Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cells ; 12(21)2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37947627

RESUMEN

Cyclophilin E (CypE) belongs to the cyclophilin family and exhibits peptidyl-prolyl cis-trans isomerase (PPIase) activity. It participates in various biological processes through the regulation of peptidyl-prolyl isomerization. However, the specific role of CypE in osteoblast differentiation has not yet been elucidated. In this study, we first discovered the positive impact of CypE on osteoblast differentiation through gain or loss of function experiments. Mechanistically, CypE enhances the transcriptional activity of Runx2 through its PPIase activity. Furthermore, we identified the involvement of the Akt signaling pathway in CypE's function in osteoblast differentiation. Taken together, our findings indicate that CypE plays an important role in osteoblast differentiation as a positive regulator by increasing the transcriptional activity of Runx2.


Asunto(s)
Ciclofilinas , Osteoblastos , Ciclofilinas/genética , Osteoblastos/metabolismo
2.
Int J Mol Sci ; 23(16)2022 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-36012517

RESUMEN

Cyclophilin A (CypA) is a ubiquitously expressed and highly conserved protein with peptidyl-prolyl cis-trans isomerase activity that is involved in various biological activities by regulating protein folding and trafficking. Although CypA has been reported to positively regulate osteoblast differentiation, the mechanistic details remain largely unknown. In this study, we aimed to elucidate the mechanism of CypA-mediated regulation of osteoblast differentiation. Overexpression of CypA promoted osteoblast differentiation in bone morphogenic protein 4 (BMP4)-treated C2C12 cells, while knockdown of CypA inhibited osteoblast differentiation in BMP4-treated C2C12. CypA and Runx2 were shown to interact based on immunoprecipitation experiments and CypA increased Runx2 transcriptional activity in a dose-dependent manner. Our results indicate that this may be because CypA can increase the DNA binding affinity of Runx2 to Runx2 binding sites such as osteoblast-specific cis-acting element 2. Furthermore, to identify factors upstream of CypA in the regulation of osteoblast differentiation, various kinase inhibitors known to affect osteoblast differentiation were applied during osteogenesis. Akt inhibition resulted in the most significant suppression of osteogenesis in BMP4-induced C2C12 cells overexpressing CypA. Taken together, our results show that CypA positively regulates osteoblast differentiation by increasing the DNA binding affinity of Runx2, and Akt signaling is upstream of CypA.


Asunto(s)
Ciclofilina A , Osteogénesis , Diferenciación Celular , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Ciclofilina A/genética , Ciclofilina A/metabolismo , ADN/metabolismo , Osteoblastos/metabolismo , Osteogénesis/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo
3.
J Cell Physiol ; 237(9): 3554-3564, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35696549

RESUMEN

Nonalcoholic steatohepatitis (NASH) is a liver disease characterized by fat accumulation and chronic inflammation in the liver. Dynein light chain of 8 kDa (LC8) was identified previously as an inhibitor of nuclear factor kappa B (NF-κB), a key regulator of inflammation, however, its role in NASH remains unknown. In this study, we investigated whether LC8 can alleviate NASH using a mouse model of methionine and choline-deficient (MCD) diet-induced NASH and examined the underlying mechanism. LC8 transgenic (Tg) mice showed lower hepatic steatosis and less progression of NASH, including hepatic inflammation and fibrosis, compared to wild-type (WT) mice after consuming an MCD diet. The hepatic expression of lipogenic genes was lower, while that of lipolytic genes was greater in LC8 Tg mice than WT mice, which might be associated with resistance of LC8 Tg mice to hepatic steatosis. Consumption of an MCD diet caused oxidative stress, IκBα phosphorylation, and subsequent p65 liberation from IκBα and nuclear translocation, resulting in induction of proinflammatory cytokines and chemokines. However, these effects of MCD diet were reduced by LC8 overexpression. Collectively, these results suggest that LC8 alleviates MCD diet-induced NASH by inhibiting NF-κB through binding to IκBα to interfere with IκBα phosphorylation and by reducing oxidative stress via scavenging reactive oxygen species. Thus, boosting intracellular LC8 could be a potential therapeutic strategy for patients with NASH.


Asunto(s)
Dineínas , FN-kappa B , Enfermedad del Hígado Graso no Alcohólico , Estrés Oxidativo , Animales , Colina/metabolismo , Dineínas Citoplasmáticas , Dieta , Modelos Animales de Enfermedad , Dineínas/genética , Dineínas/metabolismo , Inflamación/metabolismo , Hígado/metabolismo , Hígado/patología , Metionina/metabolismo , Ratones , Ratones Endogámicos C57BL , Inhibidor NF-kappaB alfa/metabolismo , FN-kappa B/metabolismo , Enfermedad del Hígado Graso no Alcohólico/genética
4.
Proc Natl Acad Sci U S A ; 117(16): 8900-8911, 2020 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-32253314

RESUMEN

Signaling pathways that sense amino acid abundance are integral to tissue homeostasis and cellular defense. Our laboratory has previously shown that halofuginone (HF) inhibits the prolyl-tRNA synthetase catalytic activity of glutamyl-prolyl-tRNA synthetase (EPRS), thereby activating the amino acid response (AAR). We now show that HF treatment selectively inhibits inflammatory responses in diverse cell types and that these therapeutic benefits occur in cells that lack GCN2, the signature effector of the AAR. Depletion of arginine, histidine, or lysine from cultured fibroblast-like synoviocytes recapitulates key aspects of HF treatment, without utilizing GCN2 or mammalian target of rapamycin complex 1 pathway signaling. Like HF, the threonyl-tRNA synthetase inhibitor borrelidin suppresses the induction of tissue remodeling and inflammatory mediators in cytokine-stimulated fibroblast-like synoviocytes without GCN2, but both aminoacyl-tRNA synthetase (aaRS) inhibitors are sensitive to the removal of GCN1. GCN1, an upstream component of the AAR pathway, binds to ribosomes and is required for GCN2 activation. These observations indicate that aaRS inhibitors, like HF, can modulate inflammatory response without the AAR/GCN2 signaling cassette, and that GCN1 has a role that is distinct from its activation of GCN2. We propose that GCN1 participates in a previously unrecognized amino acid sensor pathway that branches from the canonical AAR.


Asunto(s)
Aminoacil-ARNt Sintetasas/antagonistas & inhibidores , Antiinflamatorios/farmacología , Artritis Reumatoide/tratamiento farmacológico , Piperidinas/farmacología , Quinazolinonas/farmacología , Transducción de Señal/efectos de los fármacos , Aminoácidos/metabolismo , Aminoacil-ARNt Sintetasas/metabolismo , Animales , Antiinflamatorios/uso terapéutico , Artritis Reumatoide/inmunología , Artritis Reumatoide/patología , Artritis Reumatoide/cirugía , Línea Celular , Fibroblastos , Técnicas de Silenciamiento del Gen , Células Endoteliales de la Vena Umbilical Humana , Humanos , Pulmón/citología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Ratones Noqueados , Piperidinas/uso terapéutico , Cultivo Primario de Células , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Quinazolinonas/uso terapéutico , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , RNA-Seq , Transducción de Señal/inmunología , Membrana Sinovial/citología , Membrana Sinovial/patología , Sinoviocitos , Transactivadores/genética , Transactivadores/metabolismo
5.
Biochem J ; 477(1): 121-136, 2020 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-31845979

RESUMEN

Vlk is a secreted tyrosine kinase that plays crucial roles during vertebrate embryonic development including skeletal formation. Genetic studies suggest that Vlk can modulate the Hedgehog signaling pathway during skeletal development. Despite its potential roles as an extracellular regulator of signaling pathways, little is known regarding the molecular functions of Vlk. Here we show that Vlk can negatively regulate the Hedgehog signaling pathway. We found that Vlk can induce lysosomal degradation of Smoothened, a crucial transmembrane signal transducer of the Hedgehog pathway, through the interaction with the extracellular domain of Smoothened (Smo-ECD). In addition, we observed that Vlk can attenuate Hedgehog signaling-induced ciliary localization of Smoothened. Furthermore, Vlk-mediated suppression of Hedgehog signaling can be diminished by tyrosine-to-phenylalanine substitutions in Smo-ECD. Taken together, these results suggest that Vlk may function as a signaling regulator in extracellular space to modulate the Hedgehog pathway.


Asunto(s)
Proteínas Hedgehog/metabolismo , Proteínas Tirosina Quinasas/fisiología , Proteolisis , Receptor Smoothened/metabolismo , Animales , Cilios/metabolismo , Células HEK293 , Humanos , Lisosomas/metabolismo , Ratones , Células 3T3 NIH
6.
Nat Commun ; 9(1): 3434, 2018 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-30143647

RESUMEN

Secreted Wnts play crucial roles in synaptogenesis and synapse maintenance, but endogenous factors promoting synapse elimination in central neurons remain unknown. Here we show that proline-rich 7 (PRR7) induces specific removal of excitatory synapses and acts as a Wnt inhibitor. Remarkably, transmembrane protein PRR7 is activity-dependently released by neurons via exosomes. Exosomal PRR7 is uptaken by neurons through membrane fusion and eliminates excitatory synapses in neighboring neurons. Conversely, PRR7 knockdown in sparse neurons greatly increases excitatory synapse numbers in all surrounding neurons. These non-cell autonomous effects of PRR7 are effectively negated by augmentation or blockade of Wnt signaling. PRR7 exerts its effect by blocking the exosomal secretion of Wnts, activation of GSK3ß, and promoting proteasomal degradation of PSD proteins. These data uncover a proximity-dependent, reciprocal mechanism for the regulation of excitatory synapse numbers in local neurons and demonstrate the significance of exosomes in inter-neuronal signaling in the vertebrate brain.


Asunto(s)
Exosomas/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Sinapsis/metabolismo , Proteínas Wnt/metabolismo , Animales , Células Cultivadas , Femenino , Células HEK293 , Hipocampo/metabolismo , Humanos , Inmunohistoquímica , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Neurogénesis/genética , Neurogénesis/fisiología , Neuronas/metabolismo , Ratas , Transducción de Señal/genética , Transducción de Señal/fisiología
7.
Sci Rep ; 7(1): 13278, 2017 10 16.
Artículo en Inglés | MEDLINE | ID: mdl-29038508

RESUMEN

Wnt signaling controls critical developmental processes including tissue/body patterning. Here we report the identification of a novel regulator of Wnt signaling, OTTOGI (OTG), isolated from a large-scale expression screening of human cDNAs in zebrafish embryos. Overexpression of OTG in zebrafish embryos caused dorso-anteriorized phenotype, inhibited the expression of Wnt target genes, and prevented nuclear accumulation of ß-catenin. Conversely, knockdown of zebrafish otg using specific antisense morpholino promoted nuclear accumulation of ß-catenin and caused ventralization. However, OTG failed to rescue headless-like phenotype induced by inhibition of GSK-3ß activity, suggesting that OTG acts upstream of GSK-3ß. OTG bound specifically to Frizzled8 (Fz8) receptor and caused retention of Fz8 in the endoplasmic reticulum possibly by preventing N-linked glycosylation of Fz8. Taken together, our data indicate that OTG functions as a novel negative regulator of Wnt signaling during development by the modulation of cell surface expression of Fz receptor.


Asunto(s)
Membrana Celular/metabolismo , Receptores de Superficie Celular/metabolismo , Vía de Señalización Wnt , Proteínas de Pez Cebra/metabolismo , Animales , ADN Complementario/genética , Desarrollo Embrionario/genética , Retículo Endoplásmico/metabolismo , Expresión Génica , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Glicosilación , Humanos , Fenotipo , Unión Proteica , Transporte de Proteínas , Transcriptoma , Proteínas de Pez Cebra/genética
8.
Mol Cell Endocrinol ; 436: 150-8, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27475846

RESUMEN

Pin1 is a peptidylprolyl cis/trans isomerase and it has a unique enzymatic activity of catalyzing isomerization of the peptide bond between phospho-serine/threonine and proline. Through the conformational change of its substrates, Pin1 regulates diverse biological processes including adipogenesis. In mouse embryonic fibroblasts and 3T3-L1 preadipocytes, overexpression of Pin1 enhances adipocyte differentiation whereas inhibition of Pin1 activity suppresses it. However, the precise functions of Pin1 during adipogenesis are not clear. In the present study, we investigated the potential targets of Pin1 during adipogenesis. We found that Pin1 interacts directly with and regulates the transcriptional activity of PPARγ, a key regulator of adipogenesis. In addition, ERK activity and Ser273 of PPARγ, a potential ERK phosphorylation target site, are important for the regulation of PPARγ function by Pin1 in 3T3-L1 cells. Taken together our results suggest a novel regulatory mechanism of Pin1 during adipogenesis, in which Pin1 enhances adipocyte differentiation by regulating the function of PPARγ.


Asunto(s)
Adipocitos/citología , Adipocitos/metabolismo , Diferenciación Celular , Peptidilprolil Isomerasa de Interacción con NIMA/metabolismo , PPAR gamma/metabolismo , Transcripción Genética , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Animales , Butadienos/farmacología , Diferenciación Celular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Nitrilos/farmacología , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Unión Proteica/genética , Serina/metabolismo , Transcripción Genética/efectos de los fármacos
9.
Mol Cell Endocrinol ; 407: 85-97, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25802190

RESUMEN

Osterix, a zinc-finger transcription factor, is required for osteoblast differentiation and new bone formation during embryonic development. The c-Src of tyrosine kinase is involved in a variety of cellular signaling pathways, leading to the induction of DNA synthesis, cell proliferation, and cytoskeletal reorganization. Src activity is tightly regulated and its dysregulation leads to constitutive activation and cellular transformation. The function of Osterix can be also modulated by post-translational modification. But the precise molecular signaling mechanisms between Osterix and c-Src are not known. In this study we investigated the potential regulation of Osterix function by c-Src in osteoblast differentiation. We found that c-Src activation increases protein stability, osteogenic activity and transcriptional activity of Osterix. The siRNA-mediated knockdown of c-Src decreased the protein levels and transcriptional activity of Osterix. Conversely, Src specific inhibitor, SU6656, decreased the protein levels and transcriptional activity of Osterix. The c-Src interacts with and phosphorylates Osterix. These results suggest that c-Src signaling modulates osteoblast differentiation at least in part through Osterix.


Asunto(s)
Células Epiteliales/metabolismo , Mioblastos/metabolismo , Osteoblastos/metabolismo , Factores de Transcripción/genética , Familia-src Quinasas/genética , Animales , Proteína Tirosina Quinasa CSK , Diferenciación Celular , Línea Celular , Proliferación Celular , Citoesqueleto/metabolismo , Citoesqueleto/ultraestructura , ADN/biosíntesis , ADN/genética , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Regulación de la Expresión Génica , Genes Reporteros , Células HEK293 , Humanos , Indoles/farmacología , Luciferasas/genética , Luciferasas/metabolismo , Ratones , Mioblastos/citología , Mioblastos/efectos de los fármacos , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteogénesis/genética , Inhibidores de Proteínas Quinasas/farmacología , Estabilidad Proteica , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Transducción de Señal , Factor de Transcripción Sp7 , Sulfonamidas/farmacología , Factores de Transcripción/metabolismo , Familia-src Quinasas/antagonistas & inhibidores , Familia-src Quinasas/metabolismo
10.
Bone ; 75: 201-9, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25744063

RESUMEN

E3 ubiquitin ligase Cbl-b and c-Cbl play important roles in bone formation and maintenance. Cbl-b and c-Cbl regulate the activity of various receptor tyrosine kinases and intracellular protein tyrosine kinases mainly by regulating the degradation of target proteins. However, the precise mechanisms of how Cbl-b and c-Cbl regulate osteoblast differentiation are not well known. In this study, we investigated potential targets of Cbl-b and c-Cbl. We found that Cbl-b and c-Cbl inhibit BMP2-induced osteoblast differentiation in mesenchymal cells. Among various osteogenic transcription factors, we identified that Cbl-b and c-Cbl suppress the protein stability and transcriptional activity of Osterix. Our results suggest that Cbl-b and c-Cbl inhibit the function of Osterix by enhancing the ubiquitin-proteasome-mediated degradation of Osterix. Taken together, we propose novel regulatory roles of Cbl-b and c-Cbl during osteoblast differentiation in which Cbl-b and c-Cbl regulate the degradation of Osterix through the ubiquitin-proteasome pathway.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Diferenciación Celular/fisiología , Osteoblastos/citología , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Factores de Transcripción/metabolismo , Animales , Línea Celular , Humanos , Immunoblotting , Inmunoprecipitación , Ratones , Osteogénesis/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción Sp7 , Transfección , Ubiquitinación
11.
Protist ; 166(1): 1-13, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25555149

RESUMEN

During the differentiation of the amoeba Naegleria pringsheimi into a flagellate, a transient complex containing γ-tubulin, pericentrin-like protein, and myosin II (GPM complex) is formed, and subsequently a pair of basal bodies is assembled from the complex. It is not understood, however, how a single GPM is formed nor how the capability to form this complex is acquired by individual cells. We hypothesized that the GPM is formed from a precursor complex and developed an antibody that recognizes Naegleria (Ng)-transacylase, a component of the precursor complex. Immunostaining of differentiating cells showed that Ng-transacylase is concentrated at a site in the amoeba and that γ-tubulin is transiently co-concentrated at the site, suggesting that the GPM is formed from a precursor, GPMp, which contains Ng-transacylase and is already present in the amoeba. Immunostaining of growing N. pringsheimi with Ng-transacylase antibody revealed the presence of one GPMp in interphase cells, but two GPMps in mitotic cells, suggesting that N. pringsheimi maintains one GPMp per cell by duplicating and segregating the complex according to its cell cycle. Our results demonstrate the existence of a cell cycle-dependent duplicating complex that provides a site for the de novo assembly of the next generation of basal bodies.


Asunto(s)
Cuerpos Basales/metabolismo , Naegleria/citología , Naegleria/fisiología , Antígenos/metabolismo , Ciclo Celular , Diferenciación Celular , Miosina Tipo II/metabolismo , Multimerización de Proteína , Tubulina (Proteína)/metabolismo
12.
Dev Biol ; 397(1): 129-39, 2015 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-25446028

RESUMEN

Members of the fibroblast growth factor (FGF) family play important roles during various developmental processes including eye development. FRS (FGF receptor substrate) proteins bind to FGFR and serve as adapters for coordinated assembly of multi-protein complexes involved in Ras/MAPK and PI3 kinase/Akt pathways. Here, we identified Xenopus laevis Frs3 (XFrs3), a homolog of vertebrate Frs3, and investigated its roles during embryogenesis. XFrs3 is expressed maternally and zygotically with specific expression patterns throughout the early development. Knockdown of XFrs3 using a specific antisense morpholino oligonucleotide (MO) caused reduction of Pax6 expression in the lens placode, and defects in the eye ranging from microphthalmia to anophthalmia. XFrs3 MO-induced defects were alleviated by wild type XFrs3 or a mutant XFrs3 (XFrs3-4YF), in which the putative tyrosine phosphorylation sites served as Grb2-binding sites are mutated. However, another XFrs3 mutant (XFrs3-2YF), in which the putative Shp2-binding sites are mutated, could not rescue the defects of XFrs3 morphants. In addition, we found that XFrs3 is important for FGF or IGF-induced ERK activation in ectodermal tissue. Taken together, our results suggest that signaling through Shp2-binding sites of XFrs3 is necessary for the eye development in Xenopus laevis.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas del Ojo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/metabolismo , Cristalino/embriología , Factores de Transcripción Paired Box/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteínas Represoras/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriología , Animales , Sitios de Unión , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Mutación , Oligonucleótidos/química , Factor de Transcripción PAX6 , Fosforilación , Unión Proteica , Transducción de Señal , Tirosina/química
13.
Cell ; 158(5): 1033-1044, 2014 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-25171405

RESUMEN

Although tyrosine phosphorylation of extracellular proteins has been reported to occur extensively in vivo, no secreted protein tyrosine kinase has been identified. As a result, investigation of the potential role of extracellular tyrosine phosphorylation in physiological and pathological tissue regulation has not been possible. Here, we show that VLK, a putative protein kinase previously shown to be essential in embryonic development, is a secreted protein kinase, with preference for tyrosine, that phosphorylates a broad range of secreted and ER-resident substrate proteins. We find that VLK is rapidly and quantitatively secreted from platelets in response to stimuli and can tyrosine phosphorylate coreleased proteins utilizing endogenous as well as exogenous ATP sources. We propose that discovery of VLK activity provides an explanation for the extensive and conserved pattern of extracellular tyrosine phosphophorylation seen in vivo, and extends the importance of regulated tyrosine phosphorylation into the extracellular environment.


Asunto(s)
Plaquetas/enzimología , Embrión de Mamíferos/enzimología , Proteínas Quinasas/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Secuencia de Aminoácidos , Animales , Desarrollo Embrionario , Glicosilación , Humanos , Ratones , Datos de Secuencia Molecular , Fosforilación , Proteínas Quinasas/química , Proteínas Quinasas/genética , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína , Proteínas Tirosina Quinasas/química , Vías Secretoras
14.
J Cell Biochem ; 115(11): 2004-11, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24924519

RESUMEN

Protein kinase A (PKA), a serine/threonine kinase, regulates bone formation, and enhances Bone morphogenetic protein (BMP)-induced osteoblast differentiation. However, the mechanisms of how PKA controls the cellular response to BMP are not well known. We investigated the effects of modulating PKA activity during BMP2-induced osteoblast differentiation, and found that PKA regulates the function of Dlx3. Dlx3 plays crucial roles in osteoblast differentiation and it is expressed in most skeletal elements during development. We found that PKA activation increases BMP2-induced expression of Dlx3 protein, and enhances the protein stability, DNA binding, and transcriptional activity of Dlx3. In addition, PKA activation induces the phosphorylation of Dlx3 at consensus PKA phosphorylation target site(s). Lastly, substitution of serine 10 in Dlx3 to alanine significantly reduces, if not completely abolishes, the phosphorylation of Dlx3 and the regulation of Dlx3 function by PKA. These results suggest that Dlx3 is a novel target of PKA, and that PKA mediates BMP signaling during osteoblast differentiation, at least in part, by phosphorylating Dlx3 and modulating the protein stability and function of Dlx3.


Asunto(s)
Proteína Morfogenética Ósea 2/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Osteoblastos/fisiología , Serina/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Sustitución de Aminoácidos , Animales , Proteína Morfogenética Ósea 2/farmacología , Diferenciación Celular , Línea Celular , Colforsina/farmacología , Células HEK293 , Proteínas de Homeodominio/química , Humanos , Isoquinolinas/farmacología , Ratones , Fosforilación , Estabilidad Proteica , Sulfonamidas/farmacología , Factores de Transcripción/química
15.
J Cell Biochem ; 115(10): 1808-15, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24905700

RESUMEN

Osterix belongs to the SP gene family and is a core transcription factor responsible for osteoblast differentiation and bone formation. Activation of protein kinase A (PKA), a serine/threonine kinase, is essential for controlling bone formation and BMP-induced osteoblast differentiation. However, the relationship between Osterix and PKA is still unclear. In this report, we investigated the precise role of the PKA pathway in regulating Osterix during osteoblast differentiation. We found that PKA increased the protein level of Osterix; PKA phosphorylated Osterix, increased protein stability, and enhanced the transcriptional activity of Osterix. These results suggest that Osterix is a novel target of PKA, and PKA modulates osteoblast differentiation partially through the regulation of Osterix.


Asunto(s)
Remodelación Ósea/fisiología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Osteoblastos/citología , Osteogénesis/fisiología , Factores de Transcripción/metabolismo , Animales , Diferenciación Celular , Línea Celular , Células HEK293 , Humanos , Ratones , Fosforilación , Factor de Transcripción Sp7 , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Transcripción Genética , Activación Transcripcional
16.
FEBS J ; 281(16): 3656-66, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24961731

RESUMEN

Runx2 plays essential roles in bone formation and chondrocyte maturation. Akt promotes osteoblast differentiation induced by the bone morphogenetic proteins BMP2 and enhances the function and transcriptional activity of Runx2. However, the precise molecular mechanism underlying the relationship between Runx2 and Akt is not well understood. In this study, we examined the role of Akt in regulating Runx2 function. We found that Akt increases the stability of Runx2 protein. However, the level of Runx2 mRNA was not affected by Akt, and we did not find any evidence for direct modification of Runx2 by Akt. Instead, we found evidence that Akt induces the phosphorylation of the Smad ubiquitination regulatory factor Smurf2 and decreases the level of Smurf2 protein through ubiquitin/proteasome-mediated degradation of Smurf2. Akt also alleviates Smurf2-mediated suppression of Runx2 transcriptional activity. Taken together, our results suggest that Akt regulates osteoblast differentiation, at least in part, by enhancing the protein stability and transcriptional activity of Runx2 through regulation of ubiquitin/proteasome-mediated degradation of Smurf2.


Asunto(s)
Diferenciación Celular , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Osteoblastos/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Ubiquitina-Proteína Ligasas/fisiología , Animales , Células HEK293 , Humanos , Ratones , Fosforilación , Unión Proteica , Mapas de Interacción de Proteínas , Estabilidad Proteica , Proteolisis , Transcripción Genética , Ubiquitinación
17.
Biochim Biophys Acta ; 1843(9): 2005-11, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24803334

RESUMEN

Transcription factor YY1 plays important roles in cell proliferation and differentiation. For example, YY1 represses the expression of muscle-specific genes and the degradation of YY1 is required for myocyte differentiation. The activity of YY1 can be regulated by various post-translational modifications; however, little is known about the regulatory mechanisms for YY1 degradation. In this report, we attempted to identify potential E3 ubiquitin ligases for YY1, and found that Smurf2 E3 ubiquitin ligase can negatively regulate YY1 protein level, but not mRNA level. Smurf2 interacted with YY1, induced the poly-ubiquitination of YY1 and shortened the half-life of YY1 protein. Conversely, an E3 ubiquitin ligase-defective mutant form of Smurf2 or knockdown of Smurf2 increased YY1 protein level. PPxY motif is a typical target recognition site for Smurf2, and the PPxY motif in YY1 was important for Smurf2 interaction and Smurf2-induced degradation of YY1 protein. In addition, Smurf2 reduced the YY1-mediated activation of a YY1-responsive reporter whereas Smurf2 knockdown increased it. Finally, Smurf2 relieved the suppression of p53 activity by YY1. Taken together, our results suggest a novel regulatory mechanism for YY1 function by Smurf2 in which the protein stability and transcriptional activity of YY1 are regulated by Smurf2 through the ubiquitin-proteasome-mediated degradation of YY1.


Asunto(s)
Proteolisis , Ubiquitina-Proteína Ligasas/metabolismo , Factor de Transcripción YY1/metabolismo , Secuencias de Aminoácidos , Regulación hacia Abajo , Células HEK293 , Humanos , Poliubiquitina/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Proteína p53 Supresora de Tumor/metabolismo , Ubiquitinación , Factor de Transcripción YY1/química
18.
FEBS Lett ; 587(22): 3640-7, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-24113655

RESUMEN

Peptidyl-prolyl isomerase 1 (Pin1) is the only enzyme known to catalyze isomerization of the pSer/Thr-Pro peptide bond. Pin1 induces conformational change of substrates and subsequently regulates diverse cellular processes. However, its role in osteoblast differentiation is not well understood. Here we show that Pin1 enhances osteoblast differentiation. Pin1 interacts and affects the protein stability and transcriptional activity of an important osteogenic transcriptional factor Runx2. Our results indicate that this regulation is likely due to suppression of poly-ubiquitination-mediated proteasomal degradation of Runx2. Our current finding suggests that Pin1 is a novel regulator of osteoblast differentiation that acts through the regulation of Runx2 function.


Asunto(s)
Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Osteoblastos/enzimología , Isomerasa de Peptidilprolil/fisiología , Animales , Diferenciación Celular , Subunidad alfa 1 del Factor de Unión al Sitio Principal/química , Regulación de la Expresión Génica , Células HEK293 , Humanos , Quinasas Quinasa Quinasa PAM/metabolismo , Ratones , Peptidilprolil Isomerasa de Interacción con NIMA , Osteoblastos/fisiología , Isomerasa de Peptidilprolil/química , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas , Estabilidad Proteica , Proteolisis , Transcripción Genética
19.
Am J Med Genet A ; 161A(8): 2040-6, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23824657

RESUMEN

The transforming growth factor ß (TGF-ß) family of growth factors are key regulators of mammalian development and their dysregulation is implicated in human disease, notably, heritable vasculopathies including Marfan (MFS, OMIM #154700) and Loeys-Dietz syndromes (LDS, OMIM #609192). We described a syndrome presenting at birth with distal arthrogryposis, hypotonia, bifid uvula, a failure of normal post-natal muscle development but no evidence of vascular disease; some of these features overlap with MFS and LDS. A de novo mutation in TGFB3 was identified by exome sequencing. Several lines of evidence indicate the mutation is hypomorphic suggesting that decreased TGF-ß signaling from a loss of TGFB3 activity is likely responsible for the clinical phenotype. This is the first example of a mutation in the coding portion of TGFB3 implicated in a clinical syndrome suggesting TGFB3 is essential for both human palatogenesis and normal muscle growth.


Asunto(s)
Artrogriposis/genética , Trastornos del Crecimiento/genética , Síndrome de Loeys-Dietz/genética , Síndrome de Marfan/genética , Debilidad Muscular/genética , Mutación/genética , Factor de Crecimiento Transformador beta3/genética , Adulto , Animales , Artrogriposis/diagnóstico , Células Cultivadas , Niño , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Femenino , Trastornos del Crecimiento/diagnóstico , Humanos , Síndrome de Loeys-Dietz/diagnóstico , Masculino , Síndrome de Marfan/diagnóstico , Debilidad Muscular/diagnóstico , Fenotipo , Transducción de Señal , Factor de Crecimiento Transformador beta3/metabolismo , Xenopus laevis/metabolismo
20.
Biochim Biophys Acta ; 1823(8): 1225-32, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22633971

RESUMEN

Protein kinase C (PKC) signaling regulates osteoblast differentiation, but little is known about its downstream effectors. We examined the effect of modulating PKC activity on osteogenic transcription factors and found that the protein level of Msx2 is affected. Msx2 is induced by osteogenic signals such as BMPs and it plays critical roles in bone formation and osteoblast differentiation. Here, we examined the role of PKC signaling in regulating the function of Msx2. We found that the inhibition of PKC signaling enhances osteogenic differentiation in BMP2-stimulated C2C12 cells. Treatment with inhibitors of PKC activity or overexpression of kinase-defective (KD), dominant-negative mutant PKC isoforms strongly reduced the level of Msx2 protein. Several PKC isoforms (α, ß, δ, and ζ) interacted with Msx2, and PKCß phosphorylated Msx2 at Thr135 and Thr141. Msx2 repressed the transcriptional activity of the osteogenic transcription factor Runx2, and this repression was relieved by inhibition of PKC activity or overexpression of the KD mutant PKC isoforms. In addition, PKC prolonged the half-life of Msx2 protein. These results suggest that PKC signaling modulates osteoblast differentiation, at least in part, through the regulation of Msx2.


Asunto(s)
Diferenciación Celular , Proteínas de Homeodominio/metabolismo , Proteína Quinasa C/fisiología , Transducción de Señal , Fosfatasa Alcalina/metabolismo , Animales , Proteína Morfogenética Ósea 2/fisiología , Línea Celular , Proliferación Celular , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Expresión Génica , Semivida , Proteínas de Homeodominio/química , Proteínas de Homeodominio/genética , Humanos , Ratones , Osteogénesis , Fosforilación , Proteína Quinasa C/química , Proteína Quinasa C/metabolismo , Proteína Quinasa C beta , Inhibidores de Proteínas Quinasas/farmacología , Procesamiento Proteico-Postraduccional , Estabilidad Proteica , Activación Transcripcional , Ubiquitinación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...