Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Mol Pharmacol ; 100(6): 588-596, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34561299

RESUMO

Tenofovir (TFV) is a key component of human immunodeficiency virus (HIV) pre-exposure prophylaxis (PrEP). TFV is a nucleotide analog reverse-transcriptase inhibitor prodrug that requires two separate phosphorylation reactions by intracellular kinases to form the active metabolite tenofovir-diphosphate (TFV-DP). Muscle-type creatine kinase (CKM) has previously been demonstrated to be the kinase most responsible for the phosphorylation of tenofovir-monophosphate (TFV-MP) to the active metabolite in colon tissue. Because of the importance of CKM in TFV activation, genetic variation in CKM may contribute to interindividual variability in TFV-DP levels. In the present study, we report 10 naturally occurring CKM mutations that reduced TFV-MP phosphorylation in vitro: T35I, R43Q, I92M, H97Y, R130H, R132C, F169L, Y173C, W211R, V280L, and N286I. Interestingly, of these 10, only 4-R130H, R132C, W211R, and N286I-reduced both canonical CKM activities: ADP phosphorylation and ATP dephosphorylation. Although positions 130, 132, and 286 are located in the active site, the other mutations that resulted in decreased TFV-MP phosphorylation occur elsewhere in the protein structure. Four of these eight mutations-T35I, R43Q, I92M, and W211R-were found to decrease the thermal stability of the protein. Additionally, the W211R mutation was found to impact protein structure both locally and at a distance. These data suggest a substrate-specific effect such that certain mutations are tolerated for canonical activities while being deleterious toward the pharmacological activity of TFV activation, which could influence PrEP outcomes. SIGNIFICANCE STATEMENT: Muscle-type creatine kinase (CKM) is important to the activation of tenofovir, a key component of HIV prophylaxis. This study demonstrates that naturally occurring CKM mutations impact enzyme function in a substrate-dependent manner such that some mutations that do not reduce canonical activities lead to reductions in the pharmacologically relevant activity. This finding at the intersection of drug metabolism and energy metabolism is important to the perspective on pharmacology of other drugs acted on by atypical drug-metabolizing enzymes.


Assuntos
Creatina Quinase Forma MM/química , Mutação , Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Fármacos Anti-HIV/química , Fármacos Anti-HIV/farmacologia , Sítios de Ligação , Creatina Quinase Forma MM/genética , Creatina Quinase Forma MM/metabolismo , Humanos , Simulação de Acoplamento Molecular , Fosforilação , Ligação Proteica , Tenofovir/química , Tenofovir/farmacologia
2.
Pharmacol Res ; 155: 104680, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32032665

RESUMO

Friedreich's ataxia (FA) is due to deficiency of the mitochondrial protein, frataxin, which results in multiple pathologies including a deadly, hypertrophic cardiomyopathy. Frataxin loss leads to deleterious accumulations of redox-active, mitochondrial iron, and suppressed mitochondrial bioenergetics. Hence, there is an urgent need to develop innovative pharmaceuticals. Herein, the activity of the novel compound, 6-methoxy-2-salicylaldehyde nicotinoyl hydrazone (SNH6), was assessed in vivo using the well-characterized muscle creatine kinase (MCK) conditional frataxin knockout (KO) mouse model of FA. The design of SNH6 incorporated a dual-mechanism mediating: (1) NAD+-supplementation to restore cardiac bioenergetics; and (2) iron chelation to remove toxic mitochondrial iron. In these studies, MCK wild-type (WT) and KO mice were treated for 4-weeks from the asymptomatic age of 4.5-weeks to 8.5-weeks of age, where the mouse displays an overt cardiomyopathy. SNH6-treatment significantly elevated NAD+ and markedly increased NAD+ consumption in WT and KO hearts. In SNH6-treated KO mice, nuclear Sirt1 activity was also significantly increased together with the NAD+-metabolic product, nicotinamide (NAM). Therefore, NAD+-supplementation by SNH6 aided mitochondrial function and cardiac bioenergetics. SNH6 also chelated iron in cultured cardiac cells and also removed iron-loading in vivo from the MCK KO heart. Despite its dual beneficial properties of supplementing NAD+ and chelating iron, SNH6 did not mitigate cardiomyopathy development in the MCK KO mouse. Collectively, SNH6 is an innovative therapeutic with marked pharmacological efficacy, which successfully enhanced cardiac NAD+ and nuclear Sirt1 activity and reduced cardiac iron-loading in MCK KO mice. No other pharmaceutical yet designed exhibits both these effective pharmacological properties.


Assuntos
Aldeídos/uso terapêutico , Cardiomiopatias/tratamento farmacológico , Ataxia de Friedreich/tratamento farmacológico , Hidrazonas/uso terapêutico , Quelantes de Ferro/uso terapêutico , NAD/metabolismo , Trifosfato de Adenosina/metabolismo , Aldeídos/farmacologia , Animais , Cardiomiopatias/metabolismo , Linhagem Celular , Creatina Quinase Forma MM/genética , Modelos Animais de Doenças , Ataxia de Friedreich/metabolismo , Hidrazonas/farmacologia , Ferro/metabolismo , Quelantes de Ferro/farmacologia , Proteínas de Ligação ao Ferro/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias Cardíacas/efeitos dos fármacos , Mitocôndrias Cardíacas/metabolismo , Ratos , Frataxina
3.
Sci Rep ; 9(1): 5429, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30931999

RESUMO

The creatine/phosphocreatine system is the principal energy buffer in mammals, but is scarcely documented in fish. We measured the gene expression of major enzymes of this system, glycine amidinotransferase (GATM), guanidinoacetate N-methyltransferase (GAMT) and muscle-type creatine kinase (CKM) in kidney, liver, and muscle tissues of fish and mammals. CKM was expressed strongly in the muscles of all examined species. In contrast, GATM and GAMT were strongly expressed in the muscle tissue of fish, but not of mammals. This indicates that creatine synthesis and usage are spatially separated in mammals, but not in fish, which is supported by RNA-Seq data of 25 species. Differences in amino acid metabolism along with methionine adenosyltransferase gene expression in muscle from fishes but not mammals further support a central metabolic role of muscle in fish, and hence different organization of the creatine/phosphocreatine biosynthesis system in higher and lower vertebrates.


Assuntos
Creatina/biossíntese , Evolução Molecular , Músculo Esquelético/metabolismo , Amidinotransferases/genética , Animais , Creatina Quinase Forma MM/genética , Peixes , Perfilação da Expressão Gênica , Músculo Esquelético/enzimologia , Análise de Sequência de RNA
4.
PLoS One ; 13(4): e0195764, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29641561

RESUMO

Tenofovir (TFV), a nucleotide reverse transcriptase inhibitor, requires two phosphorylation steps to form a competitive inhibitor of HIV reverse transcriptase. Adenylate kinase 2 (AK2) has been previously demonstrated to phosphorylate tenofovir to tenofovir-monophosphate, while creatine kinase, muscle (CKM), pyruvate kinase, muscle (PKM) and pyruvate kinase, liver and red blood cell (PKLR) each have been found to phosphorylate tenofovir-monophosphate to the pharmacologically active tenofovir-diphosphate. In the present study, genomic DNA isolated from dried blood spots collected from 505 participants from Bangkok, Thailand; Cape Town, South Africa; and New York City, USA were examined for variants in AK2, CKM, PKM, and PKLR using next-generation sequencing. The bioinformatics tools SIFT and PolyPhen predicted that 19 of the 505 individuals (3.7% frequency) carried variants in at least one kinase that would result in a decrease or loss of enzymatic activity. To functionally test these predictions, AK2 and AK2 variants were expressed in and purified from E. coli, followed by investigation of their activities towards tenofovir. Interestingly, we found that purified AK2 had the ability to phosphorylate tenofovir-monophosphate to tenofovir-diphosphate in addition to phosphorylating tenofovir to tenofovir-monophosphate. Further, four of the six AK2 variants predicted to result in a loss or decrease of enzyme function exhibited a ≥30% decrease in activity towards tenofovir in our in vitro assays. Of note, an AK2 K28R variant resulted in a 72% and 81% decrease in the formation of tenofovir-monophosphate and tenofovir-diphosphate, respectively. These data suggest that there are naturally occurring genetic variants that could potentially impact TFV activation.


Assuntos
Adenilato Quinase/genética , Creatina Quinase Forma MM/genética , Variação Genética , Infecções por HIV/tratamento farmacológico , HIV-1/efeitos dos fármacos , Piruvato Quinase/genética , Tenofovir/farmacologia , Fármacos Anti-HIV/farmacologia , Infecções por HIV/epidemiologia , Infecções por HIV/genética , Infecções por HIV/virologia , HIV-1/enzimologia , HIV-1/genética , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , África do Sul/epidemiologia , Tailândia/epidemiologia , Estados Unidos/epidemiologia
5.
PLoS One ; 12(3): e0172965, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28257486

RESUMO

PURPOSE: Exertional rhabdomyolysis can occur in individuals performing various types of exercise but it is unclear why some individuals develop this condition while others do not. Previous investigations have determined the role of several single nucleotide polymorphisms (SNPs) to explain inter-individual variability of serum creatine kinase (CK) concentrations after exertional muscle damage. However, there has been no research about the interrelationship among these SNPs. The purpose of this investigation was to analyze seven SNPs that are candidates for explaining individual variations of CK response after a marathon competition (ACE = 287bp Ins/Del, ACTN3 = p.R577X, CKMM = NcoI, IGF2 = C13790G, IL6 = 174G>C, MLCK = C37885A, TNFα = 308G>A). METHODS: Using Williams and Folland's model, we determined the total genotype score from the accumulated combination of these seven SNPs for marathoners with a low CK response (n = 36; serum CK <400 U·L-1) vs. marathoners with a high CK response (n = 31; serum CK ≥400 U·L-1). RESULTS: At the end of the race, low CK responders had lower serum CK (290±65 vs. 733±405 U·L-1; P<0.01) and myoglobin concentrations (443±328 vs. 1009±971 ng·mL-1, P<0.01) than high CK responders. Although the groups were similar in age, anthropometric characteristics, running experience and training habits, total genotype score was higher in low CK responders than in high CK responders (5.2±1.4 vs. 4.4±1.7 point, P = 0.02). CONCLUSION: Marathoners with a lower CK response after the race had a more favorable polygenic profile than runners with high serum CK concentrations. This might suggest a significant role of genetic polymorphisms in the levels of exertional muscle damage and rhabdomyolysis. Yet other SNPs, in addition to exercise training, might also play a role in the values of CK after damaging exercise.


Assuntos
Creatina Quinase Forma MM/genética , Esforço Físico , Polimorfismo de Nucleotídeo Único , Rabdomiólise/diagnóstico , Rabdomiólise/genética , Actinina/sangue , Actinina/genética , Adolescente , Adulto , Idoso , Creatina Quinase Forma MM/sangue , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Fator de Crescimento Insulin-Like II/genética , Fator de Crescimento Insulin-Like II/metabolismo , Interleucina-6/sangue , Interleucina-6/genética , Masculino , Pessoa de Meia-Idade , Mioglobina/sangue , Quinase de Cadeia Leve de Miosina/sangue , Quinase de Cadeia Leve de Miosina/genética , Peptidil Dipeptidase A/sangue , Peptidil Dipeptidase A/genética , Prognóstico , Rabdomiólise/sangue , Rabdomiólise/patologia , Corrida , Fator de Necrose Tumoral alfa/sangue , Fator de Necrose Tumoral alfa/genética
6.
J Biol Chem ; 291(49): 25306-25318, 2016 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-27738103

RESUMO

Lactate dehydrogenase (LDH) catalyzes the interconversion of pyruvate and lactate, which are critical fuel metabolites of skeletal muscle particularly during exercise. However, the physiological relevance of LDH remains poorly understood. Here we show that Ldhb expression is induced by exercise in human muscle and negatively correlated with changes in intramuscular pH levels, a marker of lactate production, during isometric exercise. We found that the expression of Ldhb is regulated by exercise-induced peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α). Ldhb gene promoter reporter studies demonstrated that PGC-1α activates Ldhb gene expression through multiple conserved estrogen-related receptor (ERR) and myocyte enhancer factor 2 (MEF2) binding sites. Transgenic mice overexpressing Ldhb in muscle (muscle creatine kinase (MCK)-Ldhb) exhibited increased exercise performance and enhanced oxygen consumption during exercise. MCK-Ldhb muscle was shown to have enhanced mitochondrial enzyme activity and increased mitochondrial gene expression, suggesting an adaptive oxidative muscle transformation. In addition, mitochondrial respiration capacity was increased and lactate production decreased in MCK-Ldhb skeletal myotubes in culture. Together, these results identified a previously unrecognized Ldhb-driven alteration in muscle mitochondrial function and suggested a mechanism for the adaptive metabolic response induced by exercise training.


Assuntos
Regulação Enzimológica da Expressão Gênica/fisiologia , L-Lactato Desidrogenase/biossíntese , Mitocôndrias Musculares/enzimologia , Músculo Esquelético/enzimologia , Condicionamento Físico Animal , Animais , Creatina Quinase Forma MM/genética , Creatina Quinase Forma MM/metabolismo , Humanos , Isoenzimas/biossíntese , Isoenzimas/genética , L-Lactato Desidrogenase/genética , Camundongos , Camundongos Transgênicos , Mitocôndrias Musculares/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/genética , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo
7.
Chem Biol ; 22(11): 1461-1469, 2015 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-26526100

RESUMO

Protein glutathionylation is an important post-translational modification that regulates many cellular processes, including energy metabolism, signal transduction, and protein homeostasis. Global profiling of glutathionylated proteins (denoted as glutathionylome) is crucial for understanding redox-regulated signal transduction. Here, we developed a novel method based on click reaction and proteomics to enrich and identify the glutathionylated peptides in Escherichia coli and Drosophila lysates, in which 937 and 1,930 potential glutathionylated peptides were identified, respectively. Bioinformatics analysis showed that the cysteine residue next to negatively charged amino acid residues has a higher frequency of glutathionylation. Importantly, we found that most proteins associated with metabolic pathways were glutathionylated and that the glutathionylation sites of metabolic enzymes were highly conserved among different species. Our results indicate that the glutathione analog is a useful tool to characterize protein glutathionylation, and glutathionylation of metabolic enzymes, which play important roles in regulating cellular metabolism, is conserved.


Assuntos
Proteínas de Drosophila/química , Drosophila/metabolismo , Proteínas de Escherichia coli/química , Escherichia coli/metabolismo , Glutationa/análogos & derivados , Sondas Moleculares/química , Sequência de Aminoácidos , Animais , Ciclo do Ácido Cítrico , Química Click , Creatina Quinase Forma MM/química , Creatina Quinase Forma MM/genética , Creatina Quinase Forma MM/metabolismo , Cisteína/química , Cisteína/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas de Escherichia coli/metabolismo , Glutationa/síntese química , Humanos , Malato Desidrogenase/antagonistas & inibidores , Malato Desidrogenase/metabolismo , Dados de Sequência Molecular , Peptídeos/análise , Peptídeos/química , Peptídeos/metabolismo , Processamento de Proteína Pós-Traducional , Proteômica , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
8.
Am J Physiol Cell Physiol ; 308(11): C919-31, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25810257

RESUMO

Myoblast fusion is critical for proper muscle growth and regeneration. During myoblast fusion, the localization of some molecules is spatially restricted; however, the exact reason for such localization is unknown. Creatine kinase B (CKB), which replenishes local ATP pools, localizes near the ends of cultured primary mouse myotubes. To gain insights into the function of CKB, we performed a yeast two-hybrid screen to identify CKB-interacting proteins. We identified molecules with a broad diversity of roles, including actin polymerization, intracellular protein trafficking, and alternative splicing, as well as sarcomeric components. In-depth studies of α-skeletal actin and α-cardiac actin, two predominant muscle actin isoforms, demonstrated their biochemical interaction and partial colocalization with CKB near the ends of myotubes in vitro. In contrast to other cell types, specific knockdown of CKB did not grossly affect actin polymerization in myotubes, suggesting other muscle-specific roles for CKB. Interestingly, knockdown of CKB resulted in significantly increased myoblast fusion and myotube size in vitro, whereas knockdown of creatine kinase M had no effect on these myogenic parameters. Our results suggest that localized CKB plays a key role in myotube formation by limiting myoblast fusion during myogenesis.


Assuntos
Creatina Quinase Forma BB/genética , Desenvolvimento Muscular/genética , Fibras Musculares Esqueléticas/enzimologia , Mioblastos/enzimologia , Actinas/genética , Actinas/metabolismo , Processamento Alternativo , Animais , Fusão Celular , Creatina Quinase Forma BB/antagonistas & inibidores , Creatina Quinase Forma BB/metabolismo , Creatina Quinase Forma MM/genética , Creatina Quinase Forma MM/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Fibras Musculares Esqueléticas/citologia , Mioblastos/citologia , Polimerização , Cultura Primária de Células , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transporte Proteico , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Transdução de Sinais , Técnicas do Sistema de Duplo-Híbrido
9.
Biochim Biophys Acta ; 1852(5): 732-41, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25615794

RESUMO

HHcy has been implicated in elderly frailty, but the underlying mechanisms are poorly understood. Using C57 and CBS+/- mice and C2C12 cell line, we investigated mechanisms behind HHcy induced skeletal muscle weakness and fatigability. Possible alterations in metabolic capacity (levels of LDH, CS, MM-CK and COX-IV), in structural proteins (levels of dystrophin) and in mitochondrial function (ATP production) were examined. An exercise regimen was employed to reverse HHcy induced changes. CBS+/- mice exhibited more fatigability, and generated less contraction force. No significant changes in muscle morphology were observed. However, there is a corresponding reduction in large muscle fiber number in CBS+/- mice. Excess fatigability was not due to changes in key enzymes involved in metabolism, but was due to reduced ATP levels. A marginal reduction in dystrophin levels along with a decrease in mitochondrial transcription factor A (mtTFA) were observed. There was also an increase in the mir-31, and mir-494 quantities that were implicated in dystrophin and mtTFA regulation respectively. The molecular changes elevated during HHcy, with the exception of dystrophin levels, were reversed after exercise. In addition, the amount of NRF-1, one of the transcriptional regulators of mtTFA, was significantly decreased. Furthermore, there was enhancement in mir-494 levels and a concomitant decline in mtTFA protein quantity in homocysteine treated cells. These changes in C2C12 cells were also accompanied by an increase in DNMT3a and DNMT3b proteins and global DNA methylation levels. Together, these results suggest that HHcy plays a causal role in enhanced fatigability through mitochondrial dysfunction which involves epigenetic changes.


Assuntos
Epigênese Genética , Hiper-Homocisteinemia/fisiopatologia , Mitocôndrias/metabolismo , Debilidade Muscular/fisiopatologia , Músculo Esquelético/fisiopatologia , Trifosfato de Adenosina/metabolismo , Animais , Western Blotting , Linhagem Celular , Creatina Quinase Forma MM/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , DNA Metiltransferase 3A , Proteínas de Ligação a DNA/metabolismo , Feminino , Expressão Gênica , Hiper-Homocisteinemia/genética , Hiper-Homocisteinemia/metabolismo , Técnicas In Vitro , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/genética , Proteínas Mitocondriais/metabolismo , Contração Muscular/genética , Contração Muscular/fisiologia , Debilidade Muscular/genética , Debilidade Muscular/metabolismo , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Fator 1 Nuclear Respiratório/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Natação , Fatores de Transcrição/metabolismo , DNA Metiltransferase 3B
10.
PLoS One ; 9(7): e102492, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25058652

RESUMO

Low temperatures may cause severe growth inhibition and mortality in fish. In order to understand the mechanism of cold tolerance, a transgenic zebrafish Tg (smyd1:m3ck) model was established to study the effect of energy homeostasis during cold stress. The muscle-specific promoter Smyd1 was used to express the carp muscle form III of creatine kinase (M3-CK), which maintained enzymatic activity at a relatively low temperature, in zebrafish skeletal muscle. In situ hybridization showed that M3-CK was expressed strongly in the skeletal muscle. When exposed to 13 °C, Tg (smyd1:m3ck) fish maintained their swimming behavior, while the wild-type could not. Energy measurements showed that the concentration of ATP increased in Tg (smyd1:m3ck) versus wild-type fish at 28 °C. After 2 h at 13 °C, ATP concentrations were 2.16-fold higher in Tg (smyd1:m3ck) than in wild-type (P<0.05). At 13 °C, the ATP concentration in Tg (smyd1:m3ck) fish and wild-type fish was 63.3% and 20.0%, respectively, of that in wild-type fish at 28 °C. Microarray analysis revealed differential expression of 1249 transcripts in Tg (smyd1:m3ck) versus wild-type fish under cold stress. Biological processes that were significantly overrepresented in this group included circadian rhythm, energy metabolism, lipid transport, and metabolism. These results are clues to understanding the mechanisms underlying temperature acclimation in fish.


Assuntos
Aclimatação/genética , Animais Geneticamente Modificados , Regulação da Expressão Gênica , Músculo Esquelético/metabolismo , Peixe-Zebra/genética , Trifosfato de Adenosina/metabolismo , Animais , Transporte Biológico , Carpas/genética , Carpas/metabolismo , Ritmo Circadiano/genética , Temperatura Baixa , Creatina Quinase Forma MM/genética , Creatina Quinase Forma MM/metabolismo , Metabolismo Energético/genética , Perfilação da Expressão Gênica , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Metabolismo dos Lipídeos , Anotação de Sequência Molecular , Regiões Promotoras Genéticas , Transgenes , Peixe-Zebra/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
11.
Cell Death Dis ; 5: e1094, 2014 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-24577092

RESUMO

MYOGENIN is a member of the muscle regulatory factor family that orchestrates an obligatory step in myogenesis, the terminal differentiation of skeletal muscle cells. A paradoxical feature of alveolar rhabdomyosarcoma (ARMS), a prevalent soft tissue sarcoma in children arising from cells with a myogenic phenotype, is the inability of these cells to undergo terminal differentiation despite the expression of MYOGENIN. The chimeric PAX3-FOXO1 fusion protein which results from a chromosomal translocation in ARMS has been implicated in blocking cell cycle arrest, preventing myogenesis from occurring. We report here that PAX3-FOXO1 enhances glycogen synthase kinase 3ß (GSK3ß) activity which in turn represses MYOGENIN activity. MYOGENIN is a GSK3ß substrate in vitro on the basis of in vitro kinase assays and MYOGENIN is phosphorylated in ARMS-derived RH30 cells. Constitutively active GSK3ß(S9A) increased the level of a phosphorylated form of MYOGENIN on the basis of western blot analysis and this effect was reversed by neutralization of the single consensus GSK3ß phosphoacceptor site by mutation (S160/164A). Congruently, GSK3ß inhibited the trans-activation of an E-box reporter gene by wild-type MYOGENIN, but not MYOGENIN with the S160/164A mutations. Functionally, GSK3ß repressed muscle creatine kinase (MCK) promoter activity, an effect which was reversed by the S160/164A mutated MYOGENIN. Importantly, GSK3ß inhibition or exogenous expression of the S160/164A mutated MYOGENIN in ARMS reduced the anchorage independent growth of RH30 cells in colony-formation assays. Thus, sustained GSK3ß activity represses a critical regulatory step in the myogenic cascade, contributing to the undifferentiated, proliferative phenotype in alveolar rhabdomyosarcoma (ARMS).


Assuntos
Quinase 3 da Glicogênio Sintase/metabolismo , Miogenina/metabolismo , Rabdomiossarcoma Alveolar/enzimologia , Animais , Células COS , Linhagem Celular Tumoral , Proliferação de Células , Chlorocebus aethiops , Creatina Quinase Forma MM/genética , Creatina Quinase Forma MM/metabolismo , Estimulação Elétrica , Ativação Enzimática , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Genótipo , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/genética , Glicogênio Sintase Quinase 3 beta , Camundongos , Mutação , Miogenina/genética , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/metabolismo , Fatores de Transcrição Box Pareados/genética , Fatores de Transcrição Box Pareados/metabolismo , Fenótipo , Fosforilação , Regiões Promotoras Genéticas , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Rabdomiossarcoma Alveolar/genética , Rabdomiossarcoma Alveolar/patologia , Fatores de Tempo , Transcrição Gênica , Transfecção
12.
Yi Chuan ; 35(5): 637-42, 2013 May.
Artigo em Chinês | MEDLINE | ID: mdl-23732671

RESUMO

Many intracellular signaling pathways regulate skeletal muscle differentiation. Among them, PI3K/AKT pathway plays an important role. But the mechanisms of chromatin regulation remain unclear. In this study, the murine C2C12 myoblast cell line was used to investigate the expression of Myogenin and MCK genes during the skeletal muscle differentiation. Western blotting analysis showed that the expression of Myogenin and MCK protein was increased significantly after PI3K/AKT activator treatment for 24 h during the C2C12 cell differentiation and the expression of H3K27me3 demethylase UTX was also increased. Chromatin immunoprecipitation (ChIP) and quantitative PCR (Q-PCR) analysis showed that the enrichment of H3K27me3 on the promoter regions of Myogenin and MCK genes and the enhancer region of MCK gene were decreased. It was opposite to the PI3K/AKT inhibitor treatment. We concluded that the PI3K/AKT pathway maybe regulate skeletal muscle differentiation by regulating the expression of UTX gene to change the enrichment of H3K27me3 on the target genes.


Assuntos
Creatina Quinase Forma MM/genética , Regulação da Expressão Gênica , Músculo Esquelético/metabolismo , Miogenina/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Diferenciação Celular , Linhagem Celular , Creatina Quinase Forma MM/metabolismo , Camundongos , Músculo Esquelético/citologia , Músculo Esquelético/enzimologia , Miogenina/genética , Fosfatidilinositol 3-Quinases/genética , Proteínas Proto-Oncogênicas c-akt/genética , Transdução de Sinais
13.
Oncogene ; 32(5): 651-62, 2013 Jan 31.
Artigo em Inglês | MEDLINE | ID: mdl-22710712

RESUMO

Rhabdomyosarcoma (RMS) is the commonest soft-tissue sarcoma in childhood and is characterized by expression of myogenic proteins, including the transcription factors MyoD and myogenin. There are two main subgroups, embryonal RMS and alveolar RMS (ARMS). Most ARMS are associated with chromosomal translocations that have breakpoints in introns of either PAX3 or PAX7, and FOXO1A. These translocations create chimeric transcription factors termed PAX3/FOXO1A and PAX7/FOXO1A respectively. Upon ectopic PAX3/FOXO1A expression, together with other genetic manipulation in mice, both differentiating myoblasts and satellite cells (the resident stem cells of postnatal muscle) can give rise to tumours with ARMS characteristics. As PAX3 and PAX7 are part of transcriptional networks that regulate muscle stem cell function in utero and during early postnatal life, PAX3/FOXO1A and PAX7/FOXO1A may subvert normal PAX3 and PAX7 functions. Here we examined how PAX3/FOXO1A and PAX7/FOXO1A affect myogenesis in satellite cells. PAX3/FOXO1A or PAX7/FOXO1A inhibited myogenin expression and prevented terminal differentiation in murine satellite cells: the same effect as dominant-negative (DN) Pax3 or Pax7 constructs. The transcription of MyoD-target genes myogenin and muscle creatine kinase were suppressed by PAX3/FOXO1A or PAX7/FOXO1A in C2C12 myogenic cells again as seen with Pax3/7DN. PAX3/FOXO1A or PAX7/FOXO1A did not inhibit the transcriptional activity of MyoD by perturbing MyoD expression, localization, phosphorylation or interaction with E-proteins. Chromatin immunoprecipitation on the myogenin promoter showed that PAX3/FOXO1A or PAX7/FOXO1A did not prevent MyoD from binding. However, PAX3/FOXO1A or PAX7/FOXO1A reduced occupation of the myogenin promoter by RNA polymerase II and decreased acetylation of histone H4, but did not directly bind to the myogenin promoter. Together, these observations reveal that PAX3/FOXO1A and PAX7/FOXO1A act to prevent myogenic differentiation via suppression of the transcriptional activation of MyoD-target genes.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Proteína MyoD/metabolismo , Proteínas de Fusão Oncogênica/metabolismo , Fator de Transcrição PAX7/metabolismo , Fatores de Transcrição Box Pareados/metabolismo , Rabdomiossarcoma Alveolar/genética , Células Satélites de Músculo Esquelético/metabolismo , Células-Tronco/metabolismo , Animais , Diferenciação Celular/genética , Linhagem Celular , Creatina Quinase Forma MM/genética , Proteína Forkhead Box O1 , Regulação da Expressão Gênica , Camundongos , Miogenina/genética , Fator de Transcrição PAX3
14.
Proc Natl Acad Sci U S A ; 109(50): 20590-5, 2012 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-23169664

RESUMO

There is no effective treatment for the cardiomyopathy of the most common autosomal recessive ataxia, Friedreich ataxia (FA). This disease is due to decreased expression of the mitochondrial protein, frataxin, which leads to alterations in mitochondrial iron (Fe) metabolism. The identification of potentially toxic mitochondrial Fe deposits in FA suggests Fe plays a role in its pathogenesis. Studies using the muscle creatine kinase (MCK) conditional frataxin knockout mouse that mirrors the disease have demonstrated frataxin deletion alters cardiac Fe metabolism. Indeed, there are pronounced changes in Fe trafficking away from the cytosol to the mitochondrion, leading to a cytosolic Fe deficiency. Considering Fe deficiency can induce apoptosis and cell death, we examined the effect of dietary Fe supplementation, which led to body Fe loading and limited the cardiac hypertrophy in MCK mutants. Furthermore, this study indicates a unique effect of heart and skeletal muscle-specific frataxin deletion on systemic Fe metabolism. Namely, frataxin deletion induces a signaling mechanism to increase systemic Fe levels and Fe loading in tissues where frataxin expression is intact (i.e., liver, kidney, and spleen). Examining the mutant heart, native size-exclusion chromatography, transmission electron microscopy, Mössbauer spectroscopy, and magnetic susceptibility measurements demonstrated that in the absence of frataxin, mitochondria contained biomineral Fe aggregates, which were distinctly different from isolated mammalian ferritin molecules. These mitochondrial aggregates of Fe, phosphorus, and sulfur, probably contribute to the oxidative stress and pathology observed in the absence of frataxin.


Assuntos
Ataxia de Friedreich/metabolismo , Ferro/metabolismo , Mitocôndrias Cardíacas/metabolismo , Animais , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Cardiomegalia/prevenção & controle , Creatina Quinase Forma MM/genética , Creatina Quinase Forma MM/metabolismo , Modelos Animais de Doenças , Ataxia de Friedreich/genética , Ataxia de Friedreich/patologia , Humanos , Ferro/sangue , Proteína 2 Reguladora do Ferro/metabolismo , Ferro da Dieta/administração & dosagem , Proteínas de Ligação ao Ferro/antagonistas & inibidores , Proteínas de Ligação ao Ferro/genética , Proteínas de Ligação ao Ferro/metabolismo , Fígado/metabolismo , Camundongos , Camundongos Knockout , Camundongos Mutantes , Microscopia Eletrônica de Transmissão , Miocárdio/metabolismo , Miocárdio/ultraestrutura , Transdução de Sinais , Espectroscopia de Mossbauer , Frataxina
15.
Am J Physiol Heart Circ Physiol ; 303(7): H844-52, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22886411

RESUMO

Reduced myofibrillar ATP availability during prolonged myocardial ischemia may limit post-ischemic mechanical function. Because creatine kinase (CK) is the prime energy reserve reaction of the heart and because it has been difficult to augment ATP synthesis during and after ischemia, we used mice that overexpress the myofibrillar isoform of creatine kinase (CKM) in cardiac-specific, conditional fashion to test the hypothesis that CKM overexpression increases ATP delivery in ischemic-reperfused hearts and improves functional recovery. Isolated, retrograde-perfused hearts from control and CKM mice were subjected to 25 min of global, no-flow ischemia and 40 min of reperfusion while cardiac function [rate pressure product (RPP)] was monitored. A combination of (31)P-nuclear magnetic resonance experiments at 11.7T and biochemical assays was used to measure the myocardial rate of ATP synthesis via CK (CK flux) and intracellular pH (pH(i)). Baseline CK flux was severalfold higher in CKM hearts (8.1 ± 1.0 vs. 32.9 ± 3.8, mM/s, control vs. CKM; P < 0.001) with no differences in phosphocreatine concentration [PCr] and RPP. End-ischemic pH(i) was higher in CKM hearts than in control hearts (6.04 ± 0.12 vs. 6.37 ± 0.04, control vs. CKM; P < 0.05) with no differences in [PCr] and [ATP] between the two groups. Post-ischemic PCr (66.2 ± 1.3 vs. 99.1 ± 8.0, %preischemic levels; P < 0.01), CK flux (3.2 ± 0.4 vs. 14.0 ± 1.2 mM/s; P < 0.001) and functional recovery (13.7 ± 3.4 vs. 64.9 ± 13.2%preischemic RPP; P < 0.01) were significantly higher and lactate dehydrogenase release was lower in CKM than in control hearts. Thus augmenting cardiac CKM expression attenuates ischemic acidosis, reduces injury, and improves not only high-energy phosphate content and the rate of CK ATP synthesis in postischemic myocardium but also recovery of contractile function.


Assuntos
Trifosfato de Adenosina/metabolismo , Creatina Quinase Forma MM/metabolismo , Metabolismo Energético , Contração Miocárdica , Isquemia Miocárdica/enzimologia , Traumatismo por Reperfusão Miocárdica/enzimologia , Miocárdio/enzimologia , Acidose/enzimologia , Acidose/fisiopatologia , Animais , Creatina Quinase Forma MM/genética , Modelos Animais de Doenças , Metabolismo Energético/genética , Concentração de Íons de Hidrogênio , Cinética , L-Lactato Desidrogenase/metabolismo , Espectroscopia de Ressonância Magnética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Contração Miocárdica/genética , Isquemia Miocárdica/genética , Isquemia Miocárdica/fisiopatologia , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Fosfocreatina/metabolismo , Regulação para Cima
16.
J Clin Invest ; 122(1): 291-302, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22201686

RESUMO

ATP is required for normal cardiac contractile function, and it has long been hypothesized that reduced energy delivery contributes to the contractile dysfunction of heart failure (HF). Despite experimental and clinical HF data showing reduced metabolism through cardiac creatine kinase (CK), the major myocardial energy reserve and temporal ATP buffer, a causal relationship between reduced ATP-CK metabolism and contractile dysfunction in HF has never been demonstrated. Here, we generated mice conditionally overexpressing the myofibrillar isoform of CK (CK-M) to test the hypothesis that augmenting impaired CK-related energy metabolism improves contractile function in HF. CK-M overexpression significantly increased ATP flux through CK ex vivo and in vivo but did not alter contractile function in normal mice. It also led to significantly increased contractile function at baseline and during adrenergic stimulation and increased survival after thoracic aortic constriction (TAC) surgery-induced HF. Withdrawal of CK-M overexpression after TAC resulted in a significant decline in contractile function as compared with animals in which CK-M overexpression was maintained. These observations provide direct evidence that the failing heart is "energy starved" as it relates to CK. In addition, these data identify CK as a promising therapeutic target for preventing and treating HF and possibly diseases involving energy-dependent dysfunction in other organs with temporally varying energy demands.


Assuntos
Creatina Quinase Forma MM/metabolismo , Insuficiência Cardíaca/fisiopatologia , Insuficiência Cardíaca/terapia , Trifosfato de Adenosina/biossíntese , Animais , Creatina Quinase Forma MM/genética , Modelos Animais de Doenças , Dobutamina/farmacologia , Metabolismo Energético , Expressão Gênica , Insuficiência Cardíaca/patologia , Técnicas In Vitro , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Contração Miocárdica/genética , Contração Miocárdica/fisiologia , Perfusão , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
17.
Cancer Invest ; 29(5): 353-9, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21599511

RESUMO

To determine the adeno-associated virus (AAV) serotype that most efficiently mediates muscle expression of antiangiogenic proteins, we injected four different serotype (1, 2, 7, and 8) AAV vectors encoding mouse endostatin (mEnd) or human soluble FLK-1 (hsFLK-1) into a quadriceps muscle of C57BL/6 mice. The highest plasma levels of therapeutic protein were observed in AAV8-injected mice (8 > 7 > 1 > 2). Sustained expression of mEnd was detected for 6 months, whereas concentrations of hsFLK-1 declined to the background level within 2 weeks caused by neutralizing anti-hsFLK-1 antibody. These data demonstrate that AAV8 (mEnd) serotype is the most efficient mediator for protein expression.


Assuntos
Dependovirus/metabolismo , Endostatinas/biossíntese , Técnicas de Transferência de Genes , Vetores Genéticos , Músculo Quadríceps/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/biossíntese , Animais , Anticorpos/sangue , Creatina Quinase Forma MM/genética , Dependovirus/classificação , Dependovirus/genética , Endostatinas/sangue , Endostatinas/genética , Ensaio de Imunoadsorção Enzimática , Células HeLa , Humanos , Injeções Intramusculares , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Regiões Promotoras Genéticas , Sorotipagem , Fatores de Tempo , Transfecção , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/sangue , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/imunologia
18.
Am J Physiol Regul Integr Comp Physiol ; 300(6): R1316-25, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21451138

RESUMO

Past simulations of oxidative ATP metabolism in skeletal muscle have predicted that elimination of the creatine kinase (CK) reaction should result in dramatically faster oxygen consumption dynamics during transitions in ATP turnover rate. This hypothesis was investigated. Oxygen consumption of fast-twitch (FT) muscle isolated from wild-type (WT) and transgenic mice deficient in the myoplasmic (M) and mitochondrial (Mi) CK isoforms (MiM CK(-/-)) were measured at 20°C at rest and during electrical stimulation. MiM CK(-/-) muscle oxygen consumption activation kinetics during a step change in contraction rate were 30% faster than WT (time constant 53 ± 3 vs. 69 ± 4 s, respectively; mean ± SE, n = 8 and 6, respectively). MiM CK(-/-) muscle oxygen consumption deactivation kinetics were 380% faster than WT (time constant 74 ± 4 s vs. 264 ± 4 s, respectively). Next, the experiments were simulated using a computational model of the oxidative ATP metabolic network in FT muscle featuring ADP and Pi feedback control of mitochondrial respiration (J. A. L. Jeneson, J. P. Schmitz, N. A. van den Broek, N. A. van Riel, P. A. Hilbers, K. Nicolay, J. J. Prompers. Am J Physiol Endocrinol Metab 297: E774-E784, 2009) that was reparameterized for 20°C. Elimination of Pi control via clamping of the mitochondrial Pi concentration at 10 mM reproduced past simulation results of dramatically faster kinetics in CK(-/-) muscle, while inclusion of Pi control qualitatively explained the experimental observations. On this basis, it was concluded that previous studies of the CK-deficient FT muscle phenotype underestimated the contribution of Pi to mitochondrial respiratory control.


Assuntos
Creatina Quinase Forma MM/deficiência , Creatina Quinase Forma MM/metabolismo , Mitocôndrias Musculares/fisiologia , Fibras Musculares de Contração Rápida/metabolismo , Músculo Esquelético/metabolismo , Consumo de Oxigênio/fisiologia , Fosfatos/metabolismo , Difosfato de Adenosina/metabolismo , Animais , Fenômenos Biomecânicos , Respiração Celular/fisiologia , Creatina Quinase Forma MM/genética , Cinética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Modelos Animais , Modelos Teóricos , Fenótipo
19.
Mol Biotechnol ; 49(1): 1-10, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21197588

RESUMO

Expression of a therapeutic gene in the skeletal muscle is a practical strategy to compensate a patients' insufficient circulating factor. Its clinical application requires a muscle-targeting vector capable of inducing a continuous high-level transgene expression. We modified an adeno-associated virus serotype 2 (AAV2) vector expressing luciferase from the mouse muscle creatine kinase gene promoter-enhancer (Ckm). First, AAVS1 insulator was inserted into the vector genome for transcriptional enhancement. This increased transduction of mouse quadriceps muscle by 11-fold at 4 weeks after intramuscular injection. Second, two capsid modifications were combined (21F capsid): incorporation of a segment of AAV1 capsid to produce a hybrid capsid and substitution of a tyrosine with a phenylalanine. Use of 21F capsid increased muscle transduction further by 18-fold, resulting in 200-fold higher efficacy than that of the unmodified vector. Compared with a vector having human elongation factor 1α promoter which showed similar efficacy in the muscle, this vector having Ckm transduced non-muscle organs less efficiently after intravenous administration. The AAV2 vector composed of the modified genome and capsid provides a backbone to develop a clinical vector expressing a therapeutic gene in the muscle.


Assuntos
Dependovirus/genética , Vetores Genéticos , Músculo Esquelético/citologia , Transdução Genética , Animais , Proteínas do Capsídeo/genética , Proteínas do Capsídeo/metabolismo , Linhagem Celular , Creatina Quinase Forma MM/genética , Creatina Quinase Forma MM/metabolismo , Feminino , Regulação Viral da Expressão Gênica , Terapia Genética , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Músculo Esquelético/metabolismo , Transgenes
20.
Methods Mol Biol ; 709: 3-19, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21194018

RESUMO

Gene therapy for muscular dystrophies requires efficient gene delivery to the striated musculature and specific, high-level expression of the therapeutic gene in a physiologically diverse array of muscles. This can be achieved by the use of recombinant adeno-associated virus vectors in conjunction with muscle-specific regulatory cassettes. We have constructed several generations of regulatory cassettes based on the enhancer and promoter of the muscle creatine kinase gene, some of which include heterologous enhancers and individual elements from other muscle genes. Since the relative importance of many control elements varies among different anatomical muscles, we are aiming to tailor these cassettes for high-level expression in cardiac muscle, and in fast and slow skeletal muscles. With the achievement of efficient intravascular gene delivery to isolated limbs, selected muscle groups, and heart in large animal models, the design of cassettes optimized for activity in different muscle types is now a practical goal. In this protocol, we outline the key steps involved in the design of regulatory cassettes for optimal activity in skeletal and cardiac muscle, and testing in mature muscle fiber cultures. The basic principles described here can also be applied to engineering tissue-specific regulatory cassettes for other cell types.


Assuntos
Terapia Genética , Músculo Esquelético/metabolismo , Distrofias Musculares/terapia , Miocárdio/metabolismo , Creatina Quinase Forma MM/genética , Dependovirus/genética , Elementos Facilitadores Genéticos , Expressão Gênica , Engenharia Genética , Vetores Genéticos , Humanos , Distrofias Musculares/genética , Regiões Promotoras Genéticas , Transcrição Gênica
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA