Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Oncogene ; 35(50): 6378-6388, 2016 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-27132508

RESUMO

Cancer cells are known to execute reprogramed metabolism of glucose, amino acids and lipids. Here, we report a significant role of cholesterol metabolism in cancer metastasis. By using label-free Raman spectromicroscopy, we found an aberrant accumulation of cholesteryl ester in human pancreatic cancer specimens and cell lines, mediated by acyl-CoA cholesterol acyltransferase-1 (ACAT-1) enzyme. Expression of ACAT-1 showed a correlation with poor patient survival. Abrogation of cholesterol esterification, either by an ACAT-1 inhibitor or by shRNA knockdown, significantly suppressed tumor growth and metastasis in an orthotopic mouse model of pancreatic cancer. Mechanically, ACAT-1 inhibition increased intracellular free cholesterol level, which was associated with elevated endoplasmic reticulum stress and caused apoptosis. Collectively, our results demonstrate a new strategy for treating metastatic pancreatic cancer by inhibiting cholesterol esterification.


Assuntos
Ésteres do Colesterol/metabolismo , Neoplasias Pancreáticas/patologia , Acetil-CoA C-Acetiltransferase/antagonistas & inibidores , Acetil-CoA C-Acetiltransferase/fisiologia , Animais , Apoptose , Linhagem Celular Tumoral , Proliferação de Células , Estresse do Retículo Endoplasmático , Esterificação , Humanos , Masculino , Camundongos , Metástase Neoplásica , PTEN Fosfo-Hidrolase/fisiologia , Neoplasias Pancreáticas/metabolismo
2.
Oncogene ; 32(15): 1950-8, 2013 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-22665051

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers owing to a number of characteristics including difficulty in establishing early diagnosis and the absence of effective therapeutic regimens. A large number of genetic alterations have been ascribed to PDAC with mutations in the KRAS2 proto-oncogene thought to be an early event in the progression of disease. Recent lineage-tracing studies have shown that acinar cells expressing mutant Kras(G12D) are induced to transdifferentiate, generating duct-like cells through a process known as acinar-ductal metaplasia (ADM). ADM lesions then convert to precancerous pancreatic intraepithelial neoplasia (PanIN) that progresses to PDAC over time. Thus, understanding the earliest events involved in ADM/PanIN formation would provide much needed information on the molecular pathways that are instrumental in initiating this disease. As studying the transition of acinar cells to metaplastic ductal cells in vivo is complicated by analysis of the entire organ, an in vitro three dimensional (3D) culture system was used to model ADM outside the animal. Kras(G12D)-expressing acinar cells rapidly underwent ADM in 3D culture, forming ductal cysts that silenced acinar genes and activated duct genes, characteristics associated with in vivo ADM/PanIN lesions. Analysis of downstream KRAS signaling events established a critical importance for the Raf/MEK/ERK pathway in ADM induction. In addition, forced expression of the acinar-restricted transcription factor Mist1, which is critical to acinar cell organization, significantly attenuated Kras(G12D)-induced ADM/PanIN formation. These results suggest that maintaining MIST1 activity in Kras(G12D)-expressing acinar cells can partially mitigate the transformation activity of oncogenic KRAS. Future therapeutics that target both the MAPK pathway and Mist1 transcriptional networks may show promising efficacy in combating this deadly disease.


Assuntos
Células Acinares/fisiologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/patologia , Metaplasia/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Animais , Carcinoma in Situ/genética , Carcinoma in Situ/metabolismo , Linhagem Celular , Transformação Celular Neoplásica , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Metaplasia/patologia , Camundongos , Camundongos Transgênicos , Ductos Pancreáticos/patologia , Neoplasias Pancreáticas/metabolismo , Lesões Pré-Cancerosas/genética , Lesões Pré-Cancerosas/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/genética , Transdução de Sinais , Quinases raf/metabolismo
3.
J Cell Biol ; 155(4): 519-30, 2001 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-11696558

RESUMO

The pancreas is a complex organ that consists of separate endocrine and exocrine cell compartments. Although great strides have been made in identifying regulatory factors responsible for endocrine pancreas formation, the molecular regulatory circuits that control exocrine pancreas properties are just beginning to be elucidated. In an effort to identify genes involved in exocrine pancreas function, we have examined Mist1, a basic helix-loop-helix transcription factor expressed in pancreatic acinar cells. Mist1-null (Mist1(KO)) mice exhibit extensive disorganization of exocrine tissue and intracellular enzyme activation. The exocrine disorganization is accompanied by increases in p8, RegI/PSP, and PAP1/RegIII gene expression, mimicking the molecular changes observed in pancreatic injury. By 12 m, Mist1(KO) mice develop lesions that contain cells coexpressing acinar and duct cell markers. Analysis of the factors involved in cholecystokinin (CCK) signaling reveal inappropriate levels of the CCK receptor A and the inositol-1,4,5-trisphosphate receptor 3, suggesting that a functional defect exists in the regulated exocytosis pathway of Mist1(KO) mice. Based on these observations, we propose that Mist1(KO) mice represent a new genetic model for chronic pancreas injury and that the Mist1 protein serves as a key regulator of acinar cell function, stability, and identity.


Assuntos
Sequências Hélice-Alça-Hélice , Pâncreas/citologia , Transativadores , Fatores de Transcrição/fisiologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Colecistocinina/metabolismo , Proteínas do Citoesqueleto/biossíntese , Desenvolvimento Embrionário e Fetal , Feminino , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pâncreas/embriologia , Pâncreas/lesões , Proteínas Associadas a Pancreatite , Fenótipo , Fosfoproteínas/metabolismo , Transdução de Sinais , Fatores de Transcrição/genética , Proteína da Zônula de Oclusão-1 , beta Catenina
5.
Oncogene ; 20(11): 1276-86, 2001 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-11313872

RESUMO

Oncogenic Ras (H-Ras G12V) inhibits skeletal myogenesis through multiple signaling pathways. Previously, we demonstrated that the major downstream effectors of Ras (i.e., MEK/MAPK, RalGDS and Rac/Rho) play a minor, if any, role in the differentiation-defective phenotype of Ras myoblasts. Recently, NFkappaB, another Ras signaling target, has been shown to inhibit myogenesis presumably by stimulating cyclin D1 accumulation and cell cycle progression. In this study, we address the involvement of NFkappaB activation in the Ras-induced inhibition of myogenesis. Using H-Ras G12V and three G12V effector-loop variants, we detect high levels of NFkappaB transcriptional activity in C3H10T1/2-MyoD cells treated with differentiation medium. Myogenesis is blocked by all Ras proteins tested, yet only in the case of H-Ras G12V are cyclin D1 levels increased and cell cycle progression maintained. Expression of IkappaBalpha SR, an inhibitor of NFkappaB, does not reverse the differentiation-defective phenotype of Ras expressing cultures, but does induce differentiation in cultures treated with tumor necrosis factor (TNFalpha) or in cultures expressing the RelA/p65 subunit of NFkappaB. These data confirm that NFkappaB is a target of Ras and suggest that the cellular actions of NFkappaB require additional signals that are discriminated by the Ras effector-loop variants. Results with IkappaBalpha SR convincingly demonstrate that H-Ras G12V does not rely on NFkappaB activity to block myogenesis, an observation that continues to implicate another unidentified signaling pathway(s) in the inhibition of skeletal myogenesis by Ras.


Assuntos
Proteínas I-kappa B , Músculo Esquelético/citologia , NF-kappa B/metabolismo , Proteínas ras/metabolismo , Animais , Ciclo Celular , Diferenciação Celular , Células Cultivadas , Proteínas de Ligação a DNA/metabolismo , Camundongos , Fatores de Regulação Miogênica/metabolismo , Inibidor de NF-kappaB alfa , NF-kappa B/antagonistas & inibidores , Transdução de Sinais , Transcrição Gênica
6.
J Biol Chem ; 275(18): 13677-82, 2000 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-10788486

RESUMO

The 10T1/2-MRF4 fibroblast/myogenic cell system was used to address the following interrelated questions: whether distinct signaling pathways underlie myogenic inhibition by basic fibroblast growth factor (bFGF) and transforming growth factor (TGF)-beta; which of these pathways also up-regulates the fibroblast intermediate conductance calcium-activated potassium channel, FIK, a positive regulator of cell proliferation; and whether FIK up-regulation underlies some or all myogenic inhibitory signaling events. The results show that myogenic inhibition in 10T1/2-MRF4 cells, by both bFGF and TGF-beta, requires activation of the Ras/mitogen-activated protein (MAP) kinase/MAP kinase-ERK kinase (MEK)/extracellular signal-regulated kinase (ERK) pathway, and resultant FIK up-regulation. We show that FIK is instrumental in MEK-dependent suppression of acetylcholine receptor channel expression but that MEK activation and FIK up-regulation are not essential to suppression of myosin heavy chain and myotube formation. These data indicate that Ras/MEK/ERK induction of FIK is pivotal to regulation of certain myogenic events by both receptor tyrosine kinases and TGF-beta receptor, and this is also the first demonstration of chronic FIK up-regulation by the TGF-beta receptor family. Furthermore, the results define the physiologic signaling requirements for growth factor-stimulated FIK up-regulation, whereas previous work has concentrated on constitutive FIK up-regulation in cells stably transfected with oncoprotein signaling molecules. This study, together with earlier work showing that FIK positively regulates cell proliferation, establishes this member of the IK channel family as a multifunctional, growth factor-regulated signaling molecule.


Assuntos
Fator 2 de Crescimento de Fibroblastos/farmacologia , Fibroblastos/fisiologia , Músculo Esquelético/fisiologia , Canais de Potássio Cálcio-Ativados , Canais de Potássio/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Receptores Proteína Tirosina Quinases/fisiologia , Fator de Crescimento Transformador beta/farmacologia , Proteínas ras/fisiologia , Animais , Diferenciação Celular/fisiologia , Células Cultivadas , Canais de Potássio Ativados por Cálcio de Condutância Intermediária , Músculo Esquelético/citologia , Receptor EphA8 , Canais de Potássio Ativados por Cálcio de Condutância Baixa , Regulação para Cima/efeitos dos fármacos
7.
Anat Rec ; 259(2): 157-67, 2000 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-10820318

RESUMO

Mist1 is a basic helix-loop-helix transcription factor that represses E-box-mediated transcription. Previous studies have suggested that the Mist1 gene is expressed in a wide range of tissues, although a complete characterization of Mist1 protein accumulation in the adult organism has not been described. In an effort to identify specific cell types that contain the Mist1 protein, antibodies specific for Mist1 were generated and used in Western blot and immunohistochemical assays. Our studies show that the Mist1 protein is present in many different tissues but that it is restricted to cell types that are exclusively secretory in nature. Pancreatic acinar cells, serous or seromucous cells of the salivary glands, chief cells of the stomach, and secretory cells of the prostate and seminal vesicle show high levels of Mist1 protein, whereas nonserous exocrine cells, including the mucus-producing cells of the salivary glands, remain Mist1 negative. These results identify Mist1 as the first transcription factor that exhibits this unique serous-specific expression pattern and suggest that Mist1 may have a key role in establishing and maintaining a pathway responsible for the exocytosis of serous secretions.


Assuntos
Glândulas Exócrinas/metabolismo , Fatores de Transcrição/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Western Blotting , Celulas Principais Gástricas/metabolismo , Glândulas Exócrinas/citologia , Mucosa Gástrica/metabolismo , Expressão Gênica , Soros Imunes , Imuno-Histoquímica , Óperon Lac , Masculino , Camundongos , Camundongos Transgênicos , Pâncreas/citologia , Pâncreas/metabolismo , Próstata/citologia , Próstata/metabolismo , Glândulas Salivares/citologia , Glândulas Salivares/metabolismo , Glândulas Seminais/citologia , Glândulas Seminais/metabolismo , Membrana Serosa/metabolismo , Estômago/citologia , Fatores de Transcrição/metabolismo , beta-Galactosidase/metabolismo
8.
J Cell Sci ; 113 ( Pt 9): 1553-64, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10751147

RESUMO

Muscle LIM protein (MLP) is a striated muscle-specific factor that enhances myogenic differentiation and is critical to maintaining the structural integrity of the contractile apparatus. The ability of MLP to regulate myogenesis is particularly interesting since it exhibits multiple subcellular localizations, being found in both nuclear and cytoplasmic compartments. Despite extensive biochemical analyses on MLP, the mechanism(s) by which it influences the myogenic program remains largely undefined. To further examine the role of MLP as a positive myogenic regulator, a yeast two-hybrid screen was employed to identify cytoplasmic-associated MLP binding partners. From this screen, the cytoskeletal protein betaI-spectrin was isolated. Protein interaction assays demonstrate that MLP and betaI-spectrin associate with one another in vivo as well as when tested under several in vitro binding conditions. betaI-spectrin binds specifically to MLP but not to the MLP related proteins CRP1 and CRP2 or to other LIM domain containing proteins. The MLP:beta-spectrin interaction is mediated by the second LIM motif of MLP and by repeat 7 of beta-spectrin. Confocal microscopy studies also reveal that MLP co-localizes with beta-spectrin at the sarcolemma overlying the Z- and M-lines of myofibrils in both cardiac and skeletal muscle tissue. Given that beta-spectrin is a known costamere protein, we propose that sarcolemma-associated MLP also serves as a key costamere protein, stabilizing the association of the contractile apparatus with the sarcolemma by linking the beta-spectrin network to the alpha-actinin crosslinked actin filaments of the myofibril.


Assuntos
Proteínas do Citoesqueleto/metabolismo , Proteínas Musculares/metabolismo , Espectrina/metabolismo , Animais , Linhagem Celular , Humanos , Proteínas com Domínio LIM , Músculo Esquelético/metabolismo , Ligação Proteica , Ratos , Ratos Sprague-Dawley
9.
Cytogenet Cell Genet ; 86(3-4): 219-22, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-10575209

RESUMO

Mist1 is a basic helix-loop-helix (bHLH) transcription factor that is highly expressed in the adult pancreas. In this study, we isolated the mouse Mist1 gene and established its primary DNA sequence and complete genomic structure. Fluorescence in situ hybridization mapping located the Mist1 gene to the telomere of mouse chromosome 5 at position 5G2-5G3, an area which is syntenic to human chromosome 13q and which contains several additional pancreatic regulatory genes including IPF1 and CDX2.


Assuntos
Mapeamento Cromossômico , Camundongos/genética , Fatores de Transcrição/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Bandeamento Cromossômico , Cromossomos Humanos Par 13 , Clonagem Molecular , Sequências Hélice-Alça-Hélice , Humanos , Hibridização in Situ Fluorescente , Dados de Sequência Molecular , Pâncreas/metabolismo , Fatores de Transcrição/química
10.
J Biol Chem ; 273(28): 17696-701, 1998 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-9651367

RESUMO

Oncogenic Ras inhibits the differentiation of skeletal muscle cells through the activation of multiple downstream signaling pathways, including a Raf-dependent, mitogen-activated or extracellular signal-regulated kinase kinase/mitogen-activated protein kinase (MEK/MAPK)-independent pathway. Here we report that a non-Raf binding Ras effector-loop variant (H-Ras G12V,E37G), which retains interaction with the Ral guanine nucleotide dissociation stimulator (RalGDS), inhibits the conversion of MyoD-expressing C3H10T1/2 mouse fibroblasts to skeletal muscle. We show that H-Ras G12V,E37G, RalGDS, and the membrane-localized RalGDS CAAX protein inhibit the activity of alpha-actin-Luc, a muscle-specific reporter gene containing a necessary E-box and serum response factor (SRF) binding site, while a RalGDS protein defective for Ras interaction has no effect on alpha-actin-Luc transcription. H-Ras G12V,E37G does not activate endogenous MAPK, but does increase SRF-dependent transcription. Interestingly, RalGDS, RalGDS CAAX, and RalA G23V inhibit H-Ras G12V, E37G-induced expression of an SRF-regulated reporter gene, demonstrating that signaling through RalGDS does not duplicate the action of H-Ras G12V,E37G in this system. As additional evidence for this, we show that H-Ras G12V,E37G inhibits the expression of troponin I-Luc, an SRF-independent muscle-specific reporter gene, whereas RalGDS and RalGDS CAAX do not. Although our studies show that signaling through RalGDS can interfere with the expression of reporter genes dependent on SRF activity (including alpha-actin-Luc), our studies also provide strong evidence that an additional signaling molecule(s) activated by H-Ras G12V,E37G is required to achieve the complete inhibition of the myogenic differentiation program.


Assuntos
Proteínas de Ligação ao GTP/fisiologia , Desenvolvimento Muscular , Músculo Esquelético/crescimento & desenvolvimento , Proteínas Proto-Oncogênicas p21(ras)/fisiologia , Actinas/genética , Animais , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Diferenciação Celular , Linhagem Celular , Proteínas de Ligação a DNA/metabolismo , Genes Reporter , Luciferases/genética , Camundongos , Camundongos Endogâmicos C3H , Proteínas Nucleares/metabolismo , Fator de Resposta Sérica , Ativação Transcricional , Fator ral de Troca do Nucleotídeo Guanina , Proteínas rap de Ligação ao GTP
11.
EMBO J ; 17(5): 1412-22, 1998 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-9482738

RESUMO

A good model system to examine aspects of positive and negative transcriptional regulation is the muscle-specific regulatory factor, MyoD, which is a basic helix-loop-helix (bHLH) transcription factor. Although MyoD has the ability to induce skeletal muscle terminal differentiation in a variety of non-muscle cell types, MyoD activity itself is highly regulated through protein-protein interactions involving several different co-factors. Here we describe the characterization of a novel bHLH protein, Mist1, and how it influences MyoD function. We show that Mist1 accumulates in myogenic stem cells (myoblasts) and then decreases as myoblasts differentiate into myotubes. Mist1 functions as a negative regulator of MyoD activity, preventing muscle differentiation and the concomitant expression of muscle-specific genes. Mist1-induced inhibition occurs through a combination of mechanisms, including the formation of inactive MyoD-Mist1 heterodimers and occupancy of specific E-box target sites by Mist1 homodimers. Mist1 lacks a classic transcription activation domain and instead possesses an N-terminal repressor region capable of inhibiting heterologous activators. Thus, Mist1 may represent a new class of repressor molecules that play a role in controlling the transcriptional activity of MyoD, ensuring that expanding myoblast populations remain undifferentiated during early embryonic muscle formation.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Músculo Esquelético/citologia , Proteína MyoD/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo , Sequência de Aminoácidos , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos , Diferenciação Celular , DNA/metabolismo , Proteínas de Ligação a DNA/metabolismo , Dimerização , Fibroblastos/química , Sequências Hélice-Alça-Hélice , Proteína 1 Inibidora de Diferenciação , Camundongos , Dados de Sequência Molecular , Fibras Musculares Esqueléticas/química , Músculo Esquelético/química , Proteína MyoD/genética , Proteínas Recombinantes de Fusão , Proteínas Repressoras/análise , Proteínas Repressoras/genética , Fatores de Transcrição TCF , Proteína 1 Semelhante ao Fator 7 de Transcrição , Fatores de Transcrição/análise , Fatores de Transcrição/genética
12.
Mol Cell Biol ; 17(8): 4750-60, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9234731

RESUMO

The muscle LIM protein (MLP) is a muscle-specific LIM-only factor that exhibits a dual subcellular localization, being present in both the nucleus and in the cytoplasm. Overexpression of MLP in C2C12 myoblasts enhances skeletal myogenesis, whereas inhibition of MLP activity blocks terminal differentiation. Thus, MLP functions as a positive developmental regulator, although the mechanism through which MLP promotes terminal differentiation events remains unknown. While examining the distinct roles associated with the nuclear and cytoplasmic forms of MLP, we found that nuclear MLP functions through a physical interaction with the muscle basic helix-loop-helix (bHLH) transcription factors MyoD, MRF4, and myogenin. This interaction is highly specific since MLP does not associate with nonmuscle bHLH proteins E12 or E47 or with the myocyte enhancer factor-2 (MEF2) protein, which acts cooperatively with the myogenic bHLH proteins to promote myogenesis. The first LIM motif in MLP and the highly conserved bHLH region of MyoD are responsible for mediating the association between these muscle-specific factors. MLP also interacts with MyoD-E47 heterodimers, leading to an increase in the DNA-binding activity associated with this active bHLH complex. Although MLP lacks a functional transcription activation domain, we propose that it serves as a cofactor for the myogenic bHLH proteins by increasing their interaction with specific DNA regulatory elements. Thus, the functional complex of MLP-MyoD-E protein reveals a novel mechanism for both initiating and maintaining the myogenic program and suggests a global strategy for how LIM-only proteins may control a variety of developmental pathways.


Assuntos
Proteínas Musculares/metabolismo , Músculo Esquelético/citologia , Proteína MyoD/metabolismo , Fatores de Transcrição , Animais , Diferenciação Celular , Núcleo Celular/química , Citoplasma/química , Proteínas de Ligação a DNA/metabolismo , Dimerização , Sequências Hélice-Alça-Hélice , Proteínas com Domínio LIM , Camundongos , Desenvolvimento Muscular , Proteínas Musculares/análise , Músculo Esquelético/crescimento & desenvolvimento , Fatores de Regulação Miogênica/metabolismo , Miogenina/metabolismo , Ligação Proteica , Ratos , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Transcrição TCF , Proteína 1 Semelhante ao Fator 7 de Transcrição
13.
Mol Cell Biol ; 17(7): 3547-55, 1997 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-9199290

RESUMO

The ability of basic helix-loop-helix muscle regulatory factors (MRFs), such as MyoD, to convert nonmuscle cells to a myogenic lineage is regulated by numerous growth factor and oncoprotein signaling pathways. Previous studies have shown that H-Ras 12V inhibits differentiation to a skeletal muscle lineage by disrupting MRF function via a mechanism that is independent of the dimerization, DNA binding, and inherent transcriptional activation properties of the proteins. To investigate the intracellular signaling pathway(s) that mediates the inhibition of MRF-induced myogenesis by oncogenic Ras, we tested two transformation-defective H-Ras 12V effector domain variants for their ability to alter terminal differentiation. H-Ras 12V,35S retains the ability to activate the Raf/MEK/mitogen-activated protein (MAP) kinase cascade, whereas H-Ras 12V,40C is unable to interact directly with Raf-1 yet still influences other signaling intermediates, including Rac and Rho. Expression of each H-Ras 12V variant in C3H10T1/2 cells abrogates MyoD-induced activation of the complete myogenic program, suggesting that MAP kinase-dependent and -independent Ras signaling pathways individually block myogenesis in this model system. However, additional studies with constitutively activated Rac1 and RhoA proteins revealed no negative effects on MyoD-induced myogenesis. Similarly, treatment of Ras-inhibited myoblasts with the MEK1 inhibitor PD98059 revealed that elevated MAP kinase activity is not a significant contributor to the H-Ras 12V effect. These data suggest that an additional Ras pathway, distinct from the well-characterized MAP kinase and Rac/Rho pathways known to be important for the transforming function of activated Ras, is primarily responsible for the inhibition of myogenesis by H-Ras 12V.


Assuntos
Proteínas Quinases Dependentes de Cálcio-Calmodulina/fisiologia , Proteínas de Ligação ao GTP/fisiologia , Quinases de Proteína Quinase Ativadas por Mitógeno , Músculo Esquelético/citologia , Proteínas Proto-Oncogênicas p21(ras)/fisiologia , Animais , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Regulação da Expressão Gênica no Desenvolvimento , MAP Quinase Quinase 1 , Camundongos , Proteína MyoD/fisiologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/fisiologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas c-raf , Transdução de Sinais , Proteínas rac de Ligação ao GTP , Proteínas rho de Ligação ao GTP
14.
Dev Dyn ; 208(3): 299-312, 1997 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9056635

RESUMO

MRF4 is a muscle-specific transcription factor that belongs to a family of basic helix-loop-helix proteins known as the myogenic regulatory factors (MRFs). In vitro studies have shown that expression of the MRF4 gene is controlled by a proximal promoter element (-336 to +71) that binds the muscle-specific transcription factors MEF2 and myogenin to activate transcription. To examine further the regulatory elements necessary for endogenous MRF4 gene expression during development, transgenic mice were generated that contained either a proximal MRF4 promoter-LacZ reporter gene (-336 MRF4-nLacZ) or a MRF4-LacZ reporter gene containing 8.5 kb of 5' flanking sequence (-8500 MRF4-nLacZ). Characterization of individual transgenic mouse lines throughout development revealed that expression of both transgenes is restricted to skeletal muscle tissue. However, unlike previous in vitro data, the proximal promoter transgene exhibits only limited transcriptional activity at all developmental time points, whereas the -8500 MRF4-nLacZ lines fully recapitulate the later developmental expression patterns and exhibit transcription in myotomal cells during somitic differentiation. Tissue culture analysis of myogenic cells isolated from E12.5, E16.5, and adults confirmed that the -8500 MRF4-nLacZ transgene is expressed in greater than 90% of the myotubes for all myogenic populations. These results indicate that 8.5 kb of MRF4 5' flanking sequence contains all the regulatory elements necessary for late MRF4 expression and that at least some of these elements lie upstream of the -336 proximal promoter. It is also likely that distant upstream regulatory sequences control early somitic MRF4 expression. These findings, coupled with previous in vitro studies, suggest that the early and late developmental expression patterns of the MRF4 gene are controlled by distinct sets of regulatory elements.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Músculo Esquelético/embriologia , Músculo Esquelético/fisiologia , Fatores de Regulação Miogênica/genética , Animais , Células Cultivadas , Técnica Indireta de Fluorescência para Anticorpo , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Fatores de Regulação Miogênica/metabolismo , Miogenina , Somitos/metabolismo , Fatores de Tempo , Distribuição Tecidual , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
15.
Dev Biol ; 182(1): 101-13, 1997 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-9073453

RESUMO

Basic helix-loop-helix (bHLH) proteins often belong to a family of transcription factors that bind to the DNA target sequence -CANNTG- (E-box) that is present in the promoter or enhancer regions of numerous developmentally regulated genes. In this study, we report the isolation and initial characterization of a novel bHLH factor, termed Mist1, that was identified by virtue of its ability to interact with E-box regulatory elements in a yeast "one-hybrid" screening procedure. Northern analysis revealed that Mist1 transcripts are expressed in several adult tissues, including stomach, liver, lung, and spleen but no expression is detected in the heart, brain, kidney, or testis. During mouse embryogenesis, Mist1 mRNA is first observed at E10.5 in the primitive gut and in the developing lung bud. Expression persists through E16.5 and remains restricted primarily to the epithelial lining. Mist1 also is detected in skeletal muscle tissues beginning at E12.5, persisting throughout all embryonic stages examined although in older embryos and in the adult expression becomes severely reduced. At later developmental times, Mist1 transcripts also are found in the pancreas, submandibular gland, and adult spleen. As predicted, the Mist1 protein is nuclear and binds efficiently to E-box sites as a homodimer. Mist1 also is capable of binding to E-box elements when complexed as a heterodimer with the widely expressed E-proteins, E12 and E47. Surprisingly, although Mist1 binds to E-boxes in vivo, the Mist1 protein lacks a functional transcription activation domain. These observations suggest that Mist1 may function as a unique regulator of gene expression in several different embryonic and postnatal cell lineages.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/biossíntese , Desenvolvimento Embrionário e Fetal , Regulação da Expressão Gênica no Desenvolvimento , Sequências Hélice-Alça-Hélice , Fatores de Transcrição/biossíntese , Transcrição Gênica , Sequência de Aminoácidos , Animais , Sequência de Bases , Fatores de Transcrição Hélice-Alça-Hélice Básicos/química , Sítios de Ligação , Células COS , Linhagem Celular , Sequência Consenso , Elementos Facilitadores Genéticos , Humanos , Hibridização In Situ , Camundongos , Dados de Sequência Molecular , Especificidade de Órgãos , Regiões Promotoras Genéticas , RNA Mensageiro/biossíntese , Ratos , Proteínas Recombinantes/biossíntese , Homologia de Sequência de Aminoácidos , Fatores de Transcrição/química , Transfecção
16.
Mol Cell Biol ; 16(4): 1604-13, 1996 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8657135

RESUMO

The myogenic regulatory factors (MRFs) are a subclass of a much larger group of basic helix-loop-helix transcription factors which includes members of the E protein such as E47, E2-2, and HEB. Although the MRFs are unique in their ability to confer a myogenic phenotype on nonmuscle cells, they require E protein partners to form a MRF-E protein heterodimer, which represents the functional myogenesis-inducing complex. The mechanisms controlling homodimer and heterodimer formation in vivo remain largely unknown, although it is likely that posttranslational modification of one or both basic helix-loop-helix partners is critical to this regulatory event. In this respect, MyoD and MRF4, both members of the MRF family, exist in vivo as phosphoproteins and contains multiple consensus phosphorylation sites, including sites for casein kinase II (CKII) phosphorylation. In this study, we demonstrate that overexpression of CKII increases the transcriptional activities of MRF4 and MyoD in vivo. Interestingly, mutation of the individual CKII sites within MRF4 and MyoF does not alter the ability of CKII to enhance MRF transcriptional activity, suggesting that the effect of CKII expression on the MRFs is indirect. Given that the MRFs require dimerization with E protein partners to activate muscle-specific transcription, the effects of CKII expression on E protein function also were examined. Our studies show that E47 serves as an in vitro substrate for CKII and that CKII-phosphorylated E-47 proteins no longer bind to DNA. These observations were confirmed by in vivo experiments showing that overexpressing of CKII produces a dramatic reduction in E47 homodimer-directed transcription. We conclude from these studies that CKII may act as a positive regulator of myogenesis by preventing E protein homodimers from binding to muscle gene regulatory elements.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteína MyoD/metabolismo , Fatores de Regulação Miogênica/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica , Sequência de Aminoácidos , Animais , Caseína Quinase II , Linhagem Celular , Proteínas de Ligação a DNA/genética , Fibroblastos , Camundongos , Dados de Sequência Molecular , Mutação , Proteína MyoD/genética , Fatores de Regulação Miogênica/genética , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Transdução de Sinais , Fatores de Transcrição/genética
17.
DNA Cell Biol ; 15(1): 1-8, 1996 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8561893

RESUMO

Basic helix-loop-helix (bHLH) transcription factors play diverse roles in controlling many developmental events. Although a great deal is understood about how bHLH factors activate gene transcription via E-box DNA consensus sequences, studies of bHLH factor function in higher eukaryotes often have been hindered by the presence of multiple family members. As a first step in developing a simplified in vivo system to examine bHLH factor activities, we examined whether the bHLH muscle regulatory factors MRF4 and MyoD function appropriately in yeast. We show that Gal4-MRF4 fusion proteins, or native MRF4 proteins, activate expression of an E-box HIS3 reporter gene whereas MyoD proteins remain inactive. Deletion of the MRF4 transcription activation domain (TAD) or point mutations that abolish MRF4 DNA interactions inhibit HIS3 expression. Substitution of the MRF4 TAD with the Gal4 TAD also produces a functional protein, demonstrating that these transcription activation domains are functionally equivalent in yeast. Replacement of the MRF4 TAD with the related MyoD TAD, however, generates an inactive protein, suggesting that some specificity exists between bHLH family members. Using this experimental system, we also demonstrate that mammalian cDNA libraries can be screened successfully for cDNAs encoding novel bHLH proteins that interact with E-box targets. Thus, this in vivo yeast system provides a novel approach to facilitate functional studies of bHLH factor regulation.


Assuntos
Sequências Hélice-Alça-Hélice , Proteína MyoD/genética , Fatores de Regulação Miogênica/genética , Saccharomyces cerevisiae/genética , Fatores de Transcrição/genética , Animais , Deleção de Genes , Regulação da Expressão Gênica , Genes Reporter/genética , Mamíferos , Proteína MyoD/metabolismo , Fatores de Regulação Miogênica/metabolismo , Mutação Puntual , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Saccharomyces cerevisiae/metabolismo , Fatores de Transcrição/metabolismo
18.
FASEB J ; 9(15): 1595-604, 1995 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-8529839

RESUMO

Embryonic skeletal muscle development has become a paradigm for understanding the molecular basis of how cell lineages are established and how cells differentiate into specialized structures. Most vertebrate muscles are derived from individual somites that produce two distinct muscle populations: the myotomal muscles that generate the axial and trunk musculature and a second migratory cell population that colonizes regions of the developing limbs. In both instances, muscle differentiation is accompanied by cell cycle arrest, fusion of individual myoblasts into multinucleate myotubes, and the transcriptional activation of muscle-specific genes. Recent experimental progress has led to greater understanding of the molecular mechanisms that control myogenesis in the embryo. Most of the advances have come from the identification and isolation of regulatory genes that are involved in controlling specific transcriptional events. In particular, the muscle regulatory factor (MRF) and myocyte enhancer factor 2 (MEF2) families have been implicated in establishing the myogenic lineage as well as controlling terminal differentiation. Two additional transcription factors, Pax-3 and MLP, also appear to play a role in the production of a mature muscle cell. This review focuses on these four vertebrate transcription factor families and discusses the experimental evidence that these factors play important, non-overlapping roles in regulating skeletal muscle development.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Proteínas Musculares/fisiologia , Músculo Esquelético/embriologia , Fatores de Transcrição/classificação , Sequência de Aminoácidos , Animais , Proteínas de Ligação a DNA/fisiologia , Fatores de Transcrição MEF2 , Dados de Sequência Molecular , Família Multigênica , Músculo Esquelético/metabolismo , Fatores de Regulação Miogênica/fisiologia , Fator de Transcrição PAX3 , Fatores de Transcrição Box Pareados , Fatores de Transcrição/fisiologia
19.
Mol Cell Biol ; 15(10): 5205-13, 1995 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-7565669

RESUMO

MRF4, MyoD, myogenin, and Myf-5 are muscle-specific basic helix-loop-helix transcription factors that share the ability to activate the expression of skeletal muscle genes such as those encoding alpha-actin, myosin heavy chain, and the acetylcholine receptor subunits. The muscle regulatory factors (MRFs) also exhibit the unique capacity to initiate the myogenic program when ectopically expressed in a variety of nonmuscle cell types, most notably C3H10T1/2 fibroblasts (10T1/2 cells). The commitment of myoblasts to terminal differentiation, although positively regulated by the MRFs, also is controlled negatively by a variety of agents, including several growth factors and oncoproteins such as fibroblast growth factor (FGF-2), transforming growth factor beta 1 (TGF-beta 1), and Ras p21Val. The molecular mechanisms by which these varied agents alter myogenic terminal differentiation events remain unclear. In an effort to establish whether Ras p21Val represses MRF activity by directly targeting the MRF proteins, we examined the DNA binding and transcription activation potentials of MRF4 and MyoD when expressed in 10T1/2 cells or in 10T1/2 cells expressing Ras p21Val. Our results demonstrate that Ras p21Val inhibits terminal differentiation events by targeting the basic domain of the MRFs, and yet the mechanism underlying this inhibition does not involve altering the DNA binding or the inherent transcriptional activity of these regulatory factors. In contrast, FGF-2 and TGF-beta 1 block terminal differentiation by repressing the transcriptional activity of the MRFs. We conclude that the Ras p21Val block in differentiation operates via an intracellular signaling pathway that is distinct from the FGF-2 and TGF-beta 1 pathways.


Assuntos
Regulação da Expressão Gênica/fisiologia , Sequências Hélice-Alça-Hélice , Músculos/citologia , Fatores de Regulação Miogênica/fisiologia , Proteína Oncogênica p21(ras)/fisiologia , Diferenciação Celular , Linhagem Celular , DNA/metabolismo , Proteínas de Ligação a DNA/metabolismo , Fator 2 de Crescimento de Fibroblastos/farmacologia , Fibroblastos , Proteínas de Ligação ao GTP/fisiologia , Músculos/metabolismo , Proteína MyoD/metabolismo , Proteína MyoD/fisiologia , Fatores de Regulação Miogênica/metabolismo , Regiões Promotoras Genéticas/genética , Receptores Colinérgicos/genética , Proteínas Recombinantes de Fusão/biossíntese , Transdução de Sinais/genética , Fatores de Transcrição TCF , Proteína 1 Semelhante ao Fator 7 de Transcrição , Fatores de Transcrição/metabolismo , Fatores de Transcrição/fisiologia , Ativação Transcricional/efeitos dos fármacos , Fator de Crescimento Transformador beta/farmacologia , Proteínas rap de Ligação ao GTP
20.
Mol Cell Biol ; 15(5): 2707-18, 1995 May.
Artigo em Inglês | MEDLINE | ID: mdl-7739551

RESUMO

The basic helix-loop-helix muscle regulatory factor (MRF) gene family encodes four distinct muscle-specific transcription factors known as MyoD, myogenin, Myf-5, and MRF4. These proteins represent key regulatory factors that control many aspects of skeletal myogenesis. Although the MRFs often exhibit overlapping functional activities, their distinct expression patterns during embryogenesis suggest that each protein plays a unique role in controlling aspects of muscle development. As a first step in determining how MRF4 gene expression is developmentally regulated, we examined the ability of the MRF4 gene to be expressed in a muscle-specific fashion in vitro. Our studies show that the proximal MRF4 promoter contains sufficient information to direct muscle-specific expression. Located within the proximal promoter are a single MEF2 site and E box that are required for maximum MRF4 expression. Mutation of the MEF2 site or E box severely impairs the ability of this promoter to produce a muscle-specific response. In addition, the MEF2 site and E box function in concert to synergistically activate the MRF4 gene in nonmuscle cells coexpressing MEF2 and myogenin proteins. Thus, the MRF4 promoter is regulated by the MEF2 and basic helix-loop-helix MRF protein family through a cross-regulatory circuitry. Surprisingly, the MRF4 promoter itself is not transactivated by MRF4, suggesting that this MRF gene is not subject to an autoregulatory pathway as previously implied by other studies. Understanding the molecular mechanisms regulating expression of each MRF gene is central to fully understanding how these factors control developmental events.


Assuntos
Proteínas de Ligação a DNA/genética , Desenvolvimento Muscular , Músculos/metabolismo , Fatores de Regulação Miogênica/genética , Miogenina/genética , Fatores de Transcrição/genética , Animais , Sequência de Bases , Sítios de Ligação/genética , Linhagem Celular , Embrião de Galinha , Clonagem Molecular , DNA/genética , Proteínas de Ligação a DNA/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Óperon Lac , Fatores de Transcrição MEF2 , Camundongos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Miogenina/metabolismo , Regiões Promotoras Genéticas , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA