Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Breast Cancer Res ; 21(1): 63, 2019 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-31101121

RESUMEN

BACKGROUND: HER2-driven breast cancer is correlated with poor prognosis, especially during its later stages. Numerous studies have shown the importance of the integrin α3ß1 during the initiation and progression of breast cancer; however, its role in this disease is complex and often opposite during different stages and in different types of tumors. In this study, we aim to elucidate the role of integrin α3ß1 in a genetically engineered mouse model of HER2-driven mammary tumorigenesis. METHODS: To investigate the role of α3ß1 in HER2-driven tumorigenesis in vivo, we generated a HER2-driven MMTV-cNeu mouse model of mammary tumorigenesis with targeted deletion of Itga3 (Itga3 KO mice). We have further used several established triple-negative and HER2-overexpressing human mammary carcinoma cell lines and generated ITGA3-knockout cells to investigate the role of α3ß1 in vitro. Invasion of cells was assessed using Matrigel- and Matrigel/collagen I-coated Transwell assays under static or interstitial fluid flow conditions. The role of α3ß1 in initial adhesion to laminin and collagen was assessed using adhesion assays and immunofluorescence. RESULTS: Tumor onset in mice was independent of the presence of α3ß1. In contrast, the depletion of α3ß1 reduced the survival of mice and increased tumor growth and vascularization. Furthermore, Itga3 KO mice were significantly more likely to develop lung metastases and had an increased metastatic burden compared to WT mice. In vitro, the deletion of ITGA3 caused a significant increase in the cellular invasion of HER2-overexpressing SKBR3, AU565, and BT474 cells, but not of triple-negative MDA-MB-231. This invasion suppressing function of α3ß1 in HER2-driven cells depended on the composition of the extracellular matrix and the interstitial fluid flow. CONCLUSION: Downregulation of α3ß1 in a HER2-driven mouse model and in HER2-overexpressing human mammary carcinoma cells promotes progression and invasiveness of tumors. The invasion-suppressive role of α3ß1 was not observed in triple-negative mammary carcinoma cells, illustrating the tumor type-specific and complex function of α3ß1 in breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Integrina alfa3beta1/deficiencia , Receptor ErbB-2/genética , Animales , Biomarcadores de Tumor , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/mortalidad , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Inmunofenotipificación , Estimación de Kaplan-Meier , Ratones , Ratones Noqueados , Metástasis de la Neoplasia , Receptor ErbB-2/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología
2.
Front Immunol ; 9: 1231, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29963041

RESUMEN

DC-SIGN is an antigen uptake receptor expressed on dendritic cells (DCs) with specificity for glycans present on a broad variety of pathogens and is capable of directing its cargo to MHC-I and MHC-II pathways for the induction of CD8+ and CD4+ T cell responses, respectively. Therefore, DC-SIGN is a very promising target for the delivery of antigen for anti-cancer vaccination. Although the endocytic route leading to MHC-II presentation is characterized to a large extent, the mechanisms controlling DC-SIGN targeted cross-presentation of exogenous peptides on MHC-I, are not completely resolved yet. In this paper, we used imaging flow cytometry and antigen-specific CD8+ T cells to investigate the intracellular fate of DC-SIGN and its cargo in human DCs. Our data demonstrates that immature DCs and toll-like receptor 4 (TLR4) stimulated DCs had similar internalization capacity and were both able to cross-present antigen targeted via DC-SIGN. Interestingly, simultaneous triggering of TLR4 and DC-SIGN on DCs resulted in the translocation of cargo to the cytosol, leading to proteasome-dependent processing and increased CD8+ T cell activation. Understanding the dynamics of DC-SIGN-mediated uptake and processing is essential for the design of optimal DC-SIGN-targeting vaccination strategies aimed at enhancing CD8+ T cell responses.


Asunto(s)
Presentación de Antígeno/inmunología , Antígenos/inmunología , Moléculas de Adhesión Celular/metabolismo , Antígenos de Histocompatibilidad Clase I/inmunología , Lectinas Tipo C/metabolismo , Receptores de Superficie Celular/metabolismo , Receptor Toll-Like 4/metabolismo , Biomarcadores , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Membrana Celular/metabolismo , Reactividad Cruzada , Endosomas/metabolismo , Humanos , Unión Proteica , Transporte de Proteínas
3.
Matrix Biol ; 57-58: 244-257, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28043890

RESUMEN

Laminins are a major constituent of the basement membranes of the kidney collecting system. Integrins, transmembrane receptors formed by non-covalently bound α and ß subunits, serve as laminin receptors, but their role in development and homeostasis of the kidney collecting system is poorly defined. Integrin α3ß1, one of the major laminin receptors, plays a minor role in kidney collecting system development, while the role of α6 containing integrins (α6ß1 and α6ß4), the other major laminin receptors, is unknown. Patients with mutations in α6 containing integrins not only develop epidermolysis bullosa, but also have abnormalities in the kidney collecting system. In this study, we show that selectively deleting the α6 or ß4 integrin subunits at the initiation of ureteric bud development in mice does not affect morphogenesis. However, the collecting system becomes dilated and dysmorphic as the mice age. The collecting system in both null genotypes was also highly susceptible to unilateral ureteric obstruction injury with evidence of excessive tubule dilatation and epithelial cell apoptosis. Mechanistically, integrin α6-null collecting duct cells are unable to withstand high mechanical force when adhered to laminin. Thus, we conclude that α6 integrins are important for maintaining the integrity of the kidney collecting system by enhancing tight adhesion of the epithelial cells to the basement membrane. These data give a mechanistic explanation for the association between kidney collecting system abnormalities in patients and epidermolysis bullosa.


Asunto(s)
Membrana Basal/metabolismo , Integrina alfa6beta1/genética , Integrina alfa6beta4/genética , Túbulos Renales Colectores/metabolismo , Laminina/genética , Obstrucción Ureteral/metabolismo , Animales , Apoptosis , Membrana Basal/patología , Adhesión Celular , Movimiento Celular , Proliferación Celular , Células Epiteliales/metabolismo , Células Epiteliales/patología , Fibrosis , Regulación de la Expresión Génica , Humanos , Integrina alfa6beta1/deficiencia , Integrina alfa6beta4/deficiencia , Túbulos Renales Colectores/patología , Laminina/metabolismo , Ratones , Ratones Noqueados , Unión Proteica , Transducción de Señal , Uréter/cirugía , Obstrucción Ureteral/patología , Obstrucción Ureteral/cirugía
4.
Oncotarget ; 8(8): 13730-13746, 2017 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-28099149

RESUMEN

Metabolic reprogramming is a very heterogeneous phenomenon in cancer. It mostly consists on increased glycolysis, lactic acid formation and extracellular acidification. These events have been associated to increased activity of the hypoxia inducible factor, HIF-1α. This study aimed at defining the metabolic program activated by HIF-1α in oropharyngeal squamous cell carcinomas (SCC) and assessing its clinical impact. Global gene/miRNA expression was analyzed in SCC-derived cells exposed to hypoxia. Expression of HIF-1α, the carbonic anhydrase CAIX, and the lactate/H+ transporters MCT1 and MCT4 were analyzed by immunohistochemistry in 246 SCCs. Cell-based analysis revealed that HIF-1α-driven metabolic program includes over-expression of glycolytic enzymes and the microRNA miR-210 coupled to down-regulation of its target, the iron-sulfur cluster assembly protein, ISCU. pH-regulator program entailed over-expression of CAIX, but not MCT1 or MCT4. Accordingly, significant overlapping exists between over-expression of HIF-1α and CAIX, but not HIF-1α and MCT1 or MCT4, in tumor cells. Increased miR-210 and concomitant decreased ISCU RNA levels were found in ~40% of tumors and this was significantly associated with HIF-1α and CAIX, but not MCT1 or MCT4, over-expression. HIF-1α and/or CAIX over-expression was associated with high recurrence rate and low overall survival of surgically treated patients. By contrast, clinically significant correlations were not found in tumors with MCT1 or MCT4 over-expression. This is the first study that provides in vivo evidences of coordinated activation of HIF-1α, CAIX, miR-210 and ISCU in carcinoma and association with poor prognosis, a finding with important implications for the development of metabolic-targeting therapies against hypoxia.


Asunto(s)
Anhidrasa Carbónica IX/metabolismo , Carcinoma de Células Escamosas/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Proteínas Hierro-Azufre/metabolismo , MicroARNs/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Proteínas Musculares/metabolismo , Neoplasias Orofaríngeas/metabolismo , Simportadores/metabolismo , Carcinoma de Células Escamosas/genética , Línea Celular Tumoral , Femenino , Neoplasias de Cabeza y Cuello/genética , Humanos , Masculino , Persona de Mediana Edad , Neoplasias Orofaríngeas/genética , Transducción de Señal , Carcinoma de Células Escamosas de Cabeza y Cuello , Regulación hacia Arriba
5.
J Cell Sci ; 128(20): 3714-9, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26330528

RESUMEN

Hemidesmosomes have been extensively studied with immunofluorescence microscopy, but owing to its limited resolution, the precise organization of hemidesmosomes remains poorly understood. We studied hemidesmosome organization in cultured keratinocytes with two- and three-color super-resolution microscopy. We observed that, in the cell periphery, nascent hemidesmosomes are associated with individual keratin filaments and that ß4 integrin (also known as ITGB4) is distributed along, rather than under, keratin filaments. By applying innovative methods to quantify molecular distances, we demonstrate that the hemidesmosomal plaque protein plectin interacts simultaneously and asymmetrically with ß4 integrin and keratin. Furthermore, we show that BP180 (BPAG2, also known as collagen XVII) and BP230 (BPAG1e, an epithelial splice variant of dystonin) are characteristically arranged within hemidesmosomes with BP180 surrounding a central core of BP230 molecules. In skin cross-sections, hemidesmosomes of variable sizes could be distinguished with BP230 and plectin occupying a position in between ß4 integrin and BP180, and the intermediate filament system. In conclusion, our data provide a detailed view of the molecular architecture of hemidesmosomes in cultured keratinocytes and skin.


Asunto(s)
Autoantígenos/metabolismo , Proteínas Portadoras/metabolismo , Proteínas del Citoesqueleto/metabolismo , Hemidesmosomas/metabolismo , Integrina beta4/metabolismo , Queratinocitos/metabolismo , Queratinas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Colágenos no Fibrilares/metabolismo , Piel/metabolismo , Autoantígenos/genética , Proteínas Portadoras/genética , Proteínas del Citoesqueleto/genética , Distonina , Hemidesmosomas/genética , Hemidesmosomas/ultraestructura , Humanos , Integrina beta4/genética , Queratinocitos/ultraestructura , Queratinas/genética , Microscopía Fluorescente , Proteínas del Tejido Nervioso/genética , Colágenos no Fibrilares/genética , Piel/ultraestructura , Colágeno Tipo XVII
6.
Mol Biol Cell ; 26(13): 2402-17, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25971800

RESUMEN

Epidermolysis bullosa simplex associated with late-onset muscular dystrophy (EBS-MD) is an autosomal recessive disorder resulting from mutations in the plectin gene. The majority of these mutations occur within the large exon 31 encoding the central rod domain and leave the production of a low-level rodless plectin splice variant unaffected. To investigate the function of the rod domain, we generated rodless plectin mice through conditional deletion of exon 31. Rodless plectin mice develop normally without signs of skin blistering or muscular dystrophy. Plectin localization and hemidesmosome organization are unaffected in rodless plectin mice. However, superresolution microscopy revealed a closer juxtaposition of the C-terminus of plectin to the integrin ß4 subunit in rodless plectin keratinocytes. Wound healing occurred slightly faster in rodless plectin mice than in wild-type mice, and keratinocytes migration was increased in the absence of the rod domain. The faster migration of rodless plectin keratinocytes is not due to altered biochemical properties because, like full-length plectin, rodless plectin is a dimeric protein. Our data demonstrate that rodless plectin can functionally compensate for the loss of full-length plectin in mice. Thus the low expression level of plectin rather than the absence of the rod domain dictates the development of EBS-MD.


Asunto(s)
Epidermólisis Ampollosa Simple/metabolismo , Distrofia Muscular de Cinturas/metabolismo , Plectina/metabolismo , Animales , Citoesqueleto/metabolismo , Citoesqueleto/patología , Epidermólisis Ampollosa Simple/genética , Epidermólisis Ampollosa Simple/patología , Femenino , Hemidesmosomas/metabolismo , Integrina beta4/metabolismo , Queratinocitos/metabolismo , Queratinocitos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Distrofia Muscular de Cinturas/genética , Distrofia Muscular de Cinturas/patología , Plectina/genética , Isoformas de Proteínas , Estructura Terciaria de Proteína
7.
Head Neck ; 37(8): 1150-62, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24798801

RESUMEN

BACKGROUND: We previously showed that activation of epidermal growth factor receptor (EGFR) induces hypoxia inducible factor-1α (HIF-1α) in head and neck squamous cell carcinoma (HNSCC) cells. In this study, we have furthered this by investigating the mechanism of HIF-1α activation by epidermal growth factor (EGF) and its association with the sensitivity to gefitinib. METHODS: EGFR/HIF-1α signaling was tested by immunoblot, polymerase chain reaction (PCR), cell proliferation, and apoptosis assays. RESULTS: HIF-1α accumulated in cells overexpressing EGF and phosphorylated epidermal growth factor receptor (pEGFR), phosphatidylinositol-3-kinase (pPI3K), and mitogen-activated protein kinase (pMAPK). EGF-induced expression of HIF-1α and its targets, vascular endothelial growth factor (VEGF) and BNIP3, were blocked by gefitinib and PI3K-inhibitors and MAPK-inhibitors. HIF-1α-siRNAs abrogated EGF-induced BNIP3 but not VEGF expression. Gefitinib inhibited cell proliferation and induced apoptosis more strongly in cells with constitutively active EGFR/HIF-1α signaling than in cells lacking activation of these pathways. HIF-1α-siRNA treatment reduced sensitivity to gefitinib. CONCLUSION: The search for molecular predictors of sensitivity to gefitinib in HNSCC should be extended to the activation status of EGFR-downstream pathways, phosphorylated protein kinase B, pMAPK, and HIF-1α.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma de Células Escamosas/metabolismo , Receptores ErbB/metabolismo , Neoplasias de Cabeza y Cuello/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Antineoplásicos/farmacología , Apoptosis , Carcinoma de Células Escamosas/tratamiento farmacológico , Línea Celular Tumoral , Proliferación Celular , Receptores ErbB/antagonistas & inhibidores , Gefitinib , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Humanos , Immunoblotting , Técnicas In Vitro , Reacción en Cadena de la Polimerasa , Valor Predictivo de las Pruebas , Quinazolinas/farmacología , Sensibilidad y Especificidad , Transducción de Señal
8.
J Invest Dermatol ; 134(1): 221-228, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23792458

RESUMEN

Altered expression of the tetraspanin CD151 is associated with skin tumorigenesis; however, whether CD151 is causally involved in the tumorigenic process is not known. To evaluate its role in tumor formation, we subjected epidermis-specific Cd151 knockout mice to chemical skin carcinogenesis. Mice lacking epidermal Cd151 developed fewer and smaller tumors than wild-type mice after treatment with 7,12-dimethylbenzanthracene (DMBA)/12-O-tetradecanoylphorbol-13-acetate (TPA). Furthermore, Cd151-null epidermis showed a reduced hyperproliferative response to short-term treatment with TPA as compared with wild-type skin, whereas epidermal turnover was increased. Tumors were formed in equal numbers after DMBA-only treatment. We suggest that DMBA-initiated keratinocytes lacking Cd151 leave their niches in the epidermis and hair follicles in response to TPA treatment and subsequently are lost by differentiation. Because genetic ablation of Itga3 also reduced skin tumor formation, we tested whether reduced expression of α3 could further suppress tumor formation in epidermis-specific Cd151 knockout mice. Although DMBA/TPA-induced formation of skin tumors was similar in compound heterozygotes for Cd151 and Itga3 to that in wild-type mice, heterozygosity for Itga3 on a Cd151-null background diminished tumorigenesis, suggesting genetic interaction between the two genes. We thus identify CD151 as a critical factor in TPA-dependent skin carcinogenesis.


Asunto(s)
Queratinocitos/patología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Tetraspanina 24/genética , Animales , Carcinógenos/farmacología , Línea Celular , Epidermis/patología , Femenino , Eliminación de Gen , Predisposición Genética a la Enfermedad/genética , Integrina alfa3/genética , Masculino , Ratones , Ratones Noqueados , Estadificación de Neoplasias , Neoplasias Cutáneas/inducido químicamente , Acetato de Tetradecanoilforbol/farmacología
9.
Mol Biol Cell ; 24(15): 2454-66, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23761073

RESUMEN

Nesprin-3 is a nuclear envelope protein that connects the nucleus to intermediate filaments by interacting with plectin. To investigate the role of nesprin-3 in the perinuclear localization of plectin, we generated nesprin-3-knockout mice and examined the effects of nesprin-3 deficiency in different cell types and tissues. Nesprin-3 and plectin are coexpressed in a variety of tissues, including peripheral nerve and muscle. The expression level of nesprin-3 in skeletal muscle is very low and decreases during myoblast differentiation in vitro. Of interest, plectin was concentrated at the nuclear envelope in only a few cell types. This was most prominent in Sertoli cells of the testis, in which nesprin-3 is required for the localization of both plectin and vimentin at the nuclear perimeter. Testicular morphology and the position of the nucleus in Sertoli cells were normal, however, in the nesprin-3-knockout mice and the mice were fertile. Furthermore, nesprin-3 was not required for the polarization and migration of mouse embryonic fibroblasts. Thus, although nesprin-3 is critical for the localization of plectin to the nuclear perimeter of Sertoli cells, the resulting link between the nuclear envelope and the intermediate filament system seems to be dispensable for normal testicular morphology and spermatogenesis.


Asunto(s)
Proteínas de la Membrana/fisiología , Membrana Nuclear/metabolismo , Proteínas Nucleares/fisiología , Plectina/metabolismo , Células de Sertoli/fisiología , Espermatogénesis , Vimentina/metabolismo , Animales , Línea Celular , Movimiento Celular , Polaridad Celular , Células Madre Embrionarias/metabolismo , Femenino , Fibroblastos/fisiología , Hígado/metabolismo , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Desarrollo de Músculos , Mioblastos/fisiología , Especificidad de Órganos , Transporte de Proteínas , Testículo/citología
10.
J Invest Dermatol ; 133(9): 2180-90, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23549420

RESUMEN

From a forward genetic screen for epidermal defects in zebrafish, we identified a loss-of-function mutation in Kindlin-1, an essential regulator of integrin function. The mutation generates a premature stop codon, deleting the integrin-binding site. The mutant zebrafish develops cell-matrix and cell-cell adhesion defects in the basal epidermis leading to progressive fin rupturing, and was therefore designated rupturing-of-fins (rof). Similar defects were observed in the epidermis of Kindler syndrome patients, carrying a loss-of-function mutation in kindlin-1. Mutational analysis and rescue experiments in zebrafish revealed that residues K610, W612, and I647 in the F3 domain are essential for Kindlin-1 function in vivo, and that Kindlin-2 can functionally compensate for the loss of Kindlin-1. The fin phenotype of rof/kindlin-1 mutants resembles that of badfin mutants, carrying a mutation in integrin α3. We show here that this mutation impairs the biosynthesis of integrin α3ß1 and causes cell-matrix and cell-cell defects in vivo. Whereas both Integrin-linked kinase (Ilk) and Kindlin-1 cooperate with Integrin α3ß1 to resist trauma-induced epidermal defects, Kindlin-1 and Ilk, surprisingly, do not act synergistically but in parallel. Thus, the rof/kindlin-1 mutant zebrafish provides a unique model system to study epidermal adhesion mechanisms in vivo.


Asunto(s)
Aletas de Animales/fisiología , Adhesión Celular/fisiología , Células Epidérmicas , Epidermólisis Ampollosa/fisiopatología , Proteínas de la Membrana/genética , Proteínas de Neoplasias/genética , Pez Cebra/genética , Aletas de Animales/lesiones , Aletas de Animales/patología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Epidermis/fisiología , Epidermólisis Ampollosa/genética , Epidermólisis Ampollosa/patología , Enfermedades de los Peces/genética , Enfermedades de los Peces/patología , Enfermedades de los Peces/fisiopatología , Humanos , Integrina alfa3beta1/genética , Integrina alfa3beta1/fisiología , Proteínas de la Membrana/fisiología , Mutación , Proteínas de Neoplasias/fisiología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/fisiología
11.
BMC Cancer ; 13: 116, 2013 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-23497198

RESUMEN

BACKGROUND: Cytogenetic and gene expression analyses in head and neck squamous cell carcinomas (HNSCC) have allowed identification of genomic aberrations that may contribute to cancer pathophysiology. Nevertheless, the molecular consequences of numerous genetic alterations still remain unclear. METHODS: To identify novel genes implicated in HNSCC pathogenesis, we analyzed the genomic alterations present in five HNSCC-derived cell lines by array CGH, and compared high level focal gene amplifications with gene expression levels to identify genes whose expression is directly impacted by these genetic events. Next, we knocked down TRPC6, one of the most highly amplified and over-expressed genes, to characterize the biological roles of TRPC6 in carcinogenesis. Finally, real time PCR was performed to determine TRPC6 gene dosage and mRNA levels in normal mucosa and human HNSCC tissues. RESULTS: The data showed that the HNSCC-derived cell lines carry most of the recurrent genomic abnormalities previously described in primary tumors. High-level genomic amplifications were found at four chromosomal sites (11q21-q22.2, 18p11.31-p11.21, 19p13.2-p13.13, and 21q11) with associated gene expression changes in selective candidate genes suggesting that they may play an important role in the malignant behavior of HNSCC. One of the most dramatic alterations of gene transcription involved the TRPC6 gene (located at 11q21-q22.2) which has been recently implicated in tumour invasiveness. siRNA-induced knockdown of TRPC6 expression in HNSCC-derived cells dramatically inhibited HNSCC-cell invasion but did not significantly alter cell proliferation. Importantly, amplification and concomitant overexpression of TRPC6 was also found in HNSCC tumour samples. CONCLUSIONS: Altogether, these data show that TRPC6 is likely to be a target for 11q21-22.2 amplification that confers enhanced invasive behavior to HNSCC cells. Therefore, TRPC6 may be a promising therapeutic target in the treatment of HNSCC.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Carcinoma de Células Escamosas/genética , Cromosomas Humanos Par 11/genética , Neoplasias de Cabeza y Cuello/genética , Canales Catiónicos TRPC/genética , Biomarcadores de Tumor/genética , Carcinoma de Células Escamosas/metabolismo , Línea Celular Tumoral , Análisis Mutacional de ADN , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Neoplasias de Cabeza y Cuello/metabolismo , Humanos , Invasividad Neoplásica/genética , Carcinoma de Células Escamosas de Cabeza y Cuello , Canales Catiónicos TRPC/metabolismo , Canal Catiónico TRPC6
12.
Proc Natl Acad Sci U S A ; 109(52): 21468-73, 2012 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-23236172

RESUMEN

Progression through the various stages of skin tumorigenesis is correlated with an altered expression of the integrin α3ß1, suggesting that it plays an important role in the tumorigenic process. Using epidermis-specific Itga3 KO mice subjected to the 7,12-dimethylbenzanthracene (DMBA)/12-O-tetradecanoylphorbol-13-acetate two-stage skin carcinogenesis protocol, we demonstrate that efficient tumor development is critically dependent on the presence of α3ß1. In the absence of α3ß1, tumor initiation is dramatically decreased because of increased epidermal turnover, leading to a loss of DMBA-initiated label-retaining keratinocytes. Lineage tracing revealed emigration of α3-deficient keratinocytes residing in the bulge of the hair follicle toward the interfollicular epidermis. Furthermore, tumor growth and cell proliferation were strongly reduced in mice with an epidermis-specific deletion of Itga3. However, the rate of progression of α3ß1-null squamous cell carcinomas to undifferentiated, invasive carcinomas was increased. Therefore, α3ß1 critically affects skin carcinogenesis with opposing effects early and late in tumorigenesis.


Asunto(s)
Ciclo Celular , Transformación Celular Neoplásica/patología , Epidermis/metabolismo , Epidermis/patología , Integrina alfa3/metabolismo , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/prevención & control , 9,10-Dimetil-1,2-benzantraceno , Animales , Adhesión Celular , Diferenciación Celular , Linaje de la Célula , Movimiento Celular , Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Progresión de la Enfermedad , Folículo Piloso/metabolismo , Folículo Piloso/patología , Queratina-15/metabolismo , Queratinocitos/metabolismo , Queratinocitos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias Cutáneas/metabolismo , Coloración y Etiquetado
13.
J Clin Endocrinol Metab ; 97(11): E2194-200, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22977270

RESUMEN

BACKGROUND: Head and neck paragangliomas (HNPGLs) are rare tumors associated with the parasympathetic nervous system. Most are sporadic, but about one third result from germline mutations in succinate dehydrogenase (SDH) genes (SDHB, SDHC, SDHD, SDHA, or SDHAF2). Although a molecular connection between SDH dysfunction and tumor development is still unclear, the most accepted hypothesis proposes a central role of the pseudohypoxic pathway. SDH dysfunction induces abnormal stabilization of the hypoxia-inducible factors (HIFs) that regulate target genes involved in proliferation, apoptosis, angiogenesis, and metabolism. The involvement of these pathways in the development of sporadic HNPGLs is presently unknown. OBJECTIVE: To get some insights into the hypoxic/pseudohypoxic molecular basis of HNPGLs, we attempted to define the gene, microRNA (miRNA), and HIF-1α expression patterns that distinguish tumors from normal paraganglia tissue. DESIGN: Genome microarray and TaqMan low-density arrays were used to analyze gene and miRNA expression, respectively, in 17 HNPGL tumor tissues and three normal human carotid bodies. Twelve HNPGLs were used for validation of data. HIF-1α, SDHB, and iron-sulfur cluster scaffold protein (ISCU) protein expression was analyzed by immunohistochemistry. RESULTS: We found activation of a canonical HIF-1α-related gene expression signaling only in a subset of HNPGLs from patients that did not harbor germline or somatic SDH mutations. The pseudohypoxic signature consisted in the overexpression of both HIF-1α-target genes and the HIF-1α-inducible miRNA, miR-210, and down-regulation of the miR-210 target gene, ISCU1/2. A decreased level of the iron-sulfur-containing protein SDHB was found by immunohistochemical analysis performed in two of these tumors. CONCLUSIONS: Collectively, this study unveiled a putative signaling axis of HIF-1α/miRNA-210/ISCU in a subset of HNPGLs that could have an impact on SDHB protein stability by a mechanism independent of SDH mutations, thus providing a foundation to better understand the functional interplay between HIF, miR-210, and mitochondria and its relevance in the pathogenesis of HNPGLs.


Asunto(s)
Neoplasias de Cabeza y Cuello/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Proteínas Hierro-Azufre/genética , MicroARNs/genética , Paraganglioma/genética , Succinato Deshidrogenasa/genética , Adulto , Anciano , Femenino , Neoplasias de Cabeza y Cuello/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Proteínas Hierro-Azufre/metabolismo , Masculino , MicroARNs/metabolismo , Persona de Mediana Edad , Paraganglioma/metabolismo , Transducción de Señal/genética
14.
Genes Chromosomes Cancer ; 48(5): 441-54, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19235921

RESUMEN

The HIF-1alpha protein plays a key role in the cellular response to hypoxia via transcriptional regulation of genes involved in erythropoiesis, angiogenesis, and metabolism. Overexpression of HIF-1alpha is commonly found in solid tumors in significant association with increased patient mortality and resistance to therapy. The predominant mode of HIF-1alpha regulation by hypoxia occurs at the level of protein stability. In addition to hypoxia, HIF-1alpha protein stability and synthesis is regulated by nonhypoxic signals such as inactivation of tumor suppressors and activation of oncogenes. Here, we show that an increase in gene dosage may contribute to HIF-1alpha mRNA and protein overexpression in a nonhypoxic environment in head and neck squamous cell carcinomas (HNSCC). Increased HIF-1alpha gene dosage was found in one out of five HNSCC-derived cell lines and three out of 27 HNSCC primary tumors. Significantly, increased gene dosage in those samples was associated with high HIF-1alpha mRNA and protein levels. Normoxic overexpression of HIF-1alpha protein in HNSCC-derived cell lines was also paralleled by higher expression levels of HIF-1alpha target genes. Array CGH analysis confirmed the copy number increase of HIF-1alpha gene and revealed that the gene is contained within a region of amplification at 14q23-q24.2 both in the cell line and primary tumors. In addition, FISH analysis revealed the presence of 11-13 copies on a tetraploid background in SCC2 cells. These data suggest that increased HIF-1alpha gene dosage is a mechanism of HIF-1alpha protein overexpression in HNSCC that possibly prepares the cells for a higher activity in an intratumoral hypoxic environment.


Asunto(s)
Carcinoma de Células Escamosas/genética , Dosificación de Gen , Neoplasias de Cabeza y Cuello/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Carcinoma de Células Escamosas/metabolismo , Hipoxia de la Célula , Línea Celular Tumoral , Hibridación Genómica Comparativa , Receptores ErbB/metabolismo , Expresión Génica , Neoplasias de Cabeza y Cuello/metabolismo , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inmunohistoquímica , Hibridación Fluorescente in Situ , Análisis de Secuencia por Matrices de Oligonucleótidos , Transducción de Señal
15.
Acta Otorrinolaringol Esp ; 58(8): 347-51, 2007 Oct.
Artículo en Español | MEDLINE | ID: mdl-17949661

RESUMEN

INTRODUCTION AND OBJECTIVES: FAK expression is frequently elevated in human cancers, including head and neck squamous cell carcinomas. However, the regulation of FAK expression is poorly understood. The aim of this study is to establish the possible role of the p53 tumour suppressor gene in the regulation of FAK expression in squamous cell carcinomas of the larynx. MATERIAL AND METHOD: The expression of FAK and p53 proteins were studied using immunohistochemistry in 102 samples from surgically-treated squamous cell carcinomas of the supraglottic larynx (with postoperative radiotherapy in 49 cases). RESULTS: All the cases showed some degree of FAK expression, which was moderate to intense in 60 cases (59 %). Also 60 cases showed positive p53 expression (>10 % of tumour cells with nuclear staining). No relationship was observed between p53 positivity and FAK expression (P= .54). In addition, none of these proteins presented association with the prognosis of the patients. CONCLUSIONS: Our results suggests that p53 activity does not correlate with FAK expression in squamous cell carcinomas of the larynx.


Asunto(s)
Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Quinasa 1 de Adhesión Focal/genética , Genes p53 , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/patología , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Células Escamosas/cirugía , Femenino , Humanos , Neoplasias Laríngeas/cirugía , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Resultado del Tratamiento
16.
Arch Otolaryngol Head Neck Surg ; 133(2): 145-50, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17309983

RESUMEN

OBJECTIVE: To explore the value of E-cadherin and focal adhesion kinase (FAK) expression in the prediction of cervical lymph node metastases in squamous cell carcinoma of the supraglottic larynx. DESIGN: Immunohistochemical analysis of retrospectively selected cases. Patients The study population was composed of 95 previously untreated men with squamous cell carcinoma of the supraglottic larynx. Intervention All the patients underwent surgical resection of the tumor and bilateral neck dissection. MAIN OUTCOME MEASURES: E-cadherin and FAK expression in relation to nodal metastases. RESULTS: Decreased E-cadherin expression was correlated with the presence of nodal metastases (P = .006). The combination of E-cadherin and FAK expression resulted in a superior accuracy in assessing nodal metastasis (P = .001). Histological grade was also associated with nodal metastases (P = .005). Multivariate analysis confirmed that these parameters were independent predictors of nodal metastases. In addition, the cases with decreased E-cadherin and increased FAK expression presented a significantly reduced disease-specific survival (P = .005). CONCLUSION: The combination of the expression of E-cadherin and FAK could increase our ability to identify patients with clinically negative lymph nodes who are at considerable risk for occult metastases.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Cadherinas/metabolismo , Carcinoma de Células Escamosas/metabolismo , Quinasa 1 de Adhesión Focal/metabolismo , Neoplasias Laríngeas/metabolismo , Carcinoma de Células Escamosas/mortalidad , Carcinoma de Células Escamosas/secundario , Humanos , Inmunohistoquímica , Neoplasias Laríngeas/mortalidad , Neoplasias Laríngeas/patología , Modelos Logísticos , Metástasis Linfática/diagnóstico , Masculino , Análisis Multivariante , Recurrencia Local de Neoplasia , Estudios Retrospectivos , Tasa de Supervivencia
17.
Clin Cancer Res ; 12(11 Pt 1): 3272-9, 2006 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-16740747

RESUMEN

UNLABELLED: The development of human malignancies can involve the aberrant regulation of intracellular signal transduction pathways that regulate cell-extracellular matrix interactions. PURPOSE: In the current study, we aimed to evaluate focal adhesion kinase (FAK) at both genetic and protein expression levels in head and neck squamous cell carcinomas (HNSCC) and to explore the prognostic significance of FAK. EXPERIMENTAL DESIGN: A total of 211 tissue specimens, including 147 primary tumors, 56 lymph node metastases, 3 benign hyperplasias, and 5 dysplasias, were analyzed using immunohistochemistry. The fak gene dosage was determined in 33 tumors. Correlations among DNA, protein, and clinicopathologic variables were analyzed. RESULTS: FAK protein was overexpressed in HNSCCs compared with corresponding normal mucosa. High expression levels were found in 62% of the samples. Positive immunostaining was also detected in benign hyperplasias and preinvasive dysplastic lesions. All lymph node metastases examined showed FAK overexpression, with significant correlation with the expression in matched primary tumor. DNA copy number ratios for fak were higher in 39% of the tumors compared with normal mucosa. However, elevated FAK expression did not correlate with gains on DNA level, and not all cases with an amplification of the fak gene displayed protein overexpression. Similar data were obtained in five HNSCC-derived cell lines, in which FAK mRNA levels were precisely correlated with FAK protein levels. FAK protein overexpression in tumors correlated with nodal metastases. CONCLUSIONS: These findings suggest an involvement of FAK in the onset and progression of HNSCC and provide an insight into a mechanism of FAK activation alternative to gene amplification.


Asunto(s)
Carcinoma de Células Escamosas/enzimología , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Dosificación de Gen , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Neoplasias de Cabeza y Cuello/enzimología , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Movimiento Celular , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/patología , Humanos , Inmunohistoquímica , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Familia-src Quinasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...