Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 386
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Oral Pathol Med ; 45(7): 494-9, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26671844

RESUMEN

BACKGROUND: Polymorphous low-grade adenocarcinoma (PLGA) is a rare malignant tumor that usually arises in the minor salivary glands. Growth factors are cell-secreted peptides that regulate biological processes such as growth, proliferation, and differentiation. In salivary gland tumors, immunoexpression of growth factors and their receptors is associated with cell proliferation, malignant transformation, and tumor invasion. This study analyzed the expression of growth factors and receptors in PLGA, in other to better understand the mechanisms involved in the process of neoplastic cell proliferation and tumor progression. METHODS: The expression of growth factors FGF-2, PDGF-A, PDGF-B and receptors FGFR-1, FGFR-2, PDGFR-α, and EGFR was analyzed in 24 PLGA samples in comparison with normal salivary glands, by immunohistochemistry. A semi-quantitative analysis determined cell positivity in all stained sections. Scores were assigned according to percentage of reactive cells: score 0 < 10%; score 1-10 to 25%; score 2-25% to 50%; score 3->50%. The level of significance was set at 5%. RESULTS: Most of the growth factors and receptors, apart from FGFR-2, were significantly reactive in PLGA. Comparing to salivary acini, all of the reactive growth factors and receptors were significantly stronger in PLGA. Comparing to salivary ducts, the expression of FGF-2, PDGF-B, FGFR-1, and EGFR was significantly stronger in the nuclei and/or cytoplasm of the neoplastic cells. CONCLUSIONS: The increased expression of the growth factors and receptors in the PLGA, compared to normal salivary glands, may be related to cell proliferation, somehow participating in the oncogenic process.


Asunto(s)
Adenocarcinoma/metabolismo , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Receptores de Factores de Crecimiento/biosíntesis , Neoplasias de las Glándulas Salivales/metabolismo , Glándulas Salivales Menores/metabolismo , Adenocarcinoma/patología , Diferenciación Celular/fisiología , Núcleo Celular/patología , Proliferación Celular/fisiología , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Humanos , Inmunohistoquímica , Neoplasias de las Glándulas Salivales/patología , Glándulas Salivales Menores/patología
2.
Nat Genet ; 24(4): 434-7, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10742113

RESUMEN

Analysis of classical mouse mutations has been useful in the identification and study of many genes. We previously mapped Sox18, encoding an SRY-related transcription factor, to distal mouse chromosome 2. This region contains a known mouse mutation, ragged (Ra), that affects the coat and vasculature. Here we have directly evaluated Sox18 as a candidate for Ra. We found that Sox18 is expressed in the developing vascular endothelium and hair follicles in mouse embryos. Furthermore, we found no recombination between Sox18 and Ra in an interspecific backcross segregating for the Ra phenotype. We found point mutations in Sox18 in two different Ra alleles that result in missense translation and premature truncation of the encoded protein. Fusion proteins containing these mutations lack the ability to activate transcription relative to wild-type controls in an in vitro assay. Our observations implicate mutations in Sox18 as the underlying cause of the Ra phenotype, and identify Sox18 as a critical gene for cardiovascular and hair follicle formation.


Asunto(s)
Anomalías Cardiovasculares/genética , Folículo Piloso/anomalías , Proteínas del Grupo de Alta Movilidad/genética , Mutación Puntual/genética , Factores de Transcripción/genética , Alelos , Animales , Anomalías Cardiovasculares/patología , Análisis Mutacional de ADN , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Regulación del Desarrollo de la Expresión Génica , Ligamiento Genético , Folículo Piloso/metabolismo , Folículo Piloso/patología , Proteínas del Grupo de Alta Movilidad/biosíntesis , Hibridación in Situ , Endogamia , Ratones , Ratones Mutantes , Neovascularización Fisiológica/genética , Fenotipo , ARN Mensajero/biosíntesis , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Proteínas Tirosina Quinasas Receptoras/deficiencia , Proteínas Tirosina Quinasas Receptoras/genética , Receptores de Factores de Crecimiento/biosíntesis , Receptores de Factores de Crecimiento/deficiencia , Receptores de Factores de Crecimiento/genética , Receptores de Factores de Crecimiento Endotelial Vascular , Recombinación Genética , Factores de Transcripción SOXF , Factores de Transcripción/biosíntesis , Activación Transcripcional
3.
J Neurosci ; 31(32): 11437-42, 2011 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-21832174

RESUMEN

Autism spectrum disorder (ASD) is a highly heritable, behaviorally defined, heterogeneous disorder of unknown pathogenesis. Several genetic risk genes have been identified, including the gene encoding the receptor tyrosine kinase MET, which regulates neuronal differentiation and growth. An ASD-associated polymorphism disrupts MET gene transcription, and there are reduced levels of MET protein expression in the mature temporal cortex of subjects with ASD. To address the possible neurodevelopmental contribution of MET to ASD pathogenesis, we examined the expression and transcriptional regulation of MET by a transcription factor, FOXP2, which is implicated in regulation of cognition and language, two functions altered in ASD. MET mRNA expression in the midgestation human fetal cerebral cortex is strikingly restricted, localized to portions of the temporal and occipital lobes. Within the cortical plate of the temporal lobe, the pattern of MET expression is highly complementary to the expression pattern of FOXP2, suggesting the latter may play a role in repression of gene expression. Consistent with this, MET and FOXP2 also are reciprocally expressed by differentiating normal human neuronal progenitor cells (NHNPs) in vitro, leading us to assess whether FOXP2 transcriptionally regulates MET. Indeed, FOXP2 binds directly to the 5' regulatory region of MET, and overexpression of FOXP2 results in transcriptional repression of MET. The expression of MET in restricted human neocortical regions, and its regulation in part by FOXP2, is consistent with genetic evidence for MET contributing to ASD risk.


Asunto(s)
Trastorno Autístico/genética , Trastornos del Conocimiento/genética , Trastornos del Conocimiento/metabolismo , Factores de Transcripción Forkhead/genética , Regulación del Desarrollo de la Expresión Génica , Proteínas Proto-Oncogénicas c-met/biosíntesis , Receptores de Factores de Crecimiento/biosíntesis , Regiones no Traducidas 5'/genética , Trastorno Autístico/metabolismo , Trastorno Autístico/patología , Trastornos Generalizados del Desarrollo Infantil/genética , Trastornos Generalizados del Desarrollo Infantil/metabolismo , Trastornos Generalizados del Desarrollo Infantil/patología , Trastornos del Conocimiento/patología , Femenino , Factores de Transcripción Forkhead/biosíntesis , Factores de Transcripción Forkhead/metabolismo , Humanos , Recién Nacido , Masculino , Proteínas Proto-Oncogénicas c-met/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-met/genética , Receptores de Factores de Crecimiento/antagonistas & inhibidores , Receptores de Factores de Crecimiento/genética , Factores de Riesgo
4.
Exp Cell Res ; 317(2): 131-42, 2011 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-20955695

RESUMEN

AIMS: Activin A and transforming growth factor-ß1 (TGF-ß1) belong to the same family of growth and differentiation factors that modulate vascular lesion formation in distinct ways, which we wish to understand mechanistically. METHODS AND RESULTS: We investigated the expression of cell-surface receptors and activation of Smads in human vascular smooth muscle cells (SMCs) and demonstrated that activin receptor-like kinase-1 (ALK-1), ALK-4, ALK-5 and endoglin are expressed in human SMCs. As expected, TGF-ß1 activates Smad1 and Smad2 in these cells. Interestingly, activin A also induces phosphorylation of both Smads, which has not been reported for Smad1 before. Transcriptome analyses of activin A and TGF-ß1 treated SMCs with subsequent Gene-Set Enrichment Analyses revealed that many downstream gene networks are induced by both factors. However, the effect of activin A on expression kinetics of individual genes is less pronounced than for TGF-ß1, which is explained by a more rapid dephosphorylation of Smads and p38-MAPK in response to activin A. Substantial differences in expression of fibronectin, alpha-V integrin and total extracellular collagen synthesis were observed. CONCLUSIONS: Genome-wide mRNA expression analyses clarify the distinct modulation of vascular lesion formation by activin A and TGF-ß1, most significantly because activin A is non-fibrotic.


Asunto(s)
Receptores de Activinas Tipo II/metabolismo , Activinas/farmacología , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/efectos de los fármacos , Fenotipo , Factor de Crecimiento Transformador beta/farmacología , Receptores de Activinas Tipo I/metabolismo , Activinas/genética , Activinas/metabolismo , Células Cultivadas , Endotelio Vascular/citología , Humanos , Miocitos del Músculo Liso/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Factores de Crecimiento/biosíntesis , Receptores de Factores de Crecimiento/genética , Receptores de Factores de Crecimiento/metabolismo , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacología , Vena Safena/citología , Proteína Smad2/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
5.
Nat Med ; 3(11): 1222-7, 1997 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9359696

RESUMEN

The importance of angiogenesis in malignant tumor growth has been interpreted mainly in terms of oxygen and nutrient supply. Here we demonstrate its fundamental role for tumor invasion of malignant human keratinocytes in surface transplants on nude mice. Distinct patterns of angiogenesis and vascular endothelial growth factor receptor-2 (VEGFR-2) expression allowed us to distinguish between benign and malignant cells. Functional inactivation of VEGF-R2 by a blocking antibody disrupted ongoing angiogenesis and prevented invasion of malignant cells, without reducing tumor cell proliferation. The reversion of a malignant into a benign phenotype by halting angiogenesis demonstrates a significant function of vascular endothelium for tumor invasion.


Asunto(s)
Invasividad Neoplásica , Neovascularización Patológica/complicaciones , Animales , Transformación Celular Neoplásica , Factores de Crecimiento Endotelial/biosíntesis , Técnica del Anticuerpo Fluorescente Indirecta , Humanos , Hibridación in Situ , Linfocinas/biosíntesis , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Receptores de Factores de Crecimiento/biosíntesis , Receptores Mitogénicos/biosíntesis , Receptores de Factores de Crecimiento Endotelial Vascular , Transfección , Células Tumorales Cultivadas , Regulación hacia Arriba , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
7.
Exp Neurol ; 342: 113740, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33971218

RESUMEN

In Parkinson's disease (PD), long-term administration of L-dopa often leads to L-dopa-induced dyskinesia (LID), a debilitating motor complication. The p75 neurotrophin receptor (p75NTR) is likely to play a critical role in the regulation of dendritic spine density and morphology and appears to be associated with neuroinflammation, which previously has been identified as a crucial mechanism in LID. While aberrant modifications of p75NTR in neurological diseases have been extensively documented, only a few studies report p75NTR dysfunction in PD, and no data are available in LID. Here, we explored the functional role of p75NTR in LID. In LID rats, we identified that p75NTR was significantly increased in the lesioned striatum. In 6-hydroxydopamine (6-OHDA)-hemilesioned rats, specific knockdown of striatal p75NTR levels achieved by viral vector injection into the striatum prevented the development of LID and increased striatal structural plasticity. By contrast, we found that in 6-OHDA-hemilesioned rats, striatal p75NTR overexpression exacerbated LID and facilitated striatal dendritic spine losses. Moreover, we observed that the immunomodulatory drug fingolimod attenuated LID without lessening the therapeutic efficacy of L-dopa and normalized p75NTR levels. Together, these data demonstrate for the first time that p75NTR plays a pivotal role in the development of LID and that p75NTR may act as a potential novel target for the management of LID.


Asunto(s)
Discinesia Inducida por Medicamentos/metabolismo , Levodopa/toxicidad , Proteínas del Tejido Nervioso/biosíntesis , Oxidopamina/toxicidad , Trastornos Parkinsonianos/metabolismo , Receptores de Factores de Crecimiento/biosíntesis , Animales , Masculino , Trastornos Parkinsonianos/inducido químicamente , Trastornos Parkinsonianos/tratamiento farmacológico , Ratas , Ratas Sprague-Dawley
8.
Biomed Pharmacother ; 140: 111765, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34058438

RESUMEN

Xenocoumacin (Xcn) 1 and 2 are the major antibiotics produced by the insect-pathogenic bacterium Xenorhabdus nematophila. Although the antimicrobial activity of Xcns has been explored, research regarding their action on mammalian cells is lacking. We aimed to investigate the action of Xcns in the context of inflammation and angiogenesis. We found that Xcns do not impair the viability of primary endothelial cells (ECs). Particularly Xcn2, but not Xcn1, inhibited the pro-inflammatory activation of ECs: Xcn2 diminished the interaction between ECs and leukocytes by downregulating cell adhesion molecule expression and blocked critical steps of the NF-κB activation pathway including the nuclear translocation of NF-κB p65 as well as the activation of inhibitor of κBα (IκBα) and IκB kinase ß (IKKß). Furthermore, the synthesis of pro-inflammatory mediators and enzymes, nitric oxide (NO) production and prostaglandin E2 (PGE2), inducible NO synthase (iNOS), and cyclooxygenase-2 (COX-2), was evaluated in leukocytes. The results showed that Xcns reduced viability, NO release, and iNOS expression in activated macrophages. Beyond these anti-inflammatory properties, Xcn2 effectively hindered pro-angiogenic processes in HUVECs, such as proliferation, undirected and chemotactic migration, sprouting, and network formation. Most importantly, we revealed that Xcn2 inhibits de novo protein synthesis in ECs. Consequently, protein levels of receptors that mediate the inflammatory and angiogenic signaling processes and that have a short half-live are reduced by Xcn2 treatment, thus explaining the observed pharmacological activities. Overall, our research highlights that Xcn2 exhibits significant pharmacological in vitro activity regarding inflammation and angiogenesis, which is worth to be further investigated preclinically.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Antiinflamatorios/farmacología , Benzopiranos/farmacología , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Adhesión Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Selectina E/genética , Selectina E/metabolismo , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Leucocitos/efectos de los fármacos , Leucocitos/fisiología , Ratones , FN-kappa B/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/metabolismo , Receptores de Factores de Crecimiento/biosíntesis , Receptores de Factores de Crecimiento/genética , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo
9.
J Exp Med ; 180(3): 1141-6, 1994 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-8064230

RESUMEN

Psoriatic skin is characterized by microvascular hyperpermeability and angioproliferation, but the mechanisms responsible are unknown. We report here that the hyperplastic epidermis of psoriatic skin expresses strikingly increased amounts of vascular permeability factor (VPF; vascular endothelial growth factor), a selective endothelial cell mitogen that enhances microvascular permeability. Moreover, two VPF receptors, kdr and flt-1, are overexpressed by papillary dermal microvascular endothelial cells. Transforming growth factor alpha (TGF-alpha), a cytokine that is also overexpressed in psoriatic epidermis, induced VPF gene expression by cultured epidermal keratinocytes. VPF secreted by TGF-alpha-stimulated keratinocytes was bioactive, as demonstrated by its mitogenic effect on dermal microvascular endothelial cells in vitro. Together, these findings suggest that TGF-alpha regulates VPF expression in psoriasis by an autocrine mechanism, leading to vascular hyperpermeability and angiogenesis. Similar mechanisms may operate in tumors and in healing skin wounds which also commonly express both VPF and TGF-alpha.


Asunto(s)
Factores de Crecimiento Endotelial/biosíntesis , Linfocinas/biosíntesis , Psoriasis/metabolismo , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Receptores de Factores de Crecimiento/biosíntesis , Secuencia de Bases , Células Cultivadas , Factores de Crecimiento Endotelial/genética , Humanos , Linfocinas/genética , Datos de Secuencia Molecular , ARN Mensajero/análisis , Proteínas Tirosina Quinasas Receptoras/genética , Receptores de Factores de Crecimiento/genética , Receptores de Factores de Crecimiento Endotelial Vascular , Factor de Crecimiento Transformador alfa/farmacología , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
10.
J Exp Med ; 182(5): 1369-76, 1995 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-7595207

RESUMEN

We investigated the expression and distribution of keratinocyte growth factor (KGF) (FGF-7) and its receptor (KGFR) during reepithelialization of human skin. KGF mRNA levels increased rapidly by 8-10-fold and remained elevated for several days. In contrast, KGFR transcript levels decreased early but were significantly elevated by 8-9 d. A KGF-immunoglobulin G fusion protein (KGF-HFc), which specifically and sensitively detects the KGFR, localized the receptor to differentiating keratinocytes of control epidermis, but revealed a striking decrease in receptor protein expression during the intermediate period of reepithelization. Suramin, which blocked KGF binding and stripped already bound KGF from its receptor, failed to unmask KGFRs in tissue sections from the intermediate phase of wound repair. The absence of KGFR protein despite increased KGFR transcript levels implies functional receptor downregulation in the presence of increased KGF. This temporal modulation of KGF and KGFRs provides strong evidence for the functional involvement of KGF in human skin reepithelialization.


Asunto(s)
Epidermis/fisiología , Factores de Crecimiento de Fibroblastos , Sustancias de Crecimiento/biosíntesis , Receptores de Factores de Crecimiento de Fibroblastos , Receptores de Factores de Crecimiento/biosíntesis , Cicatrización de Heridas/genética , Adulto , Diferenciación Celular , Regulación hacia Abajo , Epidermis/efectos de los fármacos , Factor 10 de Crecimiento de Fibroblastos , Factor 7 de Crecimiento de Fibroblastos , Regulación de la Expresión Génica/efectos de los fármacos , Genes de Inmunoglobulinas , Humanos , Persona de Mediana Edad , ARN Mensajero/biosíntesis , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos , Proteínas Recombinantes de Fusión/biosíntesis , Trasplante de Piel , Suramina/farmacología
11.
Breast Cancer Res Treat ; 122(1): 105-10, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19760033

RESUMEN

The levels of expression of the four receptors and eleven ligands composing the epidermal growth factor family were measured using immunohistochemical staining in one hundred cases of breast cancer. All of the family were expressed to some degree in some cases; however, individual cases showed a very wide range of expression of the family from essentially none to all the factors at high levels. The highest aggregate level of expression of a receptor was HER2 followed by HER1, then HER3, then HER4. The ligands (including two splice variants of the NRG1 and NRG2 genes) broadly fell into three groups, those with the highest aggregate expression were Epigen, Epiregulin, Neuregulin 1alpha, Neuregulin 2alpha, Neuregulin 2beta, Neuregulin 4 and TGFalpha, moderate expression was seen with EGF, Neuregulin 1beta and Neuregulin 3, and relatively low levels of expression were seen of HB-EGF, Betacellulin and Amphiregulin. Statistical analysis using Spearman's Rank Correlation showed a positive correlation of expression between each of the factors. Analysing the data using the Cox Proportional Hazards model showed that, in this dataset, the most powerful predictors of relapse free interval and overall survival were the combined measurement of only Epigen and Neuregulin 4.


Asunto(s)
Neoplasias de la Mama/genética , Regulación Neoplásica de la Expresión Génica , Familia de Multigenes , Proteínas de Neoplasias/genética , Receptores de Factores de Crecimiento/genética , Anfirregulina , Betacelulina , Neoplasias de la Mama/metabolismo , Supervivencia sin Enfermedad , Familia de Proteínas EGF , Factor de Crecimiento Epidérmico/biosíntesis , Factor de Crecimiento Epidérmico/genética , Epigen , Receptores ErbB/biosíntesis , Receptores ErbB/genética , Femenino , Perfilación de la Expresión Génica , Genes erbB , Genes erbB-1 , Genes erbB-2 , Glicoproteínas/biosíntesis , Glicoproteínas/genética , Humanos , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Péptidos y Proteínas de Señalización Intercelular/genética , Ligandos , Proteínas de Neoplasias/biosíntesis , Neurregulinas/biosíntesis , Neurregulinas/genética , Pronóstico , Modelos de Riesgos Proporcionales , Receptor ErbB-2/biosíntesis , Receptor ErbB-3/biosíntesis , Receptor ErbB-4 , Receptores de Factores de Crecimiento/biosíntesis , Factor de Crecimiento Transformador alfa/biosíntesis , Factor de Crecimiento Transformador alfa/genética
12.
Mod Pathol ; 23(10): 1379-92, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20639860

RESUMEN

It remains obscure in myxofibrosarcoma about the basis of tumorigenesis, progression, and metastasis. Chromosome 7 gains are common in some sarcomas, including myxofibrosarcoma, whereas the specific oncogenes are yet to be characterized. We performed an integrative study of MET gene at 7q31.2 to elucidate its implication in myxofibrosarcoma. Focused on candidate oncogenes on chromosome 7, 385K array comparative genomic hybridization was used to profile DNA copy number alterations of 12 samples. MET transcript was successfully quantified by real-time RT-PCR for 16 laser-microdissected tumors and two myxofibrosarcoma cell lines (NMFH-1, OH931). MET immunoexpression was assessable in 86 primary localized tumors with follow-up. To analyze endogenous MET expression and activation, NMFH-1 and OH931 cells, both with wild-type MET gene, were subjected to Western blotting and hepatocyte growth factor-treated NMFH-1 cells were evaluated for the kinetics of MET tyrosine phosphorylation. Non-random large-scale gains on 7q were detected in five cases, delineating three recurrent amplicons, 7q21.11-7q21.3, 7q22.1-22.3, and 7q31.1-7q32.3, in which the locus of MET displayed increased copy number, among others. MET mRNA was upregulated in OH931, NMFH-1, and nine tumors (56%), whereas neither gene dosage nor mRNA expression of MET was associated with clinicopathological factors. In contrast, MET protein overexpression, present in 67% of cases, was highly related to deep location (P=0.004), higher grades (P=0.001), and more advanced stages (P<0.001). Importantly, MET overexpression independently portended inferior metastasis-free survival (P=0.004) and overall survival (P=0.0221). Expressing activating phospho-MET at Tyr(1234)/Tyr(1235), OH931 cells had more abundant total MET than NMFH-1 cells, whereas the latter became promptly phosphorylated on stimulation of hepatocyte growth factor. In primary myxofibrosarcomas, MET overexpression, as a frequent event, is likely driven by 7q gains with mRNA upregulation, associated with important prognosticators, and independently predictive of worse outcomes, highlighting its possible causative function in tumor aggressiveness and potentiality as a therapeutic target.


Asunto(s)
Biomarcadores de Tumor/genética , Fibrosarcoma/genética , Proteínas Proto-Oncogénicas c-met/biosíntesis , Receptores de Factores de Crecimiento/biosíntesis , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Western Blotting , Cromosomas Humanos Par 7/genética , Hibridación Genómica Comparativa , Femenino , Fibrosarcoma/mortalidad , Fibrosarcoma/patología , Perfilación de la Expresión Génica , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Pronóstico , Proteínas Proto-Oncogénicas c-met/genética , Receptores de Factores de Crecimiento/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Regulación hacia Arriba , Adulto Joven
13.
Invest Clin ; 51(3): 369-80, 2010 Sep.
Artículo en Español | MEDLINE | ID: mdl-21305773

RESUMEN

The product of the proto-oncogene C-MET (the c-Met receptor) and its ligand, hepatocyte growth factor (HGF), have been implicated in the progression of gastric cancer. The aim of this study was to analyze the expression of c-Met receptor, HGF and proliferating cell nuclear antigen (PCNA) by the immunohistochemistry method of labeled streptavidin-biotin, as well as survival, and they were correlated with anatomopathological factors in stomach specimens of 40 patients, who underwent gastrectomy for gastric cancer in the Department of General Surgery, Hospital Central Universitario "Antonio María Pineda" in Barquisimeto, Venezuela, in 2001-2004. High expression of c-Met receptor and PCNA was observed in patients with advanced stages of gastric cancer (III and IV) compared with early stages (I and II) (p<0.01). There was also overexpression of the c-Met receptor in histologic variables with low degree of differentiation, deeper tumor invasion into the submucosa, liver metastases and it is reported a lower survival rate in patients with increased receptor expression (+++ and ++++) when compared with patients with the lowest expression (+ and ++) (p<0.01). The expression of HGF was constant in both, advanced and early groups. The c-Met receptor is associated with proliferation and cell migration in Venezuelan patients with gastric cancer and could be used as a prognostic factor in this pathology.


Asunto(s)
Proteínas Proto-Oncogénicas c-met/biosíntesis , Receptores de Factores de Crecimiento/biosíntesis , Neoplasias Gástricas/etiología , Neoplasias Gástricas/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Progresión de la Enfermedad , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-met/inmunología , Receptores de Factores de Crecimiento/inmunología , Neoplasias Gástricas/inmunología
14.
Toxicology ; 439: 152477, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32360609

RESUMEN

We previously reported that exposure during gestation and lactation to a low dose of glyphosate-based herbicide (GBH) reduced the area and perimeter of male offspring mammary gland at postnatal day 60 (PND60), whereas a higher dose increased the longitudinal growth of the gland. Here, our aim was to assess whether perinatal exposure to GBH exhibits endocrine disruptive action in male mammary gland at an early time point (pre-puberty), which could be related to the changes observed after puberty. We also wanted to explore whether an early evaluation of the male rat mammary gland is appropriate to assess exposure to potential endocrine disrupting chemicals (EDCs). Pregnant rats were orally exposed, through the diet, to vehicle (saline solution), 3.5 or 350 mg/kg/day of GBH from gestational day 9 until weaning. At PND21, the male offspring were euthanized, and mammary gland samples were collected. The histology and proliferation index of the mammary glands were evaluated, and the mRNA expression of estrogen (ESR1) and androgen (AR) receptors, cyclin D1 (Ccnd1), amphiregulin (Areg), insulin-like growth factor 1 (IGF1), epidermal growth factor receptor (EGFR) and IGF1 receptor (IGF1R) were assessed. Moreover, the phosphorylated-Erk1/2 (p-ERK1/2) protein expression was determined. No differences were observed in mammary epithelial structures and AR expression between experimental groups; however, the proliferation index was reduced in GBH3.5-exposed males. This result was associated with decreased ESR1, Ccnd1, Areg, IGF1, EGFR and IGF1R mRNA expressions, as well as reduced p-Erk1/2 protein expression in these animals. ESR1, Ccnd1, IGF1R and EGFR expressions were also reduced in GBH350-exposed males. In conclusion, the mammary gland development of pre-pubertal male rats is affected by perinatal exposure to GBH. Although further studies are still needed to understand the molecular mechanisms involved in GBH350 exposure, the present results may explain the alterations observed in mammary gland growth of post-pubertal males exposed to low doses of GBH. Our results also suggest that early evaluation of the male rat mammary gland is useful in assessing exposure to potential EDCs. However, analysis of EDCs effects at later time points should not be excluded.


Asunto(s)
Disruptores Endocrinos/toxicidad , Glicina/análogos & derivados , Herbicidas/toxicidad , Glándulas Mamarias Animales/crecimiento & desarrollo , Actinas/metabolismo , Animales , Femenino , Glicina/toxicidad , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Glándulas Mamarias Animales/efectos de los fármacos , Embarazo , Efectos Tardíos de la Exposición Prenatal/patología , Ratas , Ratas Wistar , Receptores de Estrógenos/efectos de los fármacos , Receptores de Estrógenos/genética , Receptores de Factores de Crecimiento/biosíntesis , Receptores de Esteroides/biosíntesis , Glifosato
15.
Int J Cancer ; 124(8): 1778-84, 2009 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-19117057

RESUMEN

We analyzed MET protein and copy number in NSCLC with or without EGFR mutations untreated with EGFR tyrosine kinase inhibitors (TKIs). MET copy number was examined in 28 NSCLC and 4 human bronchial epithelial cell lines (HBEC) and 100 primary tumors using quantitative real-time PCR. Positive results were confirmed by array comparative genomic hybridization and fluorescence in-situ hybridization. Total and phospho-MET protein expression was determined in 24 NSCLC and 2 HBEC cell lines using Western blot. EGFR mutations were examined for exon 19 deletions, T790M, and L858R. Knockdown of EGFR with siRNA was performed to examine the relation between EGFR and MET activation. High-level MET amplification was observed in 3 of 28 NSCLC cell lines and in 2 of 100 primary lung tumors that had not been treated with EGFR-TKIs. MET protein was highly expressed and phosphorylated in all the 3 cell lines with high MET amplification. In contrast, 6 NSCLC cell lines showed phospho-MET among 21 NSCLC cell lines without MET amplification (p = 0.042). Furthermore, those 6 cell lines harboring phospho-MET expression without MET amplification were all EGFR mutant (p = 0.0039). siRNA-mediated knockdown of EGFR abolished phospho-MET expression in examined 3 EGFR mutant cell lines of which MET gene copy number was not amplified. By contrast, phospho-MET expression in 2 cell lines with amplified MET gene was not down-regulated by knockdown of EGFR. Our results indicated that MET amplification was present in untreated NSCLC and EGFR mutation or MET amplification activated MET protein in NSCLC.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Receptores ErbB/genética , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Mutación , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/genética , Receptores de Factores de Crecimiento/biosíntesis , Receptores de Factores de Crecimiento/genética , Anciano , Bronquios/citología , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Células Epiteliales/citología , Receptores ErbB/antagonistas & inhibidores , Femenino , Amplificación de Genes , Humanos , Masculino , Persona de Mediana Edad , Proteínas Proto-Oncogénicas c-met , ARN Interferente Pequeño/metabolismo
16.
Am J Obstet Gynecol ; 200(4): 457.e1-5, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19200930

RESUMEN

OBJECTIVES: To evaluate the relationship of hormone (estrogen receptor alpha, estrogen receptor beta, progesterone receptor) and growth factor receptor (insulin-like growth factor receptor, human epidermal growth factor receptor 2) expression with disease progression in uterine carcinosarcoma. STUDY DESIGN: Immunohistochemistry was performed on tissue arrays using standard methodology. Differences between groups were evaluated by the Wilcoxon rank-sum test. Interactions between tumor stage and receptor expression were determined by linear trend analysis. RESULTS: Compared with normal endometrium, carcinosarcomas exhibited low estrogen receptor alpha and progesterone receptor expression (all P < .01), but overexpressed estrogen receptor beta (P = .02). Estrogen receptor beta expression increased in advanced stage disease (P = .02). Insulin-like growth factor receptor expression was lower in carcinosarcoma compared with normal endometrium (P = .01). Human epidermal growth factor receptor 2 expression was elevated and increased with disease progression (P < .01). CONCLUSION: In uterine carcinosarcoma, estrogen receptor beta expression is elevated and increases with disease progression, whereas estrogen receptor alpha and progesterone receptor are suppressed. Human epidermal growth factor receptor 2 expression is increased, whereas insulin-like growth factor receptor is lower than in normal endometrium. These data support a potential role for estrogen receptor beta in disease progression via crosstalk with human epidermal growth factor receptor 2.


Asunto(s)
Carcinosarcoma/metabolismo , Carcinosarcoma/patología , Receptor alfa de Estrógeno/biosíntesis , Receptor beta de Estrógeno/biosíntesis , Receptores de Factores de Crecimiento/biosíntesis , Receptores de Progesterona/biosíntesis , Neoplasias Uterinas/metabolismo , Neoplasias Uterinas/patología , Carcinosarcoma/química , Progresión de la Enfermedad , Receptor alfa de Estrógeno/análisis , Receptor beta de Estrógeno/análisis , Femenino , Humanos , Análisis por Micromatrices , Estadificación de Neoplasias , Proyectos Piloto , Receptores de Factores de Crecimiento/análisis , Receptores de Progesterona/análisis , Transducción de Señal , Neoplasias Uterinas/química
17.
Clin Cancer Res ; 14(16): 5033-42, 2008 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-18698021

RESUMEN

PURPOSE: New therapeutic targets for soft-tissue sarcoma (STS) treatment are critically needed. Midkine (MK), a multifunctional cytokine, is expressed during midgestation but is highly restricted in normal adult tissues. Renewed MK expression was shown in several malignancies where protumorigenic properties were described. We evaluated the expression and function of MK in STS. EXPERIMENTAL DESIGN: Immunohistochemistry, reverse transcription-PCR, and Western blotting (WB) evaluated MK expression in human STS tissues and cell lines. WB and flow cytometry analyzed MK receptor expression. Cell growth assays evaluated the effect of MK on STS cell growth, and WB assessed MK downstream signaling. MK knock-in and knockout experiments further evaluated MK function. The growth of parental versus MK-transfected human fibrosarcoma cells was studied in vivo. RESULTS: MK was found to be overexpressed in a variety of human STS histologies. Using a rhabdomyosarcoma (RMS) tissue microarray, cytoplasmic and nuclear MK was identified; nuclear MK expression was significantly increased in metastases. Similarly, several STS cell lines expressed and secreted MK; RMS cells exhibited nuclear MK. STS cells also expressed the MK receptors protein tyrosine phosphatase zeta and lipoprotein receptor-related protein. MK significantly enhanced STS cell growth potentially via the Src and extracellular signal-regulated kinase pathways. STS cells stably transfected with MK exhibited increased growth in vitro and in vivo. MK-expressing human STS xenografts showed increased tumor-associated vasculature. Furthermore, MK knockdown resulted in decreased STS cell growth, especially in RMS cells. CONCLUSION: MK enhances STS tumor growth; our results support further investigation of MK and its receptors as therapeutic targets for human STS.


Asunto(s)
Citocinas/metabolismo , Sarcoma/metabolismo , Sarcoma/patología , Neoplasias de los Tejidos Blandos/metabolismo , Neoplasias de los Tejidos Blandos/patología , Animales , Western Blotting , Proliferación Celular , Femenino , Citometría de Flujo , Humanos , Inmunohistoquímica , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/biosíntesis , Glicoproteínas de Membrana/biosíntesis , Ratones , Ratones Desnudos , Midkina , Proteínas Tirosina Fosfatasas/biosíntesis , Receptores de Factores de Crecimiento/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Análisis de Matrices Tisulares
18.
J Mol Med (Berl) ; 86(2): 161-9, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17891365

RESUMEN

The use of recombinant adenoviruses (Ad) to express secreted antagonists of growth factors represents a powerful strategy for studying physiologic functions of growth factor pathways in experimental animals. Indeed, a single adenoviral injection can produce characteristic high-level and persistent plasma expression of soluble receptor ectodomains or secreted protein antagonists, allowing highly stringent conditional inactivation of target pathways in vivo. In this review, we describe our experience using recombinant Ad to inactivate growth factor pathways in vivo and discuss their advantages and limitations. Using our studies on vascular endothelial growth factor and Wnt systems as examples, we further describe how recombinant Ad can unveil previously unknown physiological roles of signaling pathways. Finally, we discuss the potential physiological and therapeutic relevance of our findings.


Asunto(s)
Adenoviridae/genética , Técnicas de Transferencia de Gen , Vectores Genéticos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Receptores de Factores de Crecimiento/metabolismo , Transducción de Señal , Adenoviridae/metabolismo , Animales , ADN Recombinante , Eritropoyesis/genética , Terapia Genética , Humanos , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Péptidos y Proteínas de Señalización Intercelular/genética , Receptores de Factores de Crecimiento/biosíntesis , Receptores de Factores de Crecimiento/genética , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Proteínas Recombinantes/metabolismo , Transducción de Señal/genética , Factores de Tiempo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteínas Wnt/metabolismo
19.
Neuron ; 12(5): 1161-71, 1994 May.
Artículo en Inglés | MEDLINE | ID: mdl-7514427

RESUMEN

To determine whether neurotrophins act on functionally distinct populations of adult sensory neurons, the distributions of mRNAs for TrkA and tyrosine kinase-containing isoforms of TrkB and TrkC were determined in rat DRG neurons projecting to different peripheral targets. Whereas trkA was expressed by a very high percentage of visceral afferents, trkC was expressed frequently only in muscle afferents. Among cutaneous afferents, the size distributions for trkA- and trkC-positive cells showed little overlap. The percentages and size distributions of cells labeled for the trks argue strongly that almost all trkB-expressing cells must also express trkA or trkC. These results indicate that NGF and NT-3 act on functionally distinct populations of adult sensory neurons and suggest that a sizeable number of small DRG neurons may not respond to neurotrophins via a known Trk in the adult rat.


Asunto(s)
Ganglios Espinales/metabolismo , Expresión Génica , Neuronas Aferentes/metabolismo , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Receptores de Factores de Crecimiento/biosíntesis , Receptores de Factor de Crecimiento Nervioso/biosíntesis , Vías Aferentes/metabolismo , Envejecimiento/metabolismo , Animales , Transporte Axonal , Femenino , Ganglios Espinales/citología , Ganglios Espinales/crecimiento & desarrollo , Hibridación in Situ , Masculino , Neuronas Aferentes/citología , Proteínas Tirosina Quinasas/biosíntesis , ARN Mensajero/biosíntesis , Ratas , Receptor trkA , Receptor trkB , Receptor trkC , Piel/inervación
20.
Neuron ; 34(6): 945-60, 2002 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-12086642

RESUMEN

Neurogenesis proceeds throughout life in the higher vocal center (HVC) of the adult songbird neostriatum. Testosterone induces neuronal addition and endothelial division in HVC. We asked if testosterone-induced angiogenesis might contribute importantly to HVC neuronal recruitment. Testosterone upregulated both VEGF and its endothelial receptor, VEGF-R2/Quek1/KDR, in HVC. This yielded a burst in local HVC angiogenesis. FACS-isolated HVC endothelial cells produced BDNF in a testosterone-dependent manner. In vivo, HVC BDNF rose by the third week after testosterone, lagging by over a week the rise in VEGF and VEGF-R2. In situ hybridization revealed that much of this induced BDNF mRNA was endothelial. In vivo, both angiogenesis and neuronal addition to HVC were substantially diminished by inhibition of VEGF-R2 tyrosine kinase. These findings suggest a causal interaction between testosterone-induced angiogenesis and neurogenesis in the adult forebrain.


Asunto(s)
Encéfalo/irrigación sanguínea , Encéfalo/crecimiento & desarrollo , Canarios/fisiología , Endotelio Vascular/fisiología , Neovascularización Fisiológica/fisiología , Neuronas/citología , Animales , Encéfalo/citología , Encéfalo/efectos de los fármacos , Factor Neurotrófico Derivado del Encéfalo/biosíntesis , Factor Neurotrófico Derivado del Encéfalo/metabolismo , División Celular/efectos de los fármacos , División Celular/fisiología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Células Cultivadas , Factores de Crecimiento Endotelial/biosíntesis , Endotelio Vascular/citología , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Estrógenos/farmacología , Femenino , Linfocinas/biosíntesis , Masculino , Neovascularización Fisiológica/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/fisiología , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Receptores de Factores de Crecimiento/biosíntesis , Receptores de Factores de Crecimiento Endotelial Vascular , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Testosterona/farmacología , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA