Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Tohoku J Exp Med ; 252(3): 199-208, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33087680

RESUMEN

Ependymal cells have an essential role in regulating the dynamics of the cerebrospinal fluid flow by the movement of their multiple cilia. Impaired generation or function of cilia could cause hydrocephalus due to the disordered dynamics of the cerebrospinal fluid flow. However, molecular bases regulating differentiation of the ependymal cells and their ciliogenesis have not been fully elucidated. We report here that bone morphogenetic proteins (BMPs), growth factors orchestrating tissue architecture throughout the body, inhibit ciliogenesis during ependymal cell differentiation in primary cell culture. Previous in vitro study has reported that ectopic expression of Smad6 and Smad7 promotes differentiation of embryonic stem cells into multi-ciliated ependymal-like cells. Since Smad6 and Smad7 have been known as the intracellular inhibitory factors of the BMP signaling pathway, the activation of the pathway could cause a deficit in ciliogenesis of ependymal cells. To examine whether activation of the pathway affects ciliogenesis, we investigated the effects of two BMPs, BMP2 and BMP4, on the ependymal differentiation of the primary cultured cells prepared from the neonatal mouse brain. Supplementation of BMP2 or BMP4 in culture media significantly reduced the number of cells with multiple cilia among the total cells, while most of the cells expressed FoxJ1, a master regulator of ciliogenesis. Activation of the pathway was confirmed by the phosphorylation of intracellular Smad1/5/8, downstream factors of the BMP receptors. These in vitro results suggest that inhibition of the BMP signaling pathway might be essential for ciliogenesis during the ependymal cell differentiation in vivo.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Cilios/metabolismo , Epéndimo/citología , Animales , Proteína Morfogenética Ósea 2/biosíntesis , Proteína Morfogenética Ósea 4/biosíntesis , Encéfalo/metabolismo , Diferenciación Celular , Células Cultivadas , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Fosforilación , Transducción de Señal/efectos de los fármacos , Proteína smad6/biosíntesis , Proteína smad7/biosíntesis
2.
Biol Reprod ; 103(3): 572-582, 2020 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-32432313

RESUMEN

In this study, we aimed to determine the origin of the difference, in terms of anti-Müllerian hormone production, existing between the bovine and porcine ovaries. We first confirmed by quantitative real-time-Polymerase-Chain Reaction, ELISA assay and immunohistochemistry that anti-Müllerian hormone mRNA and protein production are very low in porcine ovarian growing follicles compared to bovine ones. We then have transfected porcine and bovine granulosa cells with vectors containing the luciferase gene driven by the porcine or the bovine anti-Müllerian hormone promoter. These transfection experiments showed that the porcine anti-Müllerian hormone promoter is less active and less responsive to bone morphogenetic protein stimulations than the bovine promoter in both porcine and bovine cells. Moreover, bovine but not porcine granulosa cells were responsive to bone morphogenetic protein stimulation after transfection of a plasmidic construction including a strong response element to the bone morphogenetic proteins (12 repetitions of the GCCG sequence) upstream of the luciferase reporter gene. We also showed that SMAD6, an inhibitor of the SMAD1-5-8 pathway, is strongly expressed in porcine compared to the bovine granulosa cells. Overall, these results suggest that the low expression of anti-Müllerian hormone in porcine growing follicles is due to both a lack of activity/sensitivity of the porcine anti-Müllerian hormone promoter, and to the lack of responsiveness of porcine granulosa cells to bone morphogenetic protein signaling, potentially due to an overexpression of SMAD6 compared to bovine granulosa cells. We propose that the low levels of anti-Müllerian hormone in the pig would explain the poly-ovulatory phenotype in this species.


Asunto(s)
Hormona Antimülleriana/biosíntesis , Células de la Granulosa/metabolismo , Ovario/metabolismo , Animales , Hormona Antimülleriana/genética , Proteínas Morfogenéticas Óseas/biosíntesis , Bovinos , Femenino , Regulación del Desarrollo de la Expresión Génica , Inmunohistoquímica , Ovario/citología , Regiones Promotoras Genéticas , Transducción de Señal/efectos de los fármacos , Proteína smad6/biosíntesis , Proteína smad6/genética , Especificidad de la Especie , Porcinos
3.
Tissue Eng Part A ; 22(9-10): 707-20, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27036931

RESUMEN

Current approaches for the treatment of skeletal defects are suboptimal, principally because the ability of bone to repair and regenerate is poor. Although the promise of effective cellular therapies for skeletal repair is encouraging, these approaches are limited by the risks of infection, cellular contamination, and tumorigenicity. Development of a pharmacological approach would therefore help avoid some of these potential risks. This study identifies transforming growth factor beta (TGFß) signaling as a potential pathway for pharmacological modulation in vivo. We demonstrate that inhibition of TGFß signaling by the small molecule SB431542 potentiates calvarial skeletal repair through activation of bone morphogenetic protein (BMP) signaling on osteoblasts and dura mater cells participating in healing of calvarial defects. Cells respond to inhibition of TGFß signaling by producing higher levels of BMP2 that upregulates inhibitory Smad6 expression, thus providing a negative feedback loop to contain excessive BMP signaling. Importantly, study on human osteoblasts indicates that molecular mechanism(s) triggered by SB431542 are conserved. Collectively, these data provide insights into the use of small molecules to modulate key signaling pathways for repairing skeletal defects.


Asunto(s)
Benzamidas/farmacología , Regeneración Ósea/efectos de los fármacos , Dioxoles/farmacología , Osteoblastos , Transducción de Señal/efectos de los fármacos , Cráneo , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Animales , Proteína Morfogenética Ósea 2/biosíntesis , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Osteoblastos/metabolismo , Osteoblastos/patología , Cráneo/lesiones , Cráneo/metabolismo , Cráneo/patología , Proteína smad6/biosíntesis , Factor de Crecimiento Transformador beta/metabolismo
4.
Eur Rev Med Pharmacol Sci ; 20(4): 715-20, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26957275

RESUMEN

OBJECTIVE: Rheumatic heart disease (RHD) results due to the cross reaction of the host immune system when it develops immunity against group A streptococcal infection. This autoimmune disease progress with different pathological conditions and the genes associated with it are still less understood. MATERIALS AND METHODS: To understand the role of NKX2-5 and Smad-6 in developing an RHD, we successfully developed RHD model using BALB/c mice and we evaluate the expression of NKX2-5 and Smad-6 in different conditions. RESULTS: The disease conditions are confirmed through histological sectioning of RHD heart tissue with its associated Aschoff bodies. The histological of control heart tissue in the absence of NKX2-5 looks abnormal with an enlarged nucleus and in the absence of Smad-6 the solid nature of heart tissue loosens. The mice developed a complex form of acute RHD with tissue hardening in the absence of either NKX2-5 or Smad-6 which are confirmed in NKX2-5 or Smad-6 null mice. Immunohistochemical studies reveal that the NKX2-5 and Smad-6 expression get down regulated on developing with RHD. Through experiments, we detected that both Nkx2-5 and Smad-6 are both inter-dependable and it negatively regulated each other by inhibiting them. In the absence of NKX2-5 or Smad-6, a severe form of RHD is observed together with down-regulation of either NKX2-5 or Smad-6. CONCLUSIONS: The present investigation of NKX2-5 and Smad-6 in RHD provides a new insight of data that helps to understand the disease pathogenesis.


Asunto(s)
Proteínas de Homeodominio/biosíntesis , Cardiopatía Reumática/metabolismo , Proteína smad6/biosíntesis , Factores de Transcripción/biosíntesis , Animales , Femenino , Regulación de la Expresión Génica , Proteína Homeótica Nkx-2.5 , Proteínas de Homeodominio/genética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Cardiopatía Reumática/genética , Cardiopatía Reumática/patología , Proteína smad6/genética , Infecciones Estreptocócicas/genética , Infecciones Estreptocócicas/metabolismo , Infecciones Estreptocócicas/patología , Factores de Transcripción/genética
5.
J Steroid Biochem Mol Biol ; 152: 8-15, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25889901

RESUMEN

Melatonin has been reported to suppress adrenocorticotropin (ACTH) secretion in the anterior pituitary and cortisol production in the adrenal by different mechanisms. However, the effect of melatonin on aldosterone production has remained unknown. In this study, we investigated the role of melatonin in the regulation of aldosterone production using human adrenocortical H295R cells by focusing on the activin system expressed in the adrenal. Melatonin receptor MT1 mRNA and protein were expressed in H295R cells and the expression levels of MT1 were increased by activin treatment. Activin increased ACTH-induced, but not angiotensin II (Ang II)-induced, aldosterone production. Melatonin alone did not affect basal synthesis of either aldosterone or cortisol. However, melatonin effectively enhanced aldosterone production induced by co-treatment with ACTH and activin, although melatonin had no effect on aldosterone production induced by Ang II in combination with activin. These changes in steroidogenesis became apparent when the steroid production was evaluated by the ratio of aldosterone/cortisol. Melatonin also enhanced dibutyryl-AMP-induced aldosterone/cortisol levels in the presence of activin, suggesting a functional link to the cAMP-PKA pathway for induction of aldosterone production by melatonin and activin. In accordance with the data for steroids, ACTH-induced, but not Ang II-induced, cAMP synthesis was also amplified by co-treatment with melatonin and activin. Furthermore, the ratio of ACTH-induced mRNA level of CYP11B2 compared with that of CYP17 was amplified in the condition of treatment with both melatonin and activin. In addition, melatonin increased expression of the activin type-I receptor ALK-4 but suppressed expression of inhibitory Smads6/7, leading to the enhancement of Smad2 phosphorylation. Collectively, the results showed that melatonin facilitated aldosterone production induced by ACTH and activin via the cAMP-PKA pathway. The results also suggested that mutual enhancement of melatonin and activin receptor signaling is involved in the induction of aldosterone output by adrenocortical cells.


Asunto(s)
Activinas/farmacología , Corteza Suprarrenal/metabolismo , Hormona Adrenocorticotrópica/metabolismo , Aldosterona/biosíntesis , Melatonina/farmacología , Receptores de Activinas Tipo I/biosíntesis , Corteza Suprarrenal/citología , Hormona Adrenocorticotrópica/farmacología , Angiotensina II/metabolismo , Línea Celular Tumoral , Citocromo P-450 CYP11B2/genética , Humanos , Hidrocortisona/biosíntesis , Fosforilación , ARN Mensajero/biosíntesis , Receptor de Melatonina MT1/biosíntesis , Receptor de Melatonina MT1/genética , Proteína Smad2/metabolismo , Proteína smad6/biosíntesis , Proteína smad7/biosíntesis , Esteroide 17-alfa-Hidroxilasa/genética
6.
Cancer Res ; 72(16): 4276-85, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22773665

RESUMEN

CD133+ cancer stem cells (CSC) contribute to hepatocellular carcinoma (HCC) progression and resistance to therapy. Bone morphogenetic protein BMP4 plays an important role in hepatogenesis and hepatic stem cell differentiation, but little is known about its function in hepatic CSCs. In this study, we showed that high-dose exogenous BMP4 promotes CD133+ HCC CSC differentiation and inhibits the self-renewal, chemotherapeutic resistance, and tumorigenic capacity of these cells. Interestingly, we found that low-dose exogenous BMP4 upregulated CD133 protein expression in vitro, and endogenous BMP4 was preferentially expressed in CD133+ HCC CSCs, suggesting that low doses of BMP4 may facilitate CSC maintenance. A reduction in endogenous BMP4 levels decreased CD133 protein expression in vitro. In HCC tissues, expression of the BMP4 signaling target gene SMAD6 was positively correlated with CD133 expression. Activation of the Erk1/2 signaling pathway led to BMP4-mediated reduction in CD133 expression, which was reversed by treatment with MEK inhibitors. Taken together, our findings indicated that BMP4 might be a potent therapeutic agent in HCC that targets CSCs.


Asunto(s)
Proteína Morfogenética Ósea 4/farmacología , Carcinoma Hepatocelular/tratamiento farmacológico , Neoplasias Hepáticas/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Antígeno AC133 , Antígenos CD/biosíntesis , Proteína Morfogenética Ósea 4/genética , Proteína Morfogenética Ósea 4/metabolismo , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patología , Diferenciación Celular/efectos de los fármacos , Glicoproteínas/biosíntesis , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patología , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Células Madre Neoplásicas/patología , Péptidos , Fosforilación , Proteína smad6/biosíntesis , Proteína smad6/genética
7.
Clin Exp Nephrol ; 16(5): 690-6, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22581061

RESUMEN

BACKGROUND: Glomerular hypertension aggravates glomerular sclerosis by inducing growth factors, e.g., transforming growth factor-ß (TGF-ß) to mesangial matrix expansion. Smads are intracellular proteins that transmit signals from TGF-ß to nucleus, and Smads are also negatively regulated by inhibitory Smads (I-Smads), Smad6 and Smad7. However, little is known about the role of I-Smads in glomerular hypertension. We studied I-Smad expression in cultured mesangial cells subjected to mechanical stretch as an in vitro model of glomerular hypertension. METHODS: Rat mesangial cells were cultured under cyclic mechanical stretch conditions using the Flexercell Strain Unit. Phosphorylated Smad1 and Smad2 were determined by Western blots. The expression of Smad6 and Smad7 mRNAs was determined by Northern blots. Stretch-mediated I-Smad mRNAs of cells pre-treated with MAPK-ERK kinase inhibitor, U0126, were also determined. Localization of phospho-Smad1, Smad6 and Smad7 proteins in the glomerulus of Dahl salt-sensitive rats was determined by immunohistochemistry. RESULTS: Stretch stress increased phospho-Smad1 levels, and significantly decreased Smad6 mRNA to 32 % of control, and increased Smad7 mRNA to 136 % of control. U0126 significantly attenuated stretch-mediated decreases in Smad6 mRNA, but had no effect on stretch-mediated increases in Smad7 mRNA. Phospho-Smad1, Smad6 and Smad7 proteins were localized in podocytes and mesangial cells of Dahl rats. CONCLUSION: Mechanical stretch increases phospho-Smad1 levels and down-regulates Smad6 mRNA expression in mesangial cells. Stretch-mediated down-regulation of Smad6 is partially involved in ERK1/2 activation. These results indicate that glomerular hypertension might augment Smad1 signaling with concomitant attenuation of Smad6-mediated negative feedback.


Asunto(s)
Proteína smad6/biosíntesis , Proteína smad7/biosíntesis , Estrés Mecánico , Animales , Regulación hacia Abajo , Mesangio Glomerular/citología , Mesangio Glomerular/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas Dahl , Proteína Smad1/biosíntesis
8.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 27(5): 1100-4, 2010 Oct.
Artículo en Chino | MEDLINE | ID: mdl-21089679

RESUMEN

This experiment was designed to construct mouse Smad6 recombinant RNA interference vectors and determine their interference effects on bone marrow mesenchymal stem cells (BMSCs). Three recombinant Smad6 RNA interference vectors were constructed by molecular clone techniques with a lenti-virus vector expressing green fluorescent protein (GFP), and the correctness of recombinant vectors was verified by DNA sequencing. Mouse BMSCs were used for transfection experiments and BMP-2 was in use for osteogenic induction of MSCs. The transfection efficiency of recombinant vectors was examined by Laser confocal scanning microscope and the interference effect of recombinant vectors on Smad6 gene expression was determined by real-time RT-PCR and Western blot, respectively. Three Smad6 recombinant RNA interference vectors were successfully constructed and their correctness was proved by DNA sequencing. After transfection, GFPs were effectively expressed in MSCs and all of three recombinant vectors gained high transfection efficiency (> 95%). Both real-time PCR and Western blot examination indicated that among three recombinant vectors, No. 2 Svector had the best interference effect and the interference effect was nearly 91% at protein level. In conclusion, Mouse recombinant Smad6 RNA interference (RNAi) vector was successfully constructed and it provided an effective tool for further studies on BMP signal pathways.


Asunto(s)
Proteínas Morfogenéticas Óseas/genética , Vectores Genéticos/genética , Lentivirus/genética , Interferencia de ARN , Proteína smad6/biosíntesis , Animales , Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Fluorescentes Verdes/biosíntesis , Proteínas Fluorescentes Verdes/genética , Lentivirus/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Proteína smad6/genética , Transfección
9.
Hepatology ; 52(4): 1266-73, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20658468

RESUMEN

UNLABELLED: Hereditary hemochromatosis (HH) is a common inherited iron overload disorder. The vast majority of patients carry the missense Cys282Tyr mutation of the HFE gene. Hepcidin, the central regulator of iron homeostasis, is deficient in HH, leading to unchecked iron absorption and subsequent iron overload. The bone morphogenic protein (BMP)/small mothers against decapentaplegic (Smad) signaling cascade is central to the regulation of hepcidin. Recent data from HH mice models indicate that this pathway may be defective in the absence of the HFE protein. Hepatic BMP/Smad signaling has not been characterized in a human HFE-HH cohort to date. Hepatic expression of BMP/Smad-related genes was examined in 20 HFE-HH males with significant iron overload, and compared to seven male HFE wild-type controls using quantitative real-time reverse transcription polymerase chain reaction. Hepatic expression of BMP6 was appropriately elevated in HFE-HH compared to controls (P = 0.02), likely related to iron overload. Despite this, no increased expression of the BMP target genes hepcidin and Id1 was observed, and diminished phosphorylation of Smad1/Smad5/Smad8 protein relative to iron burden was found upon immunohistochemical analysis, suggesting that impaired BMP signaling occurs in HFE-HH. Furthermore, Smad6 and Smad7, inhibitors of BMP signaling, were up-regulated in HFE-HH compared to controls (P = 0.001 and P = 0.018, respectively). CONCLUSION: New data arising from this study suggest that impaired BMP signaling underlies the hepcidin deficiency of HFE-HH. Moreover, the inhibitory Smads, Smad6, and Smad7 are identified as potential disruptors of this signal and, hence, contributors to the pathogenesis of this disease.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/deficiencia , Proteína Morfogenética Ósea 6/fisiología , Transducción de Señal/fisiología , Proteína smad6/biosíntesis , Proteína smad7/biosíntesis , Adulto , Hemocromatosis/genética , Proteína de la Hemocromatosis , Hepcidinas , Antígenos de Histocompatibilidad Clase I/genética , Humanos , Sobrecarga de Hierro/genética , Masculino , Proteínas de la Membrana/genética , Persona de Mediana Edad , Proteína Smad8/genética , Regulación hacia Arriba
10.
Mol Cancer ; 9: 106, 2010 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-20462450

RESUMEN

BACKGROUND: To test if the expression of Smad1-8 mRNAs were predictive of survival in patients with oral squamous cell carcinoma (SCC). PATIENTS AND METHODS: We analyzed, prospectively, the expression of Smad1-8, by means of Ribonuclease Protection Assay in 48 primary, operable, oral SCC. In addition, 21 larynx, 10 oropharynx and 4 hypopharynx SCC and 65 matched adjacent mucosa, available for study, were also included. For survival analysis, patients were categorized as positive or negative for each Smad, according to median mRNA expression. We also performed real-time quantitative PCR (QRTPCR) to asses the pattern of TGFbeta1, TGFbeta2, TGFbeta3 in oral SCC. RESULTS: Our results showed that Smad2 and Smad6 mRNA expression were both associated with survival in Oral SCC patients. Cox Multivariate analysis revealed that Smad6 positivity and Smad2 negativity were both predictive of good prognosis for oral SCC patients, independent of lymph nodal status (P = 0.003 and P = 0.029, respectively). In addition, simultaneously Smad2- and Smad6+ oral SCC group of patients did not reach median overall survival (mOS) whereas the mOS of Smad2+/Smad6- subgroup was 11.6 months (P = 0.004, univariate analysis). Regarding to TGFbeta isoforms, we found that Smad2 mRNA and TGFbeta1 mRNA were inversely correlated (p = 0.05, R = -0.33), and that seven of the eight TGFbeta1+ patients were Smad2-. In larynx SCC, Smad7- patients did not reach mOS whereas mOS of Smad7+ patients were only 7.0 months (P = 0.04). No other correlations were found among Smad expression, clinico-pathological characteristics and survival in oral, larynx, hypopharynx, oropharynx or the entire head and neck SCC population. CONCLUSION: Smad6 together with Smad2 may be prognostic factors, independent of nodal status in oral SCC after curative resection. The underlying mechanism which involves aberrant TGFbeta signaling should be better clarified in the future.


Asunto(s)
Biomarcadores de Tumor/análisis , Carcinoma de Células Escamosas/metabolismo , Neoplasias de la Boca/metabolismo , Proteína Smad2/biosíntesis , Proteína smad6/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/mortalidad , Femenino , Expresión Génica , Humanos , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Neoplasias de la Boca/genética , Neoplasias de la Boca/mortalidad , Pronóstico , ARN Mensajero/análisis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína Smad2/genética , Proteína smad6/genética , Factor de Crecimiento Transformador beta1/biosíntesis , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta2/biosíntesis , Factor de Crecimiento Transformador beta2/genética , Factor de Crecimiento Transformador beta3/biosíntesis , Factor de Crecimiento Transformador beta3/genética
11.
J Appl Toxicol ; 29(4): 364-6, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19058293

RESUMEN

Retinoic acid can cause malformations of the developing nervous system. Smad signaling is involved in embryonic development. The current study investigated all-trans-retinoic acid (ATRA)-induced alteration of Smad expression in the developing neural tubes of mice. Pregnant mice were treated with a single dose of 50 mg/kg ATRA by oral gavage on embryonic day E7. Western immunoblotting was used to examine Smads proteins, particularly phosphorylated (p-) Smad1, total Smad1 and Smad6 in the neural tissue of the embryos on E8-E11 following treatment. Results showed that ATRA treatment significantly increased expression of both p-Smad1 and total Smad1, while Smad6 was decreased in neural tissues of ATRA-exposed embryos in utero from E8 to E11, a critical period for neural tube formation. Data suggest that disruption of Smad signaling may be involved in ATRA-induced neural tube defects.


Asunto(s)
Neuronas/metabolismo , Proteínas Smad/biosíntesis , Tretinoina/toxicidad , Actinas/biosíntesis , Animales , Western Blotting , Femenino , Expresión Génica/efectos de los fármacos , Ratones , Tubo Neural/efectos de los fármacos , Tubo Neural/crecimiento & desarrollo , Fosforilación , Embarazo , Proteínas Smad/genética , Proteína Smad1/biosíntesis , Proteína Smad1/genética , Proteína smad6/biosíntesis , Proteína smad6/genética
12.
Cancer Res ; 68(23): 9686-92, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19047146

RESUMEN

The malignant transformation in several types of cancer, including lung cancer, results in a loss of growth inhibition by transforming growth factor-beta (TGF-beta). Here, we show that SMAD6 expression is associated with a reduced survival in lung cancer patients. Short hairpin RNA (shRNA)-mediated knockdown of SMAD6 in lung cancer cell lines resulted in reduced cell viability and increased apoptosis as well as inhibition of cell cycle progression. However, these results were not seen in Beas2B, a normal bronchial epithelial cell line. To better understand the mechanism underlying the association of SMAD6 with poor patient survival, we used a lentivirus construct carrying shRNA for SMAD6 to knock down expression of the targeted gene. Through gene expression analysis, we observed that knockdown of SMAD6 led to the activation of TGF-beta signaling through up-regulation of plasminogen activator inhibitor-1 and phosphorylation of SMAD2/3. Furthermore, SMAD6 knockdown activated the c-Jun NH2-terminal kinase pathway and reduced phosphorylation of Rb-1, resulting in increased G0-G1 cell arrest and apoptosis in the lung cancer cell line H1299. These results jointly suggest that SMAD6 plays a critical role in supporting lung cancer cell growth and survival. Targeted inactivation of SMAD6 may provide a novel therapeutic strategy for lung cancers expressing this gene.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Proteína smad6/biosíntesis , Factor de Crecimiento Transformador beta/metabolismo , Apoptosis/fisiología , Carcinoma de Pulmón de Células no Pequeñas/genética , Ciclo Celular/fisiología , Procesos de Crecimiento Celular/fisiología , Línea Celular Tumoral , Regulación hacia Abajo , Humanos , Inmunohistoquímica , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Neoplasias Pulmonares/genética , Fosforilación , ARN Interferente Pequeño/genética , Transducción de Señal , Proteína smad6/deficiencia , Proteína smad6/genética , Transducción Genética
13.
J Biol Chem ; 283(40): 27154-64, 2008 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-18644788

RESUMEN

Bone morphogenetic proteins (BMPs) play critical roles at various stages in endochondral bone formation. In vitro studies have demonstrated that Smad7 regulates transforming growth factor-beta and BMP signals by inhibiting Smad pathways in chondrocytes. However, the in vivo roles of Smad7 during cartilage development are unknown. To investigate distinct effects of Smad7 at different stages during chondrocyte differentiation, we generated a series of conditional transgenic mice that overexpress Smad7 in chondrocytes at various steps of differentiation by using the Cre/loxP system. We generated Col11a2-lacZ(floxed)-Smad7 transgenic mice and mated them with three types of Cre transgenic mice to obtain Smad7(Prx1), Smad7(11Enh), and Smad7(11Prom) conditional transgenic mice. Smad7(Prx1) mice overexpressing Smad7 in condensing mesenchymal cells showed disturbed mesenchymal condensation associated with decreased Sox9 expression, leading to poor cartilage formation. Smad7(11Enh) mice overexpressing Smad7 in round chondrocytes showed decreased chondrocyte proliferation rates. Smad7(11Prom) mice overexpressing Smad7 in flat chondrocytes showed inhibited maturation of chondrocytes toward hypertrophy. Micromass culture of mesenchymal cells showed that BMP-induced cartilaginous nodule formation was down-regulated by overexpression of Smad7, but not Smad6. Overexpression of Smad7, but not Smad6, down-regulated the phosphorylation of p38 MAPKs. Our data provide in vivo evidence for distinct effects of Smad7 at different stages during chondrocyte differentiation and suggest that Smad7 in prechondrogenic cells inhibits chondrocyte differentiation possibly by down-regulating BMP-activated p38 MAPK pathways.


Asunto(s)
Diferenciación Celular/fisiología , Condrocitos/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Osteogénesis/fisiología , Proteína smad7/biosíntesis , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Proteínas Morfogenéticas Óseas/biosíntesis , Proteínas Morfogenéticas Óseas/genética , Condrocitos/citología , Regulación hacia Abajo/fisiología , Proteínas del Grupo de Alta Movilidad/biosíntesis , Proteínas del Grupo de Alta Movilidad/genética , Ratones , Ratones Transgénicos , Fosforilación , Factor de Transcripción SOX9 , Proteína smad6/biosíntesis , Proteína smad6/genética , Proteína smad7/genética , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Proteínas Quinasas p38 Activadas por Mitógenos/genética
14.
J Biol Chem ; 282(14): 10742-8, 2007 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-17215250

RESUMEN

BMP-2 plays an essential role in osteoblast and chondrocyte differentiation, but its signaling mechanism has not been fully defined. In the present studies, we investigated the mechanism through which BMP-2 activates the Smad6 gene. A -2006/+45 Smad6 promoter-luciferase construct was generated along with deletions and Runx2 binding site mutations to examine the role of Smad1 and Runx2 signaling following BMP-2 stimulation in osteoblasts. Transfection of Runx2 or treatment with BMP-2-stimulated promoter activity of the -2006/+45 and -1191/+45 reporters but not the -829/+45 and -374/+45 reporters. No Smad1/5 binding site is present in the -1191/-829 region of the Smad6 promoter. Mutation of the OSE2-a site (-1036/-1031) completely abolished the stimulatory effect of Runx2 as well as BMP-2 on the -2006/+45 and -1191/+45 Smad6 reporters. Gel shift and chromatin immunoprecipitation (ChIP) assays showed that Runx2 binds the OSE2-a element. ChIP assays demonstrated that Smad1 also interacts with the OSE2-a site at the Smad6 promoter through Runx2. The protein degradation of Runx2 is mediated by the E3 ubiquitin ligase Smurf1. In the present studies, we found that Smurf1 binds the OSE2-a site through Runx2 and inhibits Smad6 gene transcription. Treatment with BMP-2 and transfection of Smad1 abolished Smurf1 binding to the OSE2 site. These results show that Smad1 binding excludes Smurf1 interaction with the OSE2 site and promotes Smad6 gene transcription.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Condrocitos/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Osteoblastos/metabolismo , Proteína smad6/biosíntesis , Transcripción Genética/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Sitios de Unión/fisiología , Proteína Morfogenética Ósea 2 , Proteínas Morfogenéticas Óseas/farmacología , Línea Celular , Condrocitos/citología , Ratones , Mutación/fisiología , Especificidad de Órganos , Osteoblastos/citología , Elementos de Respuesta/fisiología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Proteína Smad1/metabolismo , Proteína Smad5/metabolismo , Proteína smad6/genética , Factor de Crecimiento Transformador beta/farmacología , Ubiquitina-Proteína Ligasas/metabolismo
15.
Dev Dyn ; 235(1): 50-9, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16245329

RESUMEN

The proepicardium (PE) migrates over the heart and forms the epicardium. A subset of these PE-derived cells undergoes epithelial-mesenchymal transformation (EMT) and gives rise to cardiac fibroblasts and components of the coronary vasculature. We report that transforming growth factor-beta (TGFbeta) 1 and TGFbeta2 increase EMT in PE explants as measured by invasion into a collagen gel, loss of cytokeratin expression, and redistribution of ZO1. The type I TGFbeta receptors ALK2 and ALK5 are both expressed in the PE. However, only constitutively active (ca) ALK2 stimulates PE-derived epithelial cell activation, the first step in transformation, whereas caALK5 stimulates neither activation nor transformation in PE explants. Overexpression of Smad6, an inhibitor of ALK2 signaling, inhibits epithelial cell activation, whereas BMP7, a known ligand for ALK2, has no effect. These data demonstrate that TGFbeta stimulates transformation in the PE and suggest that ALK2 partially mediates this effect.


Asunto(s)
Diferenciación Celular/fisiología , Epitelio/embriología , Mesodermo/metabolismo , Pericardio/embriología , Factor de Crecimiento Transformador beta/fisiología , Animales , Embrión de Pollo , Epitelio/metabolismo , Factor 1 de Crecimiento de Fibroblastos/fisiología , Factor 7 de Crecimiento de Fibroblastos/fisiología , Genes Reporteros , Proteínas de la Membrana/biosíntesis , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Mesodermo/citología , Pericardio/citología , Pericardio/metabolismo , Fosfoproteínas/biosíntesis , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteína smad6/biosíntesis , Proteína smad6/genética , Proteína de la Zonula Occludens-1
16.
J Leukoc Biol ; 79(1): 192-201, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16282533

RESUMEN

The bone morphogenetic protein receptor-2 (BMPR2) is a member of the transforming growth factor-beta receptor family and is expressed on the surface of several cell types including endothelial cells and macrophages. Recently, a cause for familial primary pulmonary hypertension (FPPH) has been identified as mutations in the gene encoding BMPR2. Three forms of pulmonary hypertension (PH) exist, including PPH, FPPH, and PH secondary to other etiologies (sporadic PH) such as drug abuse and human immunodeficiency virus (HIV) infection. It is interesting that these subtypes are histologically indistinguishable. The macrophage is a key target cell for HIV-1, significantly altering macrophage cell function upon infection. HIV-1 trans-activator of transcription (Tat), an immediate-early product of the HIV-1 lifecycle, plays an important role in mediating HIV-induced modulation of host cell function. Our laboratory has previously shown that Tat represses mannose receptor transcription in macrophages. In the current study, we examined activity from the BMPR2 promoter in the macrophage cell line U937 and potential regulation by Tat. Transfection of U937 cells with BMPR2 promoter-reporter constructs revealed dose-dependent repression of BMPR2 promoter activity in the presence of Tat. Experiments using truncations of the BMPR2 promoter localized Tat-mediated repression to the first 208 bases of the promoter. Decreased BMPR2 transcription resulted in altered downstream signaling. Similar to mothers against decapentaplegics (SMAD) phosphorylation and SMAD6 expression, in response to BMP2 treatment, were down-regulated after Tat treatment. Finally, HIV-1 infection and treatment with Tat protein of the U937 human monocytic cell line resulted in a decreased, endogenous BMPR2 transcript copy number.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo II/biosíntesis , Regulación hacia Abajo , Productos del Gen tat/metabolismo , Infecciones por VIH/metabolismo , VIH-1 , Transducción de Señal , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Células Endoteliales/metabolismo , Productos del Gen tat/farmacología , Infecciones por VIH/genética , Humanos , Hipertensión Pulmonar/genética , Lectinas Tipo C/biosíntesis , Lectinas Tipo C/genética , Macrófagos/metabolismo , Macrófagos/virología , Receptor de Manosa , Lectinas de Unión a Manosa/biosíntesis , Lectinas de Unión a Manosa/genética , Mutación , Fosforilación , Procesamiento Proteico-Postraduccional , Receptores de Superficie Celular/biosíntesis , Receptores de Superficie Celular/genética , Elementos de Respuesta/genética , Proteína smad6/biosíntesis , Proteína smad6/genética , Células U937 , Productos del Gen tat del Virus de la Inmunodeficiencia Humana
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...