Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
3.
Diabetologia ; 55(1): 183-93, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21927895

RESUMO

AIMS/HYPOTHESIS: Although myostatin-null (Mstn (-/-)) mice fail to accumulate fat in adipose tissue when fed a high-fat diet (HFD), little is known about the molecular mechanism(s) behind this phenomenon. We therefore sought to identify the signalling pathways through which myostatin regulates accumulation and/or utilisation of fat. METHODS: Wild-type, Mstn (-/-) and wild-type mice treated with soluble activin type IIB receptor (sActRIIB) were fed a control chow diet or an HFD for 12 weeks. Changes in gene expression were measured by microarray and quantitative PCR. Histological changes in white adipose tissue were assessed together with peripheral tissue fatty acid oxidation and changes in circulating hormones following HFD feeding. RESULTS: Our results demonstrate that inactivation of myostatin results in reduced fat accumulation in mice on an HFD. Molecular analysis revealed that metabolic benefits, due to lack of myostatin, are mediated through at least two independent mechanisms. First, lack of myostatin increased fatty acid oxidation in peripheral tissues through induction of enzymes involved in lipolysis and in fatty acid oxidation in mitochondria. Second, inactivation of myostatin also enhanced brown adipose formation in white adipose tissue of Mstn (-/-) mice. Consistent with the above, treatment of HFD-fed wild-type mice with the myostatin antagonist, sActRIIB, reduced the obesity phenotype. CONCLUSIONS/INTERPRETATION: We conclude that absence of myostatin results in enhanced peripheral tissue fatty acid oxidation and increased thermogenesis, culminating in increased fat utilisation and reduced adipose tissue mass. Taken together, our data suggest that anti-myostatin therapeutics could be beneficial in alleviating obesity.


Assuntos
Tecido Adiposo Marrom/metabolismo , Adiposidade , Fármacos Antiobesidade/uso terapêutico , Dieta Hiperlipídica/efeitos adversos , Ácidos Graxos/metabolismo , Miostatina/antagonistas & inibidores , Obesidade/metabolismo , Receptores de Activinas Tipo II/química , Receptores de Activinas Tipo II/uso terapêutico , Tecido Adiposo Marrom/efeitos dos fármacos , Tecido Adiposo Marrom/patologia , Adiposidade/efeitos dos fármacos , Animais , Fármacos Antiobesidade/química , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/efeitos dos fármacos , Lipólise/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Terapia de Alvo Molecular , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/metabolismo , Miostatina/genética , Miostatina/metabolismo , Obesidade/etiologia , Obesidade/patologia , Obesidade/prevenção & controle , Oxirredução/efeitos dos fármacos , RNA Mensageiro/metabolismo , Solubilidade
4.
Diabetologia ; 54(6): 1491-501, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21347623

RESUMO

AIMS/HYPOTHESIS: Myostatin-null mice (Mstn(-/-)) have reduced body fat and increased tolerance to glucose. To date the molecular mechanisms through which myostatin regulates body fat content and insulin sensitivity are not known. Therefore, the aim of the current study was to identify signalling pathways through which myostatin regulates insulin sensitivity. METHODS: Wild-type (WT) mice and Mstn(-/-) mice were fed either a control chow diet or a high fat diet (HFD) for 12 weeks. Glucose tolerance testing and insulin stimulated glucose uptake by M. extensor digitorum longus (EDL) were used as variables to determine insulin sensitivity. Quantitative PCR, Western blotting and enzyme assays were used to monitor AMP-activated protein kinase (AMPK) levels and activity. RESULTS: Mstn(-/-) mice exhibited reduced fat accumulation and peripheral insulin resistance when compared with WT mice, even when they were fed an HFD. Furthermore, treatment with a myostatin antagonist also increased insulin sensitivity during HFD. Consistent with increased insulin sensitivity, we also detected elevated levels of GLUT4, AKT, p-AKT and insulin receptor substrate-1 in Mstn(-/-) muscles. Molecular analysis showed that there is increased expression and activity of AMPK in Mstn(-/-) muscles. Furthermore, we also observed an increase in the AMPK downstream target genes, Sirt1 and Pgc-1α (also known as Ppargc1a), in skeletal muscle of Mstn(-/-) mice. CONCLUSIONS/INTERPRETATION: We conclude that myostatin inactivation leads to increased AMPK levels and activity resulting in increased insulin sensitivity of skeletal muscle. We propose that, by regulating AMPK in skeletal muscle and adipose tissues, myostatin plays a major role in regulating insulin signalling.


Assuntos
Proteínas Quinases Ativadas por AMP/fisiologia , Resistência à Insulina/fisiologia , Miostatina/deficiência , Transdução de Sinais/fisiologia , Animais , Gorduras na Dieta/metabolismo , Glucose/metabolismo , Transportador de Glucose Tipo 4/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Músculo Esquelético/metabolismo , Miostatina/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo
5.
Ann Nutr Metab ; 50(5): 476-81, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16931880

RESUMO

BACKGROUND: Skeletal muscle mass is governed by multiple IGF-1-sensitive positive regulators of muscle-specific protein synthesis (myogenic regulatory factors which includes myoD, myogenin and Myf5) and negative regulators, including the atrogenic proteins myostatin, atrogin-1 and muscle ring finger 1 (MuRF-1). The coordinated control of these myogenic and atrogenic factors in human skeletal muscle following short-term fasting is currently unknown. METHOD: Healthy adults (n = 6, age 27.6 years) undertook a 40-hour fast. Skeletal muscle biopsy (vastus lateralis) and venous blood samples were taken 3, 15 and 40 h into the fast after an initial standard high-carbohydrate meal. Gene expression of the myogenic regulator factors (myoD, myogenin and Myf5) and the atrogenic factors (myostatin, atrogin-1 and MuRF-1) were determined by real-time PCR analysis. Plasma myostatin and IGF-1 were determined by ELISA. RESULTS: There were no significant alterations in either the positive or negative regulators of muscle mass at either 15 or 40 h, when compared to gene expression measured 3 h after a meal. Similarly, plasma myostatin and IGF-1 were also unaltered at these times. CONCLUSIONS: Unlike previous observations in catabolic and cachexic diseased states, short-term fasting (40 h) fails to elicit marked alteration of the genes regulating both muscle-specific protein synthesis or atrophy. Greater periods of fasting may be required to initiate coordinated inhibition of myogenic and atrogenic gene expression.


Assuntos
Jejum/metabolismo , Regulação da Expressão Gênica , Músculo Esquelético/metabolismo , Fatores de Regulação Miogênica/metabolismo , RNA Mensageiro/metabolismo , Adulto , Biópsia por Agulha , Feminino , Humanos , Fator de Crescimento Insulin-Like I/metabolismo , Masculino , Proteínas Musculares/metabolismo , Proteína MyoD/metabolismo , Fator Regulador Miogênico 5/metabolismo , Miogenina/metabolismo , Miostatina , Reação em Cadeia da Polimerase/métodos , Proteínas Ligases SKP Culina F-Box/metabolismo , Fatores de Tempo , Fator de Crescimento Transformador beta/sangue , Fator de Crescimento Transformador beta/metabolismo , Proteínas com Motivo Tripartido , Ubiquitina-Proteína Ligases/metabolismo
6.
J Endocrinol ; 176(3): 425-37, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12630927

RESUMO

The IGF axis is nutritionally sensitive in vivo and IGFs stimulate myoblast proliferation and differentiation in vitro, while myostatin inhibits these processes in vitro. We hypothesised that underfeeding would reversibly inhibit the myogenic activity of satellite cells in vivo together with decreased IGF-I and increased myostatin in muscle. Satellite cell activity was measured indirectly from the expression of proliferating cell nuclear antigen (PCNA) and the myogenic regulatory factors (MRFs), MyoD, Myf-5 and myogenin. Young sheep were underfed (30% of maintenance) and some killed after 1, 4, 12, 17, 21 and 22 weeks. Remaining underfed animals were then re-fed a control ration of pellets and killed after 2 days, and 1, 6 and 30 weeks. Expression of PCNA and MRFs decreased during the first week of underfeeding. This coincided with reduced IGF-I and myostatin mRNA, and processed myostatin. Subsequently, Myf-5, MyoD, myostatin mRNA and processed myostatin increased, suggesting that satellite cells may have become progressively quiescent. Long-term underfeeding caused muscle necrosis in some animals and IGF-I and MRF expression was increased in these, indicating the activation of satellite cells for muscle repair. Re-feeding initiated rapid muscle growth and increased expression of PCNA, IGF-I and the MRFs concurrently with decreased myostatin proteins. In conclusion, these data indicate that IGF-I and myostatin may work in a coordinated manner to regulate the proliferation, differentiation and quiescence of satellite cells in vivo.


Assuntos
Proteínas de Ligação a DNA , Fator de Crescimento Insulin-Like I/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Miogenina/metabolismo , Distúrbios Nutricionais/metabolismo , Transativadores , Fator de Crescimento Transformador beta/metabolismo , Adaptação Fisiológica , Animais , Northern Blotting/métodos , Western Blotting/métodos , Feminino , Imuno-Histoquímica/métodos , Fator de Crescimento Insulin-Like I/genética , Músculo Esquelético/citologia , Proteína MyoD/genética , Proteína MyoD/metabolismo , Fator Regulador Miogênico 5 , Miostatina , Antígeno Nuclear de Célula em Proliferação/genética , Antígeno Nuclear de Célula em Proliferação/metabolismo , RNA Mensageiro/análise , Distribuição Aleatória , Ovinos , Fatores de Tempo , Fator de Crescimento Transformador beta/genética
7.
Exerc Sport Sci Rev ; 29(4): 155-8, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11688787

RESUMO

Myostatin, a member of the TGF beta superfamily, regulates skeletal muscle size by controlling embryonic myoblast proliferation. Recent results show that myostatin may also have a role in muscle regeneration and muscle wasting of adult animals. This review summarizes the recent developments in the regulation of myostatin gene expression and mechanism of its function.


Assuntos
Músculo Esquelético/fisiologia , Regeneração/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Animais , Expressão Gênica , Homeostase/fisiologia , Humanos , Músculo Esquelético/embriologia , Miostatina , Transcrição Gênica , Fator de Crescimento Transformador beta/genética
8.
Mol Cell Biochem ; 220(1-2): 31-7, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11451380

RESUMO

Myostatin belongs to the Transforming Growth Factor-beta (TGF-beta) superfamily and is expressed in developing and mature skeletal muscle. Biologically, the role of myostatin seems to be extremely well conserved during evolution since inactivating mutations in myostatin gene cause similar phenotype of heavy muscling in both mice and cattle. In this report we have analysed the genomic structure and neonatal expression of the bovine myostatin gene. The molecular analysis shows that the bovine myostatin gene consists of three exons and two introns. The sizes of the first and second exons are 506 and 374 base pairs (bp) respectively. The size of the third exon was found to be variable in length (1701 or 1812 or 1887 nucleotides), whereas the size of the two introns is 1840 and 2033 bps. In the first exon of bovine myostatin, a single transcription initiation site is found at 133 bps from the translation start codon ATG. Sequencing the 3' untranslated region indicated that there are multiple polyadenylation signals at 1301, 1401 and 1477 bp downstream from the translation stop codon (TGA). Furthermore, 3' RACE analysis confirmed that all three polyadenylation sites are used in vivo. Using quantitative RT-PCR we have analysed neonatal expression of myostatin gene. In both the M. biceps femoris and M. semitendinosus, the highest level of myostatin expression was observed on day 1 postnatally, then gradually reduced on days 8 and 14 postnatally. In contrast, in the M. gastrocnemius, myostatin expression was highest on day 14 and lowest on day 8. These results indicate that myostatin gene structure and function is well conserved during evolution and that neonatal expression of myostatin in a number of predominantly fast twitch muscles is differentially regulated.


Assuntos
Fator de Crescimento Transformador beta/biossíntese , Fator de Crescimento Transformador beta/genética , Regiões 3' não Traduzidas , Animais , Sequência de Bases , Bovinos , Códon de Terminação , DNA Complementar/metabolismo , Evolução Molecular , Éxons , Íntrons , Modelos Genéticos , Dados de Sequência Molecular , Fibras Musculares de Contração Rápida/metabolismo , Músculo Esquelético/embriologia , Miostatina , Fenótipo , Poliadenilação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Crescimento Transformador beta/metabolismo
9.
Am J Physiol Regul Integr Comp Physiol ; 280(5): R1488-93, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11294773

RESUMO

Excessive muscling in double-muscled cattle arises from mutations in the myostatin gene, but the role of myostatin in normal muscle development is unclear. The aim of this study was to measure the temporal relationship of myostatin and myogenic regulatory factors during muscle development in normal (NM)- and double-muscled (DM) cattle to determine the timing and possible targets of myostatin action in vivo. Myostatin mRNA peaked at the onset of secondary fiber formation (P < 0.001) and was greater in DM (P < 0.001) than in NM. MyoD expression was also elevated throughout primary and secondary fiber formation (P < 0.001) and greater in DM (P < 0.05). Expression of myogenin peaked later than MyoD (P < 0.05); however, it did not differ between NM and DM. These data show that myostatin and MyoD increase coincidentally during formation of muscle fibers, indicating a coordinated role in the terminal differentiation and/or fusion of myoblasts. Myostatin mRNA is also consistently higher in DM than NM, suggesting that a feedback loop of regulation is also disrupted in the myostatin-deficient condition.


Assuntos
Bovinos/anormalidades , Deleção de Genes , Músculo Esquelético/embriologia , Proteína MyoD/genética , Fator de Crescimento Transformador beta/genética , Animais , Desenvolvimento Embrionário e Fetal , Feto , Idade Gestacional , Membro Posterior , Miostatina , Fenótipo , RNA Mensageiro/análise , RNA Mensageiro/genética , Valores de Referência , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transcrição Gênica , Fator de Crescimento Transformador beta/deficiência
11.
J Biol Chem ; 275(51): 40235-43, 2000 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-10976104

RESUMO

Myostatin, a member of the transforming growth factor-beta (TGF-beta) superfamily, has been shown to be a negative regulator of myogenesis. Here we show that myostatin functions by controlling the proliferation of muscle precursor cells. When C(2)C(12) myoblasts were incubated with myostatin, proliferation of myoblasts decreased with increasing levels of myostatin. Fluorescence-activated cell sorting analysis revealed that myostatin prevented the progression of myoblasts from the G(1)- to S-phase of the cell cycle. Western analysis indicated that myostatin specifically up-regulated p21(Waf1, Cip1), a cyclin-dependent kinase inhibitor, and decreased the levels and activity of Cdk2 protein in myoblasts. Furthermore, we also observed that in myoblasts treated with myostatin protein, Rb was predominately present in the hypophosphorylated form. These results suggests that, in response to myostatin signaling, there is an increase in p21 expression and a decrease in Cdk2 protein and activity thus resulting in an accumulation of hypophosphorylated Rb protein. This, in turn, leads to the arrest of myoblasts in G(1)-phase of cell cycle. Thus, we propose that the generalized muscular hyperplasia phenotype observed in animals that lack functional myostatin could be as a result of deregulated myoblast proliferation.


Assuntos
Divisão Celular/fisiologia , Músculo Esquelético/citologia , Fator de Crescimento Transformador beta/fisiologia , Animais , Sequência de Bases , Bovinos , Ciclo Celular , Células Cultivadas , Primers do DNA , Hidrólise , Miostatina , Fosforilação , Proteína do Retinoblastoma/metabolismo
12.
J Cell Physiol ; 184(3): 356-63, 2000 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-10911367

RESUMO

Myostatin, a member of the TGF-beta superfamily, is a key negative regulator of skeletal muscle growth. The role of myostatin during skeletal muscle regeneration has not previously been reported. In the present studies, normal Sprague-Dawley and growth hormone (GH)-deficient (dw/dw) rats were administered the myotoxin, notexin, in the right M. biceps femoris on day 0. The dw/dw rats then received either saline or human-N-methionyl GH (200microg/100g body weight/day) during the ensuing regeneration. Normal and dw/dw M. biceps femoris were dissected on days 1, 2, 3, 5, 9 and 13, formalin-fixed, then immunostained for myostatin protein. Immunostaining for myostatin revealed high levels of protein within necrotic fibres and connective tissue of normal and dw/dw damaged muscles. Regenerating myotubes contained no myostatin at the time of fusion (peak fusion on day 5), and only low levels of myostatin were observed during subsequent myotube enlargement. Fibres which survived assault by notexin (survivor fibres) contained moderate to high myostatin immunostaining initially. The levels in both normal and dw/dw rat survivor fibres decreased on days 2-3, then increased on days 9-13. In dw/dw rats, there was no observed effect of GH administration on the levels of myostatin protein in damaged muscle. The low level of myostatin observed in regenerating myotubes in these studies suggests a negative regulatory role for myostatin in muscle regeneration.


Assuntos
Músculo Esquelético/fisiologia , Regeneração/fisiologia , Fator de Crescimento Transformador beta/fisiologia , Animais , Nanismo/genética , Nanismo/fisiopatologia , Hormônio do Crescimento Humano/farmacologia , Humanos , Imuno-Histoquímica , Masculino , Fibras Musculares Esqueléticas/fisiologia , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/lesões , Miostatina , Necrose , Ratos , Ratos Mutantes , Ratos Sprague-Dawley , Regeneração/efeitos dos fármacos
13.
Domest Anim Endocrinol ; 17(2-3): 191-7, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10527122

RESUMO

The enlarged muscles of certain breeds of cattle, such as the Belgian Blue, have been shown to result from a marked increase in the number of normal sized muscle fibers. Originally insulin-like growth factors (IGFs) were implicated in this myofiber hyperplasia, as IGFs have been shown to stimulate myoblast proliferation as well as maintain fiber differentiation. Recently it has been reported that mice lacking a myostatin gene, a member of the TGFbeta superfamily, have enhanced skeletal mass resulting from increased muscle fiber number and size. Mutations in this gene have been found in double-muscled cattle, indicating that myostatin is an inhibitor of muscle growth. Myostatin is expressed early in gestation and then maintained to adulthood in certain muscles. Myostatin expression in bovine muscle is highest during gestation when muscle fibers are forming and some of the myogenic regulatory factors have elevated expression over the same period as myostatin. Molecular expression of the IGF axis does not differ between Belgian Blue and normal muscled cattle, and IGF-II mRNA is increased throughout formation of secondary fibers in both breeds. However, myostatin and MyoD expression in muscle differ between normal and hypertrophied muscle cattle breeds. This evidence strongly suggests that lack of myostatin is associated with an increase in fiber number which then results in a marked increase in potential muscle mass in double-muscled cattle.


Assuntos
Bovinos/fisiologia , Substâncias de Crescimento/fisiologia , Músculo Esquelético/fisiologia , Fator de Crescimento Transformador beta/genética , Sequência de Aminoácidos , Animais , Feminino , Feto/fisiologia , Hiperplasia/genética , Hiperplasia/veterinária , Fator de Crescimento Insulin-Like II/genética , Fator de Crescimento Insulin-Like II/fisiologia , Dados de Sequência Molecular , Desenvolvimento Muscular , Músculo Esquelético/crescimento & desenvolvimento , Proteína MyoD/genética , Proteína MyoD/fisiologia , Miostatina , Mutação Puntual , Gravidez , Alinhamento de Sequência , Fator de Crescimento Transformador beta/fisiologia
14.
J Cell Physiol ; 180(1): 1-9, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10362012

RESUMO

Myostatin is a secreted growth and differentiating factor (GDF-8) that belongs to the transforming growth factor-beta (TGF-beta) superfamily. Targeted disruption of the myostatin gene in mice and a mutation in the third exon of the myostatin gene in double-muscled Belgian Blue cattle breed result in skeletal muscle hyperplasia. Hence, myostatin has been shown to be involved in the regulation of skeletal muscle mass in both mice and cattle. Previous published reports utilizing Northern hybridization had shown that myostatin expression was seen exclusively in skeletal muscle. A significantly lower level of myostatin mRNA was also reported in adipose tissue. Using a sensitive reverse transcription-polymerase chain reaction (RT-PCR) technique and Western blotting with anti-myostatin antibodies, we show that myostatin mRNA and protein are not restricted to skeletal muscle. We also show that myostatin expression is detected in the muscle of both fetal and adult hearts. Sequence analysis reveals that the Belgian Blue heart myostatin cDNA sequence contains an 11 nucleotide deletion in the third exon that causes a frameshift that eliminates virtually all of the mature, active region of the protein. Anti-myostatin immunostaining on heart sections also demonstrates that myostatin protein is localized in Purkinje fibers and cardiomyocytes in heart tissue. Furthermore, following myocardial infarction, myostatin expression is upregulated in the cardiomyocytes surrounding the infarct area. Given that myostatin is expressed in fetal and adult hearts and that myostatin expression is upregulated in cardiomyocytes after the infarction, myostatin could play an important role in cardiac development and physiology.


Assuntos
Fibras Musculares Esqueléticas/fisiologia , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Fator de Crescimento Transformador beta/genética , Animais , Sequência de Bases , Western Blotting , Bovinos , Sequência Conservada , DNA Complementar , Modelos Animais de Doenças , Regulação da Expressão Gênica no Desenvolvimento , Mamíferos , Dados de Sequência Molecular , Fibras Musculares Esqueléticas/química , Músculo Esquelético/química , Músculo Esquelético/citologia , Músculo Esquelético/fisiologia , Mutação/fisiologia , Miocárdio/química , Miocárdio/citologia , Miostatina , RNA Mensageiro/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos , Ovinos , Fator de Crescimento Transformador beta/análise , Fator de Crescimento Transformador beta/metabolismo , Regulação para Cima/genética
15.
Am J Physiol ; 276(4): C873-82, 1999 04.
Artigo em Inglês | MEDLINE | ID: mdl-10199818

RESUMO

The cloned epithelial cell-specific Na+/H+ exchanger (NHE) isoform NHE2 is stimulated by fibroblast growth factor (FGF), phorbol 12-myristate 13-acetate (PMA), okadaic acid (OA), and fetal bovine serum (FBS) through a change in maximal velocity of the transporter. In the present study, we used COOH-terminal truncation mutants to delineate specific domains in the COOH terminus of NHE2 that are responsible for growth factor and/or protein kinase regulation. Five truncation mutants (designated by the amino acid number at the truncation site) were stably expressed in NHE-deficient PS120 fibroblasts. The effects of PMA, FGF, OA, FBS, and W-13 [a Ca2+/calmodulin (CaM) inhibitor] were studied. Truncation mutant E2/660, but not E2/573, was stimulated by PMA. OA stimulated E2/573 but not E2/540. FGF stimulated E2/540 but not E2/499. The most truncated mutant, E2/499, was stimulated by FBS. W-13 stimulated the basal activity of the wild-type NHE2. However, W-13 had no effect on E2/755. By monitoring the emission spectra of dansylated CaM fluorescence, we showed that dansylated CaM bound directly to a purified fusion protein of glutathione S-transferase and the last 87 amino acids of NHE2 in a Ca2+-dependent manner, with a stoichiometry of 1:1 and a dissociation constant of 300 nM. Our results showed that the COOH terminus of NHE2 is organized into separate stimulatory and inhibitory growth factor/protein kinase regulatory subdomains. This organization of growth factor/protein kinase regulatory subdomains is very similar to that of NHE3, suggesting that the tertiary structures of the putative COOH termini of NHE2 and NHE3 are very similar despite the minimal amino acid identity in this part of the two proteins.


Assuntos
Fatores de Crescimento de Fibroblastos/farmacologia , Proteínas Quinases/metabolismo , Trocadores de Sódio-Hidrogênio/química , Trocadores de Sódio-Hidrogênio/metabolismo , Animais , Bovinos , Linhagem Celular , Clonagem Molecular , Cricetinae , Cricetulus , Concentração de Íons de Hidrogênio , Cinética , Pulmão , Mutagênese Sítio-Dirigida , Ácido Okadáico/farmacologia , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Deleção de Sequência , Sódio/metabolismo , Trocadores de Sódio-Hidrogênio/genética , Sulfonamidas/farmacologia , Acetato de Tetradecanoilforbol/farmacologia , Transfecção
16.
Genes Dev ; 12(2): 246-60, 1998 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-9436984

RESUMO

POU transcription factors participate in cell-identity decisions during nervous system development, yet little is known about the regulatory networks controlling their expression. We report all known Drosophila POU genes require castor (cas) for correct CNS expression. drifter and I-POU depend on cas for full expression, whereas pdm-1 and pdm-2 are negatively regulated. cas encodes a zinc finger protein that shares DNA-binding specificity with another pdm repressor: the gap segmentation gene regulator Hunchback (Hb). Our studies reveal that the embryonic CNS contains sequentially generated neuroblast sublineages that can be distinguished by their expression of either Hb, Pdm-1, or Cas. Hb and Cas may directly silence pdm expression in early and late developing sublineages, given that pdm-1 cis-regulatory DNA contains >=32 Hb/Cas-binding sites and its enhancer(s) are ectopically activated in cas- neuroblasts. In addition, the targeted misexpression of Cas in all neuroblast lineages reduces Pdm-1 expression without altering Hb expression. By ensuring correct POU gene expression boundaries, hb and cas maintain temporal subdivisions in the cell-identity circuitry controlling CNS development.


Assuntos
Sistema Nervoso Central/embriologia , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila , Drosophila/genética , Regulação da Expressão Gênica , Fatores de Transcrição/metabolismo , Animais , Sequência de Bases , Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/genética , Elementos Facilitadores Genéticos , Técnica Indireta de Fluorescência para Anticorpo , Proteínas de Homeodomínio/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Microscopia Confocal , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/metabolismo , Fatores do Domínio POU , Fatores de Transcrição/genética
17.
J Virol ; 71(12): 9792-5, 1997 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9371647

RESUMO

Rat Mx2 and rat Mx3 are two alpha/beta interferon-inducible cytoplasmic GTPases that differ in three residues in the amino-terminal third, which also contains the tripartite GTP-binding domain, and that differ in five residues in the carboxy-terminal quarter, which also contains a dimerization domain. While Mx2 is active against vesicular stomatitis virus (VSV), Mx3 lacks antiviral activity. We mapped the functional difference between Mx2 and Mx3 protein to two critical residues in the carboxy-terminal parts of the molecules. An exchange of either residue 588 or 630 of Mx2 with the corresponding residues of Mx3 abolished anti-VSV activity, and the introduction of the two Mx2 residues on an Mx3 background partially restored anti-VSV activity. These results are consistent with the facts that Mx2 and Mx3 have similar intrinsic GTPase activities and that the GTPase domain of Mx3 can fully substitute for the GTPase domain of Mx2. Nevertheless, the amino-terminal third containing the GTP-binding domain is necessary for antiviral activity, since an amino-terminally truncated Mx2 protein is devoid of anti-VSV activity. Furthermore, Fab fragments of a monoclonal antibody known to neutralize antiviral activity block GTPase activity by binding an epitope in the carboxy-terminal half of Mx2 or Mx3 protein. The results are consistent with a two-domain model in which both the conserved amino-terminal half and the less-well-conserved carboxy-terminal half of Mx proteins carry functionally important domains.


Assuntos
Antivirais/imunologia , Epitopos/imunologia , GTP Fosfo-Hidrolases/imunologia , Proteínas de Ligação ao GTP , Proteínas/imunologia , Vírus da Estomatite Vesicular Indiana/imunologia , Células 3T3 , Animais , Antivirais/genética , GTP Fosfo-Hidrolases/genética , Camundongos , Proteínas de Resistência a Myxovirus , Proteínas/genética , Ratos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia
18.
Genome Res ; 7(9): 910-6, 1997 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-9314496

RESUMO

A visibly distinct muscular hypertrophy (mh), commonly known as double muscling, occurs with high frequency in the Belgian Blue and Piedmontese cattle breeds. The autosomal recessive mh locus causing double-muscling condition in these cattle maps to bovine chromosome 2 within the same interval as myostatin, a member of the TGF-beta superfamily of genes. Because targeted disruption of myostatin in mice results in a muscular phenotype very similar to that seen in double-muscled cattle, we have evaluated this gene as a candidate gene for double-muscling condition by cloning the bovine myostatin cDNA and examining the expression pattern and sequence of the gene in normal and double-muscled cattle. The analysis demonstrates that the levels and timing of expression do not appear to differ between Belgian Blue and normal animals, as both classes show expression initiating during fetal development and being maintained in adult muscle. Moreover, sequence analysis reveals mutations in heavy-muscled cattle of both breeds. Belgian Blue cattle are homozygous for an 11-bp deletion in the coding region that is not detected in cDNA of any normal animals examined. This deletion results in a frame-shift mutation that removes the portion of the Myostatin protein that is most highly conserved among TGF-beta family members and that is the portion targeted for disruption in the mouse study. Piedmontese animals tested have a G-A transition in the same region that changes a cysteine residue to a tyrosine. This mutation alters one of the residues that are hallmarks of the TGF-beta family and are highly conserved during evolution and among members of the gene family. It therefore appears likely that the mh allele in these breeds involves mutation within the myostatin gene and that myostatin is a negative regulator of muscle growth in cattle as well as mice.


Assuntos
Bovinos/genética , Genes , Músculos/ultraestrutura , Fator de Crescimento Transformador beta/genética , Alelos , Sequência de Aminoácidos , Animais , Sequência Conservada , DNA Complementar , Mutação da Fase de Leitura , Expressão Gênica , Hipertrofia/genética , Camundongos , Dados de Sequência Molecular , Músculos/patologia , Miostatina , Fenótipo , Reação em Cadeia da Polimerase , RNA/isolamento & purificação , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Especificidade da Espécie
19.
J Biol Chem ; 271(34): 20641-9, 1996 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-8702812

RESUMO

A detailed understanding of the mechanism of lipid transport in insects has been hampered by the inability to identify the proapolipophorin gene that encodes apolipophorins I and II, the principal protein components of lipophorin, the lipid transport vehicle. Here we provide the first molecular description of the Drosophila gene encoding a retinoid- and fatty acid-binding glycoprotein (RFABG) and present evidence that it is a member of the proapolipophorin gene family. The gene, localized to the chromosome 4 (102 F region), encodes a 3351-amino acid protein that could serve as the precursor for the approximately 70-kDa and >200-kDa polypeptides associated with RFABG. The N-terminal sequence of the approximately 70-kDa polypeptide and that predicted for the >200-kDa polypeptide showed high sequence similarity to blowfly apolipophorin II and apolipophorin I, respectively. The RFABG precursor contains a signal peptide and exhibits a multidomain mosaic protein structure, which is typical of extracellular proteins. It has structural domains similar to lipid-binding proteins, namely vitellogenins and apolipoprotein B. The protein also contains a domain similar to the D domain of von Willebrand factor and mucin. The gene is expressed in the Drosophila embryo during development in cells that make up the amnioserosa and fat bodies. Immunolocalizations using specific antibodies against RFABG reveal that the protein is initially dispersed through the embryonic amnioserosa sac and latter concentrated at skeletal muscle-epidermis apodemeal contact junctions during larval development. This novel gene may play an important role in the transport of lipids, including retinoids and fatty acids, in insects.


Assuntos
Proteínas de Transporte/genética , Proteínas de Drosophila , Drosophila melanogaster/genética , Genes de Insetos , Glicoproteínas/genética , Lipoproteínas/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Proteínas de Transporte/metabolismo , Mapeamento Cromossômico , Proteínas de Ligação a Ácido Graxo , Ácidos Graxos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Glicoproteínas/metabolismo , Dados de Sequência Molecular , Estrutura Secundária de Proteína , RNA Mensageiro/genética , Retinoides/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...