynthesis , Myogenic Regulatory Factors/genetics , Nuclear Proteins/biosynthesis , Polymorphism, Single Nucleotide , Promoter Regions, Genetic/genetics , RNA Interference , RNA, Small Interfering , Transcription, Genetic/genetics
6.
Appl Biochem Biotechnol ; 172(3): 1714-24, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24254256

ABSTRACT

Myostatin (MSTN) acts as a negative regulator of skeletal muscle development. Naturally occurring inactivating mutations in the coding region and knockout as well as knockdown of MSTN result in an increase in the muscle mass. However, the effect of MSTN knockdown on the expression of myogenic regulatory factors (MRFs) has not been studied in farm animals including goats. In the present study, using different synthetic siRNAs (n = 3), we demonstrated as high as 69 (p < 0.01) and 89% downregulation of MSTN mRNA and protein in the primary caprine foetal myoblast cells. Further, we also examined the effect of MSTN knockdown on the transcripts of MRFs including MyoD, Myf5 and MYOG. The expression of Myf5 remained unaffected (p = 0.60); however, MSTN downregulation caused a significant (p < 0.05) decrease and increase of MYOG and MyoD expression, respectively. Assessment of OAS1 expression confirmed the absence of any siRNA-elicited interferon response. Our results demonstrate that the downregulation of MSTN expression was accompanied by differential expressions of MRFs without any adverse interferon response. This study also suggests the importance of siRNA-mediated knockdown of MSTN as a potential alternative to increase muscle mass and meat production.


Subject(s)
Goats/genetics , Myogenic Regulatory Factors/genetics , Myostatin/genetics , RNA, Small Interfering/genetics , Animals , Embryonic Development , Fibroblasts/metabolism , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Goats/embryology , Goats/growth & development , Myoblasts/metabolism , Myogenic Regulatory Factors/biosynthesis , RNA, Messenger/genetics
7.
Int J Cardiol ; 167(3): 698-703, 2013 Aug 10.
Article in English | MEDLINE | ID: mdl-22464481

ABSTRACT

BACKGROUND: Although skeletal muscle atrophy and changes in myosin heavy chain (MyHC) isoforms have often been observed during heart failure, their pathophysiological mechanisms are not completely defined. In this study we tested the hypothesis that skeletal muscle phenotype changes are related to myogenic regulatory factors and myostatin/follistatin expression in spontaneously hypertensive rats (SHR) with heart failure. METHODS: After developing tachypnea, SHR were subjected to transthoracic echocardiogram. Pathological evidence of heart failure was assessed during euthanasia. Age-matched Wistar-Kyoto (WKY) rats were used as controls. Soleus muscle morphometry was analyzed in histological sections, and MyHC isoforms evaluated by electrophoresis. Protein levels were assessed by Western blotting. STATISTICAL ANALYSIS: Student'st test and Pearson correlation. RESULTS: All SHR presented right ventricular hypertrophy and seven had pleuropericardial effusion. Echocardiographic evaluation showed dilation in the left chambers and left ventricular hypertrophy with systolic and diastolic dysfunction in SHR. Soleus weight and fiber cross sectional areas were lower (WKY 3615 ± 412; SHR 2035 ± 224 µm(2); P<0.001), and collagen fractional volume was higher in SHR. The relative amount of type I MyHC isoform was increased in SHR. Myogenin, myostatin, and follistatin expression was lower and MRF4 levels higher in SHR. Myogenin and follistatin expression positively correlated with fiber cross sectional areas and MRF4 levels positively correlated with I MyHC isoform. CONCLUSION: Reduced myogenin and follistatin expression seems to participate in muscle atrophy while increased MRF4 protein levels can modulate myosin heavy chain isoform shift in skeletal muscle of spontaneously hypertensive rats with heart failure.


Subject(s)
Heart Failure/pathology , Muscle, Skeletal/pathology , Muscular Diseases/metabolism , Myogenic Regulatory Factors/antagonists & inhibitors , Myogenic Regulatory Factors/biosynthesis , Animals , Follistatin-Related Proteins/antagonists & inhibitors , Follistatin-Related Proteins/biosynthesis , Heart Failure/genetics , Heart Failure/metabolism , Male , Muscle, Skeletal/metabolism , Muscular Atrophy/genetics , Muscular Atrophy/metabolism , Muscular Atrophy/pathology , Muscular Diseases/genetics , Muscular Diseases/pathology , Myosin Heavy Chains/biosynthesis , Myosin Heavy Chains/genetics , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Species Specificity
8.
Genet Mol Res ; 12(4): 6733-42, 2013 Dec 13.
Article in English | MEDLINE | ID: mdl-24391014

ABSTRACT

To confirm the entire developmental process and transition point of embryonic Pekin duck pectoral muscle, and to investigate the association between pectoral muscle development and their regulating genes, anatomical and morphological analyses of embryonic Pekin duck skeletal muscles were performed, and the expression patterns of its regulating genes were investigated. The anatomical analysis revealed that body weight increased with age, while increases in pectoral muscle weight nearly ceased after the embryo was 20 days of hatching (E20). The developmental morphological characteristics of Pekin duck pectoral muscle at the embryonic stage showed that E20 was the transition point (from proliferation to fusion) of Pekin duck pectoral muscle. The expression patterns of MRF4, MyoG, and MSTN indicated that E19 or E20 was the fastest point of pectoral muscle development and the crucial transition for Pekin duck pectoral muscle development during the embryonic stage. Together, these findings imply that E20 is the crucial transition point (from proliferation to fusion) of Pekin duck pectoral muscle and that there is no muscle fiber hypertrophy after E20. Results of this study provide further understanding of the developmental process and transition point of Pekin duck pectoral muscle during the embryo stage.


Subject(s)
Ducks/embryology , Gene Expression Profiling/veterinary , Pectoralis Muscles/embryology , Animals , Body Weight , Embryo, Nonmammalian/metabolism , Gene Expression Regulation, Developmental/genetics , Myogenic Regulatory Factors/biosynthesis , Myogenic Regulatory Factors/genetics , Myogenin/biosynthesis , Myogenin/genetics , Myostatin/biosynthesis , Myostatin/genetics , Pectoralis Muscles/anatomy & histology , Pectoralis Muscles/growth & development , RNA, Messenger/biosynthesis
9.
Biol Trace Elem Res ; 150(1-3): 166-72, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23054870

ABSTRACT

The biological function of selenium (Se) is mainly elicited through Se-containing proteins. Selenoprotein W (SelW), one member of the selenoprotein family, is essential for the normal function of the skeletal muscle system. To investigate the possible relationship of Se in the process of differentiation in chicken myoblasts and the expression of SelW, the cultured chicken embryonic myoblasts were incubated with sodium selenite at different concentrations for 72 h, and then the mRNA levels of SelW and myogenic regulatory factors (MRFs) in myoblasts were determined at 12, 24, 48, and 72 h, respectively. Furthermore, the correlation between SelW mRNA expression and MRF mRNA expression was assessed. The results showed that the sodium selenite medium enhanced the mRNA expression of SelW, Myf-5, MRF4, and myogenin in chicken myoblasts. The mRNA expression levels of MRFs were significantly correlated with those of SelW at 24, 48, and 72 h. These data demonstrate that Se is involved in the differentiation of chicken embryonic myoblasts, and SelW showed correlation with MRFs.


Subject(s)
Myoblasts/metabolism , Myogenic Regulatory Factor 5/biosynthesis , Myogenic Regulatory Factors/biosynthesis , Myogenin/biosynthesis , Selenium/metabolism , Selenoprotein W/biosynthesis , Up-Regulation , Animals , Animals, Inbred Strains , Avian Proteins/biosynthesis , Avian Proteins/genetics , Avian Proteins/metabolism , Cell Differentiation , Cell Proliferation , Cells, Cultured , Chick Embryo , Muscle Development , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Myoblasts/cytology , Myogenic Regulatory Factor 5/genetics , Myogenic Regulatory Factor 5/metabolism , Myogenic Regulatory Factors/genetics , Myogenic Regulatory Factors/metabolism , Myogenin/genetics , Myogenin/metabolism , Osmolar Concentration , RNA, Messenger/metabolism , Selenoprotein W/genetics , Selenoprotein W/metabolism , Sodium Selenite/metabolism , Time Factors
10.
J Mol Cell Cardiol ; 53(4): 578-87, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22850285

ABSTRACT

The Myocyte Enhancer Factor-2 (MEF2) family of transcription factors regulates gene expression during cardiomyocyte differentiation and adaptation of the myocardium to stress. MEF2 activity is enhanced by increasing its transcription and by MAPK-dependent phosphorylation, and is reduced by binding to class-II Histone Deacetylases and by miR-1-mediated degradation of its transcript. Here we show that MEF2 protein abundance is regulated at the translational level, determining myocyte size, during hypertrophy. In order to reduce MEF2 protein expression, its silencing through RNA interference required serum deprivation and, even in this condition, MEF2 protein abundance recovered to basal levels in presence of phenylephrine. Hypertrophic agonist stimulation of neonatal ventricular cardiomyocytes increased Mef2 expression by enhancing its translation, without changing its transcription or blocking degradation of the protein. MEF2 abundance was increased by Calcineurin overexpression in vivo and was reduced by Calcineurin inhibition in vitro, without affecting Mef2 mRNA levels. Calcineurin activity influenced expression of Polypyrimidine Tract Protein (PTB), contributing to MEF2 translation. Thus, our results show a previously unrecognized but relevant level of MEF2 activity regulation through the control of its translation that involves Calcineurin and PTB.


Subject(s)
Calcineurin/metabolism , MADS Domain Proteins/biosynthesis , MADS Domain Proteins/genetics , Myocytes, Cardiac/metabolism , Myogenic Regulatory Factors/biosynthesis , Myogenic Regulatory Factors/genetics , Polypyrimidine Tract-Binding Protein/biosynthesis , Animals , Cells, Cultured , HEK293 Cells , Humans , MEF2 Transcription Factors , Male , Mice , Mice, Transgenic , Pyrimidines/pharmacology , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering , Rats , Rats, Sprague-Dawley , Signal Transduction
11.
Exp Biol Med (Maywood) ; 237(8): 912-8, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22875341

ABSTRACT

The prenatal and early postnatal period is a key developmental window for nutrition status, and high-fat exposure in this period has been shown to be associated with type 2 diabetes, obesity and other features of metabolic disorders later in life. The present study was designed to investigate the underlying molecular mechanisms and role of relative genes involved in this process. We investigated the impact of prenatal and early postnatal exposure to a high-saturated-fat diet on the regulation of the Wnt signaling pathway and myogenic genes in skeletal muscle of rat offspring as well as the serum and muscle physiological outcomes. Timed-pregnant Sprague-Dawley rats were fed either a control (C, 16% kcal fat) or high-saturated-fat diet (HF, 45% kcal fat) throughout gestation and lactation. After weaning, female offspring were fed a control diet to generate two offspring groups: control diet-fed offspring of control diet-fed dams (C/C) and control diet-fed offspring of HF diet-fed dams (HF/C). The serum glucose of the HF/C offspring (5.58 ± 0.26 mmol/L) was significantly higher than that of C/C offspring (4.97 ± 0.28 mmol/L), and the Homeostasis Model Assessment-Insulin Resistance of HF/C offspring (2.00 ± 0.11) was also significantly higher when compared with C/C (1.84 ± 0.09). Furthermore, HF/C offspring presented excessive intramuscular fat accumulation (1.8-fold, P < 0.05) and decreased muscle glycogen (1.3-fold, P < 0.05), as well as impairment of muscle development at the age of 12 weeks. Meanwhile, we observed the repression of Wnt/ß-catenin signaling and myogenic genes in HF/C offspring. The present study indicates that prenatal and early postnatal exposure to a high-saturated-fat diet suppresses the development of skeletal muscle and myogenic genes via Wnt/ß-catenin signaling, and the inappropriate muscle development could potentially contribute to the predisposition of offspring to develop metabolic-syndrome-like phenotype in adulthood.


Subject(s)
Dietary Fats/administration & dosage , Fatty Acids/administration & dosage , Gene Expression Profiling , Gene Expression Regulation, Developmental , Signal Transduction , Animals , Blood Glucose/analysis , Diet/methods , Female , Male , Myogenic Regulatory Factors/biosynthesis , Pregnancy , Rats , Rats, Sprague-Dawley , Wnt Proteins/biosynthesis
12.
Am J Physiol Cell Physiol ; 302(11): C1621-31, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22460713

ABSTRACT

The chemokine-like receptor-1 (CMKLR1) is a G protein-coupled receptor that is activated by chemerin, a secreted plasma leukocyte attractant and adipokine. Previous studies identified that CMKLR1 is expressed in skeletal muscle in a stage-specific fashion during embryogenesis and in adult mice; however, its function in skeletal muscle remains unclear. Based on the established function of CMKLR1 in cell migration and differentiation, we investigated the hypothesis that CMKLR1 regulates the differentiation of myoblasts into myotubes. In C(2)C(12) mouse myoblasts, CMKLR1 expression increased threefold with differentiation into multinucleated myotubes. Decreasing CMKLR1 expression by adenoviral-delivered small-hairpin RNA (shRNA) impaired the differentiation of C(2)C(12) myoblasts into mature myotubes and reduced the mRNA expression of myogenic regulatory factors myogenin and MyoD while increasing Myf5 and Mrf4. At embryonic day 12.5 (E12.5), CMKLR1 knockout (CMKLR1(-/-)) mice appeared developmentally delayed and displayed significantly lower wet weights and a considerably diminished myotomal component of somites as revealed by immunolocalization of myosin heavy chain protein compared with wild-type (CMKLR1(+/+)) mouse embryos. These changes were associated with increased Myf5 and decreased MyoD protein expression in the somites of E12.5 CMKLR1(-/-) mouse embryos. Adult male CMKLR1(-/-) mice had significantly reduced bone-free lean mass and weighed less than the CMKLR1(+/+) mice. We conclude that CMKLR1 is essential for myogenic differentiation of C(2)C(12) cells in vitro, and the CMKLR1 null mice have a subtle skeletal muscle deficit beginning from embryonic life that persists during postnatal life.


Subject(s)
Muscle Cells/metabolism , Muscle Development , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Receptors, G-Protein-Coupled/metabolism , Absorptiometry, Photon , Animals , Cell Differentiation , Cells, Cultured , Male , Mice , Mice, Knockout , Muscle Cells/physiology , Muscle Fibers, Skeletal/physiology , Muscle Proteins/biosynthesis , Muscle Proteins/genetics , Muscle, Skeletal/embryology , Muscle, Skeletal/physiology , MyoD Protein/biosynthesis , MyoD Protein/genetics , Myogenic Regulatory Factor 5/biosynthesis , Myogenic Regulatory Factor 5/genetics , Myogenic Regulatory Factor 5/metabolism , Myogenic Regulatory Factors/biosynthesis , Myogenic Regulatory Factors/genetics , Myogenic Regulatory Factors/metabolism , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering , Receptors, Chemokine
13.
Development ; 139(6): 1153-63, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22318228

ABSTRACT

Chondrocyte hypertrophy is crucial for endochondral ossification, but the mechanism underlying this process is not fully understood. We report that salt-inducible kinase 3 (SIK3) deficiency causes severe inhibition of chondrocyte hypertrophy in mice. SIK3-deficient mice showed dwarfism as they aged, whereas body size was unaffected during embryogenesis. Anatomical and histological analyses revealed marked expansion of the growth plate and articular cartilage regions in the limbs, accumulation of chondrocytes in the sternum, ribs and spine, and impaired skull bone formation in SIK3-deficient mice. The primary phenotype in the skeletal tissue of SIK3-deficient mice was in the humerus at E14.5, where chondrocyte hypertrophy was markedly delayed. Chondrocyte hypertrophy was severely blocked until E18.5, and the proliferative chondrocytes occupied the inside of the humerus. Consistent with impaired chondrocyte hypertrophy in SIK3-deficient mice, native SIK3 expression was detected in the cytoplasm of prehypertrophic and hypertrophic chondrocytes in developing bones in embryos and in the growth plates in postnatal mice. HDAC4, a crucial repressor of chondrocyte hypertrophy, remained in the nuclei in SIK3-deficient chondrocytes, but was localized in the cytoplasm in wild-type hypertrophic chondrocytes. Molecular and cellular analyses demonstrated that SIK3 was required for anchoring HDAC4 in the cytoplasm, thereby releasing MEF2C, a crucial facilitator of chondrocyte hypertrophy, from suppression by HDAC4 in nuclei. Chondrocyte-specific overexpression of SIK3 induced closure of growth plates in adulthood, and the SIK3-deficient cartilage phenotype was rescued by transgenic SIK3 expression in the humerus. These results demonstrate an essential role for SIK3 in facilitating chondrocyte hypertrophy during skeletogenesis and growth plate maintenance.


Subject(s)
Bone Development , Chondrocytes/cytology , Chondrocytes/physiology , Osteogenesis , Protein Serine-Threonine Kinases/metabolism , Animals , Bone Development/genetics , Bone and Bones/abnormalities , Cell Differentiation/genetics , Cell Nucleus/metabolism , Cells, Cultured , Chondrogenesis , Collagen Type XI/genetics , Dwarfism/genetics , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Growth Plate/physiology , Histone Deacetylases/metabolism , Hypertrophy , MEF2 Transcription Factors , Mice , Mice, Knockout , Mice, Transgenic , Myogenic Regulatory Factors/biosynthesis , Osteogenesis/genetics , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/genetics
14.
J Biol Chem ; 286(41): 35339-35346, 2011 Oct 14.
Article in English | MEDLINE | ID: mdl-21868385

ABSTRACT

microRNAs (miRNAs) are 21-23-nucleotide non-coding RNAs. It has become more and more evident that this class of small RNAs plays critical roles in the regulation of gene expression at the post-transcriptional level. MEF2A is a member of the MEF2 (myogenic enhancer factor 2) family of transcription factors. Prior report showed that the 3'-untranslated region (3'-UTR) of the Mef2A gene mediated its repression; however, the molecular mechanism underlying this intriguing observation was unknown. Here, we report that MEF2A is repressed by miRNAs. We identify miR-155 as one of the primary miRNAs that significantly represses the expression of MEF2A. We show that knockdown of the Mef2A gene by siRNA impairs myoblast differentiation. Similarly, overexpression of miR-155 leads to the repression of endogenous MEF2A expression and the inhibition of myoblast differentiation. Most importantly, reintroduction of MEF2A in miR-155 overexpressed myoblasts was able to partially rescue the miR-155-induced myoblast differentiation defect. Our data therefore establish miR-155 as an important regulator of MEF2A expression and uncover its function in muscle gene expression and myogenic differentiation.


Subject(s)
Cell Differentiation/physiology , Gene Expression Regulation/physiology , MicroRNAs/metabolism , Muscle, Skeletal/metabolism , Myoblasts, Skeletal/metabolism , Myogenic Regulatory Factors/biosynthesis , 3' Untranslated Regions/physiology , Animals , COS Cells , Chlorocebus aethiops , Humans , MEF2 Transcription Factors , Mice , MicroRNAs/genetics , Muscle, Skeletal/cytology , Myoblasts, Skeletal/cytology , Myogenic Regulatory Factors/genetics
15.
Neurodegener Dis ; 8(5): 386-96, 2011.
Article in English | MEDLINE | ID: mdl-21346327

ABSTRACT

BACKGROUND: In the superoxide dismutase 1 (SOD1)-G93A mouse model of amyotrophic lateral sclerosis (ALS), skeletal muscle is a key target of mutant SOD1 toxicity. However, the expression of factors that control the regenerative potential of the muscle is unknown in this model. OBJECTIVE: To characterize the expression of satellite cell marker Pax7 and myogenic regulatory factors (MRF) in skeletal muscle of SOD1-G93A mice at different stages of the disease. METHODS: The expressions of Pax7, Myod1, Myf5 and myogenin (Myog) were determined by quantitative real-time PCR and by Western blotting from the grouped gastrocnemius, quadriceps and soleus muscles of SOD1-G93A mice at presymptomatic, symptomatic and terminal stages of the disease, and from surgically denervated wild-type gastrocnemius muscles. RESULTS: Pax7 mRNA and MYF5 protein were upregulated in presymptomatic mice, coinciding with increased muscle damage marker Rrad and chemokine Ccl5. All MRF transcripts and most proteins (excluding MYOG) were increased, starting from 3 months of age, simultaneously with increased expression of denervation marker Chrna1. However, in the terminal stage, no protein increase was evident for Pax7 or any of the MRF despite the increased mRNA levels. The transcripts for chemokine Ccl2 and chemokine receptor Cxcr4 were increased starting from the onset of symptoms. CONCLUSIONS: The characterization of Pax7 and MRF in SOD1-G93A mice reveals a progressive induction of the myogenic program at the RNA level, but a blunted protein level response at late stages of the disease. Altered posttranscriptional and posttranslational mechanisms likely to operate, as well as the potential role of chemokine signaling in mutant SOD1 muscle, are discussed.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Disease Models, Animal , Myogenic Regulatory Factors/biosynthesis , Amyotrophic Lateral Sclerosis/genetics , Animals , Gene Expression Regulation, Enzymologic , Humans , Male , Mice , Mice, Transgenic , Myogenic Regulatory Factors/genetics , Superoxide Dismutase/biosynthesis , Superoxide Dismutase/genetics
16.
Am J Physiol Endocrinol Metab ; 300(5): E790-9, 2011 May.
Article in English | MEDLINE | ID: mdl-21304067

ABSTRACT

Arthritis is a chronic inflammatory illness that induces cachexia, which has a direct impact on morbidity and mortality. Fenofibrate, a selective PPARα activator prescribed to treat human dyslipidemia, has been reported to decrease inflammation in rheumatoid arthritis patients. The aim of this study was to elucidate whether fenofibrate is able to ameliorate skeletal muscle wasting in adjuvant-induced arthritis, an experimental model of rheumatoid arthritis. On day 4 after adjuvant injection, control and arthritic rats were treated with 300 mg/kg fenofibrate until day 15, when all rats were euthanized. Fenofibrate decreased external signs of arthritis and liver TNFα and blocked arthritis-induced decreased in PPARα expression in the gastrocnemius muscle. Arthritis decreased gastrocnemius weight, which results from a decrease in cross-section area and myofiber size, whereas fenofibrate administration to arthritic rats attenuated the decrease in both gastrocnemius weight and fast myofiber size. Fenofibrate treatment prevented arthritis-induced increase in atrogin-1 and MuRF1 expression in the gastrocnemius. Neither arthritis nor fenofibrate administration modify Akt-FoxO3 signaling. Myostatin expression was not modified by arthritis, but fenofibrate decreased myostatin expression in the gastrocnemius of arthritic rats. Arthritis increased muscle expression of MyoD, PCNA, and myogenin in the rats treated with vehicle but not in those treated with fenofibrate. The results indicate that, in experimental arthritis, fenofibrate decreases skeletal muscle atrophy through inhibition of the ubiquitin-proteasome system and myostatin.


Subject(s)
Arthritis, Experimental/pathology , Fenofibrate/pharmacology , Hypolipidemic Agents/pharmacology , Muscle Proteins/biosynthesis , Muscle, Skeletal/pathology , Myostatin/biosynthesis , Myostatin/genetics , PPAR gamma/agonists , SKP Cullin F-Box Protein Ligases/biosynthesis , Ubiquitin-Protein Ligases/biosynthesis , Animals , Arthritis, Experimental/drug therapy , Atrophy , Body Weight/drug effects , Eating/drug effects , Gene Expression/drug effects , Lipids/blood , Male , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/ultrastructure , Muscle Proteins/genetics , Myogenic Regulatory Factors/biosynthesis , Myogenic Regulatory Factors/genetics , Organ Size/drug effects , Rats , Rats, Wistar , SKP Cullin F-Box Protein Ligases/genetics , Tripartite Motif Proteins , Ubiquitin-Protein Ligases/genetics
17.
J Psychopharmacol ; 25(2): 222-9, 2011 Feb.
Article in English | MEDLINE | ID: mdl-19939859

ABSTRACT

Injection of the histone deacetylases inhibitor trichostatin A to rats has been shown to decrease the reinforcing properties of cocaine. In the present study, we investigated alterations in gene expression patterns in the anterior cingulate cortex, caudate-putamen and nucleus accumbens of rats self-administering cocaine and treated with trichostatin A. As recent studies highlighted the importance of chromatin remodelling in the regulation of gene transcription in neurons, we studied the expression of Mecp2 and of several histone deacetylases. Cocaine self-administration was accompanied by an increased synthesis of Mecp2, HDAC2 and HDAC11 and by a decreased nuclear localization of HDAC5 and of the phospho-form of HDAC5, suggesting a nuclear export of this protein in response to the drug. The latter mechanism was further addressed by the demonstration of an enhanced expression of MEF2C transcription factor. Among the genes we examined, treatment with trichostatin A before each cocaine self-administration session was found to mostly affect Mecp2 and HDAC11 expression. A correlation was found between the modification of Mecp2 and MEF2C gene expression and the reinforcing property of cocaine. The two factors known to regulate gene transcription are likely to play a role in the neurobiological mechanism underlying a decrease in the reinforcing properties of cocaine.


Subject(s)
Chromatin Assembly and Disassembly/drug effects , Cocaine/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Animals , Cell Nucleus/metabolism , Cocaine/administration & dosage , Conditioning, Operant/drug effects , Corpus Striatum/drug effects , Corpus Striatum/enzymology , Gene Expression Regulation/drug effects , Gyrus Cinguli/drug effects , Gyrus Cinguli/enzymology , Histone Deacetylases/biosynthesis , MEF2 Transcription Factors , Male , Methyl-CpG-Binding Protein 2/biosynthesis , Myogenic Regulatory Factors/biosynthesis , Nucleus Accumbens/drug effects , Nucleus Accumbens/enzymology , Rats , Rats, Wistar , Self Administration , Transcription Factors/biosynthesis
18.
J Mol Cell Cardiol ; 50(3): 561-9, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21163264

ABSTRACT

Approximately 5 million people are affected with aortic valve disease (AoVD) in the United States. The most common treatment is aortic valve (AoV) replacement surgery, however, replacement valves are susceptible to failure, necessitating additional surgeries. The molecular mechanisms underlying disease progression and late AoV calcification are not well understood. Recent studies suggest that genes involved in bone and cartilage development play an active role in osteogenic-like calcification in human calcific AoVD (CAVD). In an effort to define the molecular pathways involved in AoVD progression and calcification, expression of markers of valve mesenchymal progenitors, chondrogenic precursors, and osteogenic differentiation was compared in pediatric non-calcified and adult calcified AoV specimens. Valvular interstitial cell (VIC) activation, extracellular matrix (ECM) disorganization, and markers of valve mesenchymal and skeletal chondrogenic progenitor cells were observed in both pediatric and adult AoVD. However, activated BMP signaling, increased expression of cartilage and bone-type collagens, and increased expression of the osteogenic marker Runx2 are observed in adult diseased AoVs. They are not observed in the majority of pediatric diseased valves, representing a marked distinction in the molecular profile between pediatric and adult diseased AoVs. The combined evidence suggests that an actively regulated osteochondrogenic disease process underlies the pathological changes affecting AoVD progression, ultimately resulting in stenotic AoVD. Both pediatric and adult diseased AoVs express protein markers of valve mesenchymal and chondrogenic progenitor cells while adult diseased AoVs also express proteins involved in osteogenic calcification. These findings provide specific molecular indicators of AoVD progression, which may lead to identification of early disease markers and the development of potential therapeutics.


Subject(s)
Aortic Valve/metabolism , Aortic Valve/pathology , Bone and Bones/metabolism , Cartilage/metabolism , Heart Valve Diseases/metabolism , Heart Valve Diseases/pathology , Aged , Biomarkers/metabolism , Bone Morphogenetic Proteins/biosynthesis , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Bone and Bones/pathology , Calcinosis/genetics , Calcinosis/pathology , Cell Differentiation/physiology , Cell Growth Processes/physiology , Child , Child, Preschool , Chondrogenesis/genetics , Chondrogenesis/physiology , Collagen/genetics , Collagen/metabolism , Core Binding Factor Alpha 1 Subunit/biosynthesis , Core Binding Factor Alpha 1 Subunit/genetics , Extracellular Matrix/metabolism , Heart Valve Diseases/genetics , Humans , MADS Domain Proteins/biosynthesis , MADS Domain Proteins/genetics , MEF2 Transcription Factors , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Myogenic Regulatory Factors/biosynthesis , Myogenic Regulatory Factors/genetics , Osteogenesis/genetics , Osteogenesis/physiology , Transcription Factors/biosynthesis , Transcription Factors/genetics
19.
Article in English | MEDLINE | ID: mdl-20434580

ABSTRACT

The nutritional regulation of skeletal muscle growth is very little documented in fish. The aim of the study presented here was to determine how changes in dietary plant protein sources and amino acid profiles affect the muscle growth processes of fish. Juvenile rainbow trout (Oncorhynchys mykiss) were fed two diets containing fish meal and a mixture of plant protein sources either low (control diet) or rich in soybean meal (diet S). Both diets were supplemented with crystalline indispensable amino acids (IAA) to match the rainbow trout muscle IAA profile. Diet S was also supplemented with glutamic acid, an AA present in high quantities in trout muscle. Rainbow trout fed diets C and S were not significantly different in terms of overall somatic growth or daily nitrogen gain, although their parameters of dietary protein utilisation differed. Distribution of skeletal white muscle fibre diameter and expression of certain selected muscle genes were also affected by dietary changes. In the white muscle, diet S led to a significant decrease (x0.9) in the mean and median diameters of muscle fibres, to a significant decrease (x0.6) in the expression of MyoD and to a significant increase (x1.7) in the expression of fast-MHC, with no significant changes in myogenin expression. There was no change in the expression of the genes analysed in lateral red muscle (MyoD, MyoD2, myogenin and slow-MHC). These results demonstrated that changes occurred in skeletal white muscle cellularity and expression of MyoD and fast-MHC, although overall growth and protein accretion were not modified, when a diet rich in soybean meal and glutamic acid was ingested. Present findings also indicated that the white and red muscles of rainbow trout are differently affected by nutritional changes.


Subject(s)
Amino Acids/analysis , Dietary Proteins/analysis , Muscle, Skeletal/cytology , Muscle, Skeletal/metabolism , Myogenic Regulatory Factors/biosynthesis , Myosin Heavy Chains/biosynthesis , Oncorhynchus mykiss/metabolism , Plant Proteins/chemistry , Amino Acids/metabolism , Animal Feed , Animals , Diet/veterinary , Dietary Proteins/administration & dosage , Gene Expression Regulation, Developmental , Muscle, Skeletal/growth & development , Myogenic Regulatory Factors/genetics , Myosin Heavy Chains/genetics , Oncorhynchus mykiss/growth & development , Plant Proteins/administration & dosage , Plant Proteins/metabolism , Species Specificity
20.
J Biol Chem ; 285(2): 1529-43, 2010 Jan 08.
Article in English | MEDLINE | ID: mdl-19897480

ABSTRACT

Down syndrome (DS), or Trisomy 21, is the most common genetic cause of cognitive impairment and congenital heart defects in the human population. Bioinformatic annotation has established that human chromosome 21 (Hsa21) harbors five microRNA (miRNAs) genes: miR-99a, let-7c, miR-125b-2, miR-155, and miR-802. Our laboratory recently demonstrated that Hsa21-derived miRNAs are overexpressed in DS brain and heart specimens. The aim of this study was to identify important Hsa21-derived miRNA/mRNA target pairs that may play a role, in part, in mediating the DS phenotype. We demonstrate by luciferase/target mRNA 3'-untranslated region reporter assays, and gain- and loss-of-function experiments that miR-155 and -802 can regulate the expression of the predicted mRNA target, the methyl-CpG-binding protein (MeCP2). We also demonstrate that MeCP2 is underexpressed in DS brain specimens isolated from either humans or mice. We further demonstrate that, as a consequence of attenuated MeCP2 expression, transcriptionally activated and silenced MeCP2 target genes, CREB1/Creb1 and MEF2C/Mef2c, are also aberrantly expressed in these DS brain specimens. Finally, in vivo silencing of endogenous miR-155 or -802, by antagomir intra-ventricular injection, resulted in the normalization of MeCP2 and MeCP2 target gene expression. Taken together, these results suggest that improper repression of MeCP2, secondary to trisomic overexpression of Hsa21-derived miRNAs, may contribute, in part, to the abnormalities in the neurochemistry observed in the brains of DS individuals. Finally these results suggest that selective inactivation of Hsa21-derived miRNAs may provide a novel therapeutic tool in the treatment of DS.


Subject(s)
Brain/metabolism , Chromosomes, Human, Pair 21/metabolism , Down Syndrome/metabolism , Gene Expression Regulation , MicroRNAs/metabolism , Nerve Tissue Proteins/biosynthesis , 3' Untranslated Regions/genetics , Animals , Chromosomes, Human, Pair 21/genetics , Cyclic AMP Response Element-Binding Protein/biosynthesis , Cyclic AMP Response Element-Binding Protein/genetics , Down Syndrome/genetics , Female , Humans , MADS Domain Proteins/biosynthesis , MADS Domain Proteins/genetics , MEF2 Transcription Factors , Male , Methyl-CpG-Binding Protein 2/biosynthesis , Methyl-CpG-Binding Protein 2/genetics , Mice , Mice, Knockout , MicroRNAs/genetics , Myocardium/metabolism , Myogenic Regulatory Factors/biosynthesis , Myogenic Regulatory Factors/genetics , Nerve Tissue Proteins/genetics , Organ Specificity/genetics , Transcription, Genetic/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...