Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cancers (Basel) ; 14(24)2022 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-36551660

RESUMEN

Two vascular endothelial growth factor (VEGF) receptors, FLT-1 and KDR, are expressed preferentially in proliferating endothelium. There is increasing evidence that recombinant, soluble VEGF receptor domains interfering with VEGF signaling may inhibit in vivo neoangiogenesis, tumor growth and metastatic spread. We hypothesized that a soluble form of FLT-1 receptor (sFLT-1) could inhibit the growth of pre-established tumors via an anti-angiogenic mechanism. A replication-deficient adenovirus (Ad) vector carrying the sflt-1 cDNA (Adsflt) was used to overexpress the sFLT-1 receptor in a breast cancer animal model. MCF-7 cells, which produce VEGF, were used to establish solid tumors in the mammary fat pads of female nude mice. After six weeks, tumors were injected either with Adsflt or a negative control virus (AdCMV.ßgal). After six months, average tumor volume in the Adsflt-infected group (33 ± 22 mm3) decreased by 91% relative to that of the negative control group (388 ± 94 mm3; p < 0.05). Moreover, 10 of 15 Adsflt-infected tumors exhibited complete regression. The vascular density of Adsflt-infected tumors was reduced by 50% relative to that of negative controls (p < 0.05), which is consistent with sFLT-1-mediated tumor regression through an anti-angiogenic mechanism. Moreover, cell necrosis and fibrosis associated with long-term regression of Adsflt−infected tumors were preceded by apoptosis of tumor vascular endothelial cells. Mice treated with Adsflt intratumorally showed no delay in the healing of cutaneous wounds, providing preliminary evidence that Ad-mediated sFLT-1 overexpression may be an effective anti-angiogenic therapy for cancer without the risk of systemic anti-angiogenic effects.

2.
Immunobiology ; 216(3): 343-50, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20961651

RESUMEN

The pleiotropic cytokine transforming growth factor-ß (TGF-ß) signals through different pathways among which the Smad- and the MAP-Kinase pathways are already well characterized. Both pathways utilize adaptor/chaperone molecules that facilitate or modulate the intracellular signaling events. Two of the proteins shown in vitro to play a role in Smad-dependent signaling are the TGF-ß Receptor Associated Protein-1 (TRAP1, also TGFBRAP1) and its homologue VPS39, also known as Vam6 and TRAP1-Like-Protein (TLP). We generated mice deficient for TRAP1 and VPS39/TLP, respectively. Absence of TRAP1 protein results in death at either of two defined timepoints during embryogenesis, before the blastula stage or during gastrulation, whereas most of the VPS39 deficient mice die before E6.5. Heterozygous mice show no overt phenotype. In summary, our data indicate that TRAP1 and VPS39 are nonredundant and essentially required for early embryonic development.


Asunto(s)
Blástula/embriología , Desarrollo Embrionario , Gástrula/embriología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Animales Modificados Genéticamente , Proteínas Relacionadas con la Autofagia , Northern Blotting , Western Blotting , Células Cultivadas , Expresión Génica , Genotipo , Factores de Intercambio de Guanina Nucleótido/genética , Proteínas HSP90 de Choque Térmico , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Reacción en Cadena de la Polimerasa , Transducción de Señal , Proteínas de Transporte Vesicular
3.
J Cell Biochem ; 111(2): 310-21, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20506405

RESUMEN

Hepatocyte growth factor (HGF)-stimulated mitogenesis, motogenesis and morphogenesis in various cell types begins with activation of the Met receptor tyrosine kinase and the recruitment of intracellular adaptors and kinase substrates. The adapter protein Gab1 is a critical effector and substrate of activated Met, mediating morphogenesis, among other activities, in epithelial cells. To define its role downstream of Met in hematopoietic cells, Gab1 was expressed in the HGF-responsive, Gab1-negative murine myeloid cell line 32D. Interestingly, the adhesion and motility of Gab1-expressing cells were significantly greater than parental cells, independent of growth factor treatment. Downstream of activated Met, Gab1 expression was specifically associated with rapid Shp-2 recruitment and activation, increased mitogenic potency, suppression of GATA-1 expression and concomitant upregulation of GATA-2 transcription. In addition to enhanced proliferation, continuous culture of Gab1-expressing 32D cells in HGF resulted in cell attachment, filopodia extension and phenotypic changes suggestive of monocytic differentiation. Our results suggest that in myeloid cells, Gab1 is likely to enhance HGF mitogenicity by coupling Met to Shp-2 and GATA-2 expression, thereby potentially contributing to normal myeloid differentiation as well as oncogenic transformation.


Asunto(s)
Factor de Crecimiento de Hepatocito/fisiología , Mitógenos , Morfogénesis , Células Madre Multipotentes/fisiología , Células Progenitoras Mieloides/citología , Fosfoproteínas/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Diferenciación Celular , Línea Celular , Transformación Celular Neoplásica , Factor de Transcripción GATA2 , Ratones , Células Progenitoras Mieloides/fisiología , Proteína Tirosina Fosfatasa no Receptora Tipo 11 , Proteínas Proto-Oncogénicas c-met , Seudópodos
4.
J Cell Biol ; 183(3): 409-17, 2008 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-18955553

RESUMEN

The blood-brain barrier (BBB) is confined to the endothelium of brain capillaries and is indispensable for fluid homeostasis and neuronal function. In this study, we show that endothelial Wnt/beta-catenin (beta-cat) signaling regulates induction and maintenance of BBB characteristics during embryonic and postnatal development. Endothelial specific stabilization of beta-cat in vivo enhances barrier maturation, whereas inactivation of beta-cat causes significant down-regulation of claudin3 (Cldn3), up-regulation of plamalemma vesicle-associated protein, and BBB breakdown. Stabilization of beta-cat in primary brain endothelial cells (ECs) in vitro by N-terminal truncation or Wnt3a treatment increases Cldn3 expression, BBB-type tight junction formation, and a BBB characteristic gene signature. Loss of beta-cat or inhibition of its signaling abrogates this effect. Furthermore, stabilization of beta-cat also increased Cldn3 and barrier properties in nonbrain-derived ECs. These findings may open new therapeutic avenues to modulate endothelial barrier function and to limit the devastating effects of BBB breakdown.


Asunto(s)
Barrera Hematoencefálica/fisiología , Sistema Nervioso Central/fisiología , Circulación Cerebrovascular/fisiología , Neovascularización Fisiológica/fisiología , Proteínas Wnt/fisiología , beta Catenina/fisiología , Animales , Genes Reporteros , Humanos , Ratones , Modelos Animales , Transducción de Señal , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo
5.
EMBO J ; 27(7): 993-1004, 2008 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-18337748

RESUMEN

VE-cadherin is an endothelial-specific transmembrane protein concentrated at cell-to-cell adherens junctions. Besides promoting cell adhesion and controlling vascular permeability, VE-cadherin transfers intracellular signals that contribute to vascular stabilization. However, the molecular mechanism by which VE-cadherin regulates vascular homoeostasis is still poorly understood. Here, we report that VE-cadherin expression and junctional clustering are required for optimal transforming growth factor-beta (TGF-beta) signalling in endothelial cells (ECs). TGF-beta antiproliferative and antimigratory responses are increased in the presence of VE-cadherin. ECs lacking VE-cadherin are less responsive to TGF-beta/ALK1- and TGF-beta/ALK5-induced Smad phosphorylation and target gene transcription. VE-cadherin coimmunoprecipitates with all the components of the TGF-beta receptor complex, TbetaRII, ALK1, ALK5 and endoglin. Clustered VE-cadherin recruits TbetaRII and may promote TGF-beta signalling by enhancing TbetaRII/TbetaRI assembly into an active receptor complex. Taken together, our data indicate that VE-cadherin is a positive and EC-specific regulator of TGF-beta signalling. This suggests that reduction or inactivation of VE-cadherin may contribute to progression of diseases where TGF-beta signalling is impaired.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/farmacología , Receptores de Activinas Tipo II/metabolismo , Alantoides/citología , Alantoides/efectos de los fármacos , Alantoides/metabolismo , Animales , Cadherinas/deficiencia , Movimiento Celular/efectos de los fármacos , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Dimerización , Embrión de Mamíferos/citología , Embrión de Mamíferos/efectos de los fármacos , Embrión de Mamíferos/metabolismo , Células Endoteliales/citología , Humanos , Cinética , Ratones , Modelos Biológicos , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Proteína Smad2/metabolismo , Proteína smad3/metabolismo , Transcripción Genética/efectos de los fármacos
6.
N Engl J Med ; 351(6): 552-9, 2004 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-15295048

RESUMEN

BACKGROUND: The receptors for transforming growth factor beta (TGF-beta) and their signaling intermediates make up an important tumor-suppressor pathway. The role of one of these intermediates--Smad3--in the pathogenesis of lymphoid neoplasia is unknown. METHODS: We measured Smad3 messenger RNA (mRNA) and protein in leukemia cells obtained at diagnosis from 19 children with acute leukemia, including 10 with T-cell acute lymphoblastic leukemia (ALL), 7 with pre-B-cell ALL, and 2 with acute nonlymphoblastic leukemia (ANLL). All nine exons of the SMAD3 gene (MADH3) were sequenced. Mice in which one or both alleles of Smad3 were inactivated were used to evaluate the role of Smad3 in the response of normal T cells to TGF-beta and in the susceptibility to spontaneous leukemogenesis in mice in which both alleles of the tumor suppressor p27Kip1 were deleted. RESULTS: Smad3 protein was absent in T-cell ALL but present in pre-B-cell ALL and ANLL. No mutations were found in the MADH3 gene in T-cell ALL, and Smad3 mRNA was present in T-cell ALL and normal T cells at similar levels. In mice, the loss of one allele for Smad3 impairs the inhibitory effect of TGF-beta on the proliferation of normal T cells and works in tandem with the homozygous inactivation of p27Kip1 to promote T-cell leukemogenesis. CONCLUSIONS: Loss of Smad3 protein is a specific feature of pediatric T-cell ALL. A reduction in Smad3 expression and the loss of p27Kip1 work synergistically to promote T-cell leukemogenesis in mice.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proteínas de Unión al ADN/metabolismo , Leucemia de Células T/metabolismo , Leucemia-Linfoma de Células T del Adulto/metabolismo , Linfocitos T/metabolismo , Transactivadores/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Supresoras de Tumor/genética , Adulto , Animales , Proteínas de Ciclo Celular/metabolismo , Niño , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Proteínas de Unión al ADN/genética , Exones , Eliminación de Gen , Expresión Génica , Humanos , Interleucina-2/biosíntesis , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia de Células T/genética , Leucemia-Linfoma de Células T del Adulto/genética , Ratones , Ratones Noqueados , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Análisis de Secuencia de ADN , Transducción de Señal , Proteína smad3 , Transactivadores/genética , Proteínas Supresoras de Tumor/metabolismo
7.
Blood ; 103(12): 4594-601, 2004 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-15016653

RESUMEN

Tgf-beta1-/- mice develop a progressive, lethal, inflammatory syndrome, but mechanisms leading to the spontaneous activation of Tgf-beta1-/- T cells remain unclear. Here we show the disruption of CD28 gene expression accelerates disease in Tgf-beta1-/- mice, and we link this increase in severity to a reduction in the number of CD4+CD25+ regulatory T cells. CD4+CD25+ T cells develop normally in Tgf-beta1-/- mice and display characteristic expression of cytotoxic T lymphocyte-associated antigen 4 (CTLA-4), glucocorticoid-induced tumor necrosis factor receptor (GITR), alpha(E)beta7 integrin, and Foxp3. Adoptive transfer of Tgf-beta1-/- splenocytes to Tgf-beta1+/+/Rag2-/- mice induced an autoimmune inflammatory disease with features similar to those of the Tgf-beta1-/- phenotype, and disease transfer was accelerated by the depletion of Tgf-beta1-/- CD4+CD25+ T cells from donor splenocytes. Cotransfer of Tgf- beta1-/- CD4+CD25+ T cells clearly attenuated disease in Rag2-/- recipients of CD25+-depleted Tgf-beta1-/- spleen and lymph node cells, but suppression was incomplete when compared with Tgf-beta1+/+ CD4+CD25+ T cells. These data demonstrate that CD4+CD25+ regulatory T cells develop in complete absence of endogenous transforming growth factor-beta1 (TGF-beta1) expression and that autocrine TGF-beta1 expression is not essential for these cells to suppress inflammation in vivo.


Asunto(s)
Antígenos CD28/genética , Subgrupos de Linfocitos T/inmunología , Linfocitos T/inmunología , Factor de Crecimiento Transformador beta/deficiencia , Factor de Crecimiento Transformador beta/inmunología , Animales , Antígenos CD4/genética , Separación Celular , Terapia de Inmunosupresión , Inflamación , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa , Receptores de Interleucina-2/genética , Subgrupos de Linfocitos T/citología , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta1
8.
Mol Cell Biol ; 24(6): 2251-62, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14993265

RESUMEN

Smad7 inhibits responses mediated by transforming growth factor beta (TGF-beta) and acts in a negative-feedback loop to regulate the intensity or duration of the TGF-beta signal. However, the aberrant expression and continued presence of Smad7 may cause TGF-beta resistance. Here we report that Jab1/CSN5, which is a component of the COP9 signalosome complex, associates constitutively with Smad7 and that overexpression of Jab1/CSN5 causes the translocation of Smad7 from the nucleus to the cytoplasm, promoting its degradation. Overexpression of Jab1/CSN5 increases Smad2 phosphorylation and enhances TGF-beta-induced transcriptional activity. The inhibition of endogenous Jab1/CSN5 expression by small interfering RNA (siRNA) induces Smad7 expression. This study thus defines Jab1/CSN5 as an adapter that targets Smad7 for degradation, thus releasing Smad7-mediated suppression of TGF-beta signaling.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Animales , Complejo del Señalosoma COP9 , Células COS , Línea Celular , Proteínas de Unión al ADN/genética , Regulación hacia Abajo , Humanos , Técnicas In Vitro , Péptidos y Proteínas de Señalización Intracelular , Modelos Biológicos , Complejos Multiproteicos , Péptido Hidrolasas , Unión Proteica , Proteínas/genética , ARN Interferente Pequeño/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Transducción de Señal , Proteína Smad2 , Proteína smad7 , Fracciones Subcelulares/metabolismo , Transactivadores/genética , Factores de Transcripción/genética , Transfección , Factor de Crecimiento Transformador beta/farmacología , Factor de Crecimiento Transformador beta1 , Técnicas del Sistema de Dos Híbridos
9.
J Biol Chem ; 279(15): 15167-76, 2004 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-14732719

RESUMEN

Radiation-induced fibrosis is an untoward effect of high dose therapeutic and inadvertent exposure to ionizing radiation. Transforming growth factor-beta (TGF-beta) has been proposed to be critical in tissue repair mechanisms resulting from radiation injury. Previously, we showed that interruption of TGF-beta signaling by deletion of Smad3 results in resistance to radiation-induced injury. In the current study, a small molecular weight molecule, halofuginone (100 nm), is demonstrated by reporter assays to inhibit the TGF-beta signaling pathway, by Northern blotting to elevate inhibitory Smad7 expression within 15 min, and by Western blotting to inhibit formation of phospho-Smad2 and phospho-Smad3 and to decrease cytosolic and membrane TGF-beta type II receptor (TbetaRII). Attenuation of TbetaRII levels was noted as early as 1 h and down-regulation persisted for 24 h. Halofuginone blocked TGF-beta-induced delocalization of tight junction ZO-1, a marker of epidermal mesenchymal transition, in NMuMg mammary epithelial cells and suggest halofuginone may have in vivo anti-fibrogenesis characteristics. After documenting the in vitro cellular effects, halofuginone (intraperitoneum injection of 1, 2.5, or 5 microg/mouse/day) efficacy was assessed using ionizing radiation-induced (single dose, 35 or 45 Gy) hind leg contraction in C3H/Hen mice. Halofuginone treatment alone exerted no toxicity but significantly lessened radiation-induced fibrosis. The effectiveness of radiation treatment (2 gray/day for 5 days) of squamous cell carcinoma (SCC) tumors grown in C3H/Hen was not affected by halofuginone. The results detail the molecular effects of halofuginone on the TGF-beta signal pathway and show that halofuginone may lessen radiation-induced fibrosis in humans.


Asunto(s)
Inhibidores de la Síntesis de la Proteína/uso terapéutico , Quinazolinas/uso terapéutico , Neumonitis por Radiación/tratamiento farmacológico , Factor de Crecimiento Transformador beta/metabolismo , Animales , Northern Blotting , Western Blotting , Células COS , Carcinoma de Células Escamosas/tratamiento farmacológico , Línea Celular , Línea Celular Tumoral , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Fibrosis/metabolismo , Eliminación de Gen , Genes Reporteros , Humanos , Immunoblotting , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Endogámicos C3H , Microscopía Confocal , Microscopía Fluorescente , Piperidinas , Plásmidos/metabolismo , Quinazolinonas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Proteína smad3 , Factores de Tiempo , Transactivadores/metabolismo , Factor de Crecimiento Transformador beta1
10.
Cancer Res ; 63(23): 8284-92, 2003 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-14678987

RESUMEN

The role of transforming growth factor beta in breast cancer is controversial with tumor suppressor and pro-oncogenic activities having been demonstrated. To address whether the same or different signal transduction pathways mediate these opposing activities, we manipulated the Smad2/3 signaling pathway in cells of common origin but differing degrees of malignancy derived from MCF10A human breast cells. We show that interference with endogenous Smad2/3 signaling enhances the malignancy of xenografted tumors of premalignant and well-differentiated tumor cells but strongly suppresses lung metastases of more aggressive carcinoma cells after tail vein injection. Overexpression of Smad3 in the same cells has opposite effects. The data demonstrate that the Smad2/3 signaling pathway mediates tumor suppressor and prometastatic signals, depending on the cellular context.


Asunto(s)
Neoplasias de la Mama/patología , Proteínas de Unión al ADN/fisiología , Transactivadores/fisiología , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , División Celular/fisiología , Línea Celular Tumoral , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Regulación hacia Abajo , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Metástasis de la Neoplasia , Trasplante de Neoplasias , Fosforilación , Transducción de Señal , Proteína Smad2 , Proteína smad3 , Transactivadores/antagonistas & inhibidores , Transactivadores/biosíntesis , Transactivadores/genética , Transfección , Factor de Crecimiento Transformador beta/fisiología , Trasplante Heterólogo
11.
EMBO J ; 22(17): 4465-77, 2003 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-12941698

RESUMEN

Transforming growth factor-beta (TGF-beta) is a multifunctional cytokine signaling to the nucleus through cell surface transmembrane receptor serine/threonine kinases and cytoplasmic effectors, including Smad proteins. We describe a novel modulator of this pathway, TLP (TRAP-1-like protein), which is 25% identical to the previously described Smad4 chaperone, TRAP-1, and shows identical expression patterns in human tissues. Endogenous TLP associates with both active and kinase-deficient TGF-beta and activin type II receptors, but interacts with the common-mediator Smad4 only in the presence of TGF-beta/activin signaling. Overexpression of TLP represses the ability of TGF-beta to induce transcription from SBE-Luc, a Smad3/4-specific reporter, while it potentiates transcription from ARE-Luc, a Smad2/4-specific reporter. Consistent with this, TLP inhibits the formation of Smad3/4 complexes in the absence of effects on phosphorylation of Smad3, while it affects neither Smad2 phosphorylation nor hetero-oligomerization. We propose that TLP might regulate the balance of Smad2 and Smad3 signaling by localizing Smad4 intracellularly, thus contributing to cellular specificity of TGF-beta transcriptional responses in both normal and pathophysiology.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteínas de Unión al ADN/metabolismo , Péptidos y Proteínas de Señalización Intracelular , Proteínas de la Membrana , Transactivadores/metabolismo , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Activinas/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Relacionadas con la Autofagia , Células COS , Proteínas Portadoras/química , Proteínas Portadoras/genética , Línea Celular , Clonación Molecular , ADN Complementario/genética , Endocitosis , Humanos , Cinética , Modelos Biológicos , Datos de Secuencia Molecular , Fosforilación , Estructura Terciaria de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Homología de Secuencia de Aminoácido , Transducción de Señal , Proteína Smad2 , Proteína smad3 , Proteína Smad4 , Factores de Transcripción/química , Factores de Transcripción/genética , Transfección , Proteínas de Transporte Vesicular
12.
Ann N Y Acad Sci ; 995: 1-10, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12814934

RESUMEN

Transforming growth factor-beta (TGF-beta), a key player in a large variety of physiological and disease processes, signals through transmembrane receptor serine/threonine kinases to activate novel signaling intermediates called Smad proteins, which then modulate transcription of target genes. We have utilized mice with a targeted deletion of Smad3, one of two homologous proteins involved in signaling from TGF-beta/activin, to investigate the function of this particular pathway in transducing such effects of TGF-beta. The dramatic results of the absence of Smad3 on parameters of healing of cutaneous wounds, such as reepithelialization and influx of inflammatory cells, as well as on fibrosis as modeled by radiation fibrosis of skin in mice, suggest that signaling flux through Smad3 is critical for chemotactic activity of TGF-beta, inhibitory effects of TGF-beta on keratinocyte proliferation and migration, and chemoattraction and elaboration of extracellular matrix by fibroblasts in fibrotic diseases. We recently identified a novel molecule, TLP for TRAP-1-like protein, which selectively interferes with Smad3 signaling, and are currently investigating whether levels of this protein might be altered in disease to change the relative flow of information from Smad2 and Smad3.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Transactivadores/fisiología , Factor de Crecimiento Transformador beta/fisiología , Animales , Daño del ADN , Proteínas de Unión al ADN/genética , Susceptibilidad a Enfermedades , Ratones , Ratones Noqueados , Modelos Biológicos , Transducción de Señal , Proteína Smad2 , Proteína smad3 , Transactivadores/genética , Cicatrización de Heridas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA