Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 121
Filtrar
1.
Vet Comp Oncol ; 14(2): 170-80, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24428588

RESUMEN

Insulin-like growth factor type II (IGF-II) is the main cause of non-islet cell tumour hypoglycaemia (NICTH) and insulin is thought to be the only factor causing hypoglycaemia in insulinomas. However, two case reports of pancreatic neuroendocrine tumours (PNETs) producing IGF-II have been previously published: a human and a canine patient. In this study, we investigated clinical, histopathological, immunohistochemical and ultrastructural features, and biological behaviour of canine pancreatic IGF-II-omas, a subgroup of PNETs that has not been previously characterized. Case records of 58 dogs with confirmed PNETs and hypoglycaemia were reviewed: six patients were affected by IGF-II-omas. Surgery was performed in all cases and two dogs had metastases. Four patients remained alive and in remission at 370, 440, 560 and 890 days post-diagnosis; two died of non-tumour-related causes. IGF-II-omas can be differentiated from insulinomas through hypoinsulinaemia, IGF-II positive and insulin negative immunostaining. The prevalence of this neoplasia is low, accounting for just 6% of PNETs.


Asunto(s)
Adenoma de Células de los Islotes Pancreáticos/veterinaria , Enfermedades de los Perros/metabolismo , Factor II del Crecimiento Similar a la Insulina/metabolismo , Neoplasias Pancreáticas/veterinaria , Adenoma de Células de los Islotes Pancreáticos/genética , Adenoma de Células de los Islotes Pancreáticos/metabolismo , Animales , Enfermedades de los Perros/genética , Perros , Femenino , Regulación Neoplásica de la Expresión Génica/fisiología , Factor II del Crecimiento Similar a la Insulina/genética , Masculino , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Estudios Retrospectivos
2.
Sci Rep ; 2: 494, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22768384

RESUMEN

In the mouse model of pancreas endocrine tumor, loss of Vegf (VKO) results in dramatically decreased tumor progression; however, the residual microscopic lesions show increased invasion into surrounding exocrine tissue. Double KO mice of Vegf and hypoxia inducible factor-1α (Hif-1α) showed increased life span and suppressed tumor growth due to increased apoptosis. The increased invasiveness of tumors in VKO mice was diminished in DKO mice to the levels of wild-type mice. Compared to VKO mice, DKO mice also exhibited smaller changes in the expression levels of adhesion molecules, including E-cadherin, N-cadherin, and NCAM. These changes of adhesion molecules were not observed in the primary culture of the tumor cells under hypoxic conditions. Thus, the invasive phenotype observed under angiogenesis inhibition requires Hif-1α, but is not directly caused by acute hypoxia.


Asunto(s)
Adenoma de Células de los Islotes Pancreáticos/genética , Adenoma de Células de los Islotes Pancreáticos/patología , Eliminación de Gen , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Factor A de Crecimiento Endotelial Vascular/genética , Adenoma de Células de los Islotes Pancreáticos/mortalidad , Animales , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Supervivencia Celular/genética , Modelos Animales de Enfermedad , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Ratones Noqueados , Invasividad Neoplásica/genética , Fenotipo
3.
Oncogene ; 31(11): 1459-67, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-21822305

RESUMEN

Tumor-associated macrophages have recently emerged as a key regulatory cell type during cancer progression, and have been found to promote tumor malignancy in the majority of studies performed to date. We show in this study that CD68(+) macrophages positively correlate with tumor grade and liver metastasis in human pancreatic neuroendocrine tumors (PNETs). To investigate the potential mechanisms whereby macrophages can promote PNET progression, we crossed the RIP1-Tag2 (RT2) mouse model of pancreatic islet cancer to colony-stimulating factor-1 (CSF-1)-deficient Csf1(op/op) mice, which have reduced numbers of tissue macrophages. Csf1(op/op) RT2 mice had a substantial reduction in cumulative tumor burden, which interestingly resulted from a significant decrease in angiogenic switching and tumor number, rather than an evident effect on tumor growth. In the tumors that did develop in CSF-1-deficient animals, however, there were no significant differences in tumor cell proliferation, apoptosis, angiogenesis or invasion. CSF-1 deficiency decreased macrophage infiltration by approximately 50% during all stages of RT2 tumor progression. Interestingly, several cytokines were upregulated in CSF-1-deficient RT2 tumors, and neutrophil infiltration was increased. These results show that macrophages are important for promoting PNET development and suggest that additional factors contribute to the recruitment and survival of myeloid cells in RT2 tumors in the absence of CSF-1.


Asunto(s)
Factor Estimulante de Colonias de Macrófagos/deficiencia , Macrófagos/metabolismo , Tumores Neuroendocrinos/genética , Neoplasias Pancreáticas/genética , Adenoma de Células de los Islotes Pancreáticos/genética , Adenoma de Células de los Islotes Pancreáticos/patología , Animales , Humanos , Ratones , Ratones Transgénicos , Neoplasias Pancreáticas/patología
4.
Clin Endocrinol (Oxf) ; 75(6): 791-800, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21627674

RESUMEN

CONTEXT: The MEN1 syndrome is associated with parathyroid, pancreatic and pituitary tumours and is caused by mutations in the MEN1 gene. In general, there is no genotype-phenotype correlation. OBJECTIVES: To characterize a large family with MEN1 with aggressive tumour behaviour: malignant pancreatic endocrine tumours were present in five affected subjects and were the presenting features in three subjects. DESIGN: The coding region of MEN1 was sequenced. Gene copy number analysis was performed by multiplex ligation-dependent probe amplification (MLPA) and array comparative genomic hybridization (aCGH). Loss of heterozygosity (LOH) in tumour tissue was studied by microsatellite analysis. Insulin-like growth factor II (IGF-II) and CDKN1C/p57KIP2 expression were investigated by immunohistochemistry. RESULTS: Mutation screening by conventional PCR sequence analysis of patients' peripheral blood DNA did not reveal any mutation in the MEN1 or CDKN1B gene. Gene copy number analysis by MLPA and aCGH demonstrated a novel monoallelic deletion of 5 kb genomic DNA involving the MEN1 promoter and exons 1 and 2. LOH analysis indicated somatic deletion of maternal chromosome 11, including MEN1 locus (11q13) and 11p15 imprinting control regions (ICR). Methylation analysis of ICR demonstrated ICR1 hypermethylation and ICR2 hypomethylation in the tumour specimens. ICR1 and ICR2 control the expression of IGF-2 and CDKN1C/p57KIP2, respectively. Immunohistochemistry showed that expression of paternally expressed IGF-2 was up-regulated and the maternally expressed CDKN1C/p57KIP2 was lost in the pancreatic endocrine tumours. CONCLUSIONS: Gene copy number analysis by MLPA should be considered in patients with negative conventional mutation screening. Although large MEN1 deletion causes MEN1, disruption of imprinted CDKN1C/p57KIP2 and IGF-2 gene expression may contribute to tumour progression and aggressive phenotype.


Asunto(s)
Neoplasia Endocrina Múltiple Tipo 1/genética , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogénicas/genética , Adenoma de Células de los Islotes Pancreáticos/genética , Adolescente , Adulto , Niño , Familia , Femenino , Eliminación de Gen , Estudios de Asociación Genética , Humanos , Masculino , Persona de Mediana Edad , Linaje , Fenotipo , Análisis de Secuencia de ADN , Adulto Joven
5.
Proc Natl Acad Sci U S A ; 107(40): 17268-73, 2010 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-20855625

RESUMEN

Cancer is a disease subject to both genetic and environmental influences. In this study, we used the RIP1-Tag2 (RT2) mouse model of islet cell carcinogenesis to identify a genetic locus that influences tumor progression to an invasive growth state. RT2 mice inbred into the C57BL/6 (B6) background develop both noninvasive pancreatic neuroendocrine tumors (PNET) and invasive carcinomas with varying degrees of aggressiveness. In contrast, RT2 mice inbred into the C3HeB/Fe (C3H) background are comparatively resistant to the development of invasive tumors, as are RT2 C3HB6(F1) hybrid mice. Using linkage analysis, we identified a 13-Mb locus on mouse chromosome 17 with significant linkage to the development of highly invasive PNETs. A gene residing in this locus, the anaplastic lymphoma kinase (Alk), was expressed at significantly lower levels in PNETs from invasion-resistant C3H mice compared with invasion-susceptible B6 mice, and pharmacological inhibition of Alk led to reduced tumor invasiveness in RT2 B6 mice. Collectively, our results demonstrate that tumor invasion is subject to polymorphic genetic control and identify Alk as a genetic modifier of invasive tumor growth.


Asunto(s)
Adenoma de Células de los Islotes Pancreáticos/genética , Adenoma de Células de los Islotes Pancreáticos/patología , Transformación Celular Neoplásica/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Polimorfismo Genético , Adenoma de Células de los Islotes Pancreáticos/tratamiento farmacológico , Quinasa de Linfoma Anaplásico , Animales , Cromosomas de los Mamíferos , Ligamiento Genético , Sitios Genéticos , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Neoplasias Pancreáticas/tratamiento farmacológico , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/metabolismo , Pirimidinas/uso terapéutico , Proteínas Tirosina Quinasas Receptoras
6.
Mol Cancer Res ; 6(12): 1894-907, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19074834

RESUMEN

Menin is a tumor suppressor protein mutated in patients with multiple endocrine neoplasia type 1. We show that menin is essential for canonical Wnt/beta-catenin signaling in cultured rodent islet tumor cells. In these cells, overexpression of menin significantly enhances TCF gene assay reporter activity in response to beta-catenin activation. Contrastingly, inhibition of menin expression with Men1 siRNA decreases TCF reporter gene activity. Likewise, multiple endocrine neoplasia type 1 disease associated missense mutations of menin abrogate the ability to increase TCF reporter gene activity. We show that menin physically interacts with proteins involved in the canonical Wnt signaling pathway, including beta-catenin, TCF3 (TCFL1), and weakly with TCF4 (TCFL2). Menin overexpression increases expression of the Wnt/beta-catenin downstream target gene Axin2, which is associated with increased H3K4 trimethylation of the Axin2 gene promoter. Moreover, inhibition of menin expression by siRNA abrogates H3K4 trimethylation and Axin2 gene expression. Based on these studies, we hypothesized that Wnt signaling could inhibit islet cell proliferation because loss of menin function is thought to increase endocrine tumor cell proliferation. TGP61 rodent islet tumor cells treated with a glycogen synthase kinase 3beta inhibitor that increases Wnt pathway signaling had decreased cell proliferation compared with vehicle-treated cells. Collectively, these data suggest that menin has an essential role in Wnt/beta-catenin signaling through a mechanism that eventually affects histone trimethylation of the downstream target gene Axin2, and activation of Wnt/beta-catenin signaling inhibits islet tumor cell proliferation.


Asunto(s)
Adenoma de Células de los Islotes Pancreáticos/metabolismo , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Wnt/metabolismo , Adenoma de Células de los Islotes Pancreáticos/genética , Animales , Proteína Axina , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Cricetinae , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Proteínas de Unión al ADN/genética , Histonas/metabolismo , Humanos , Riñón/citología , Metilación , Ratones , Ratones Noqueados , Neoplasia Endocrina Múltiple Tipo 1/genética , Mutación Missense , Neoplasias Pancreáticas/genética , Regiones Promotoras Genéticas/fisiología , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas/química , Proteínas Proto-Oncogénicas/genética , ARN Interferente Pequeño , Transducción de Señal/fisiología , Factor de Transcripción 4 , Factores de Transcripción/genética , beta Catenina/metabolismo
7.
Am J Gastroenterol ; 103(3): 729-32, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18341492

RESUMEN

Gastroenteropancreatic neuroendocrine tumors (GEP-NETs) are usually sporadic; however, familial (inherited) syndromes, such as the multiple endocrine neoplasia 1 (MEN-1) syndrome, von Hippel-Lindau (VHL) syndrome, neurofibromatosis (NF-1), as well as tuberous sclerosis, may be associated with proximal intestinal and pancreatic NETs. For example, 25% of gastrinoma patients have MEN-1 syndrome. Over the last two decades, the genetic basis of tumorigenesis for these familial syndromes has been clearly identified, providing clinicians with useful screening tools for affected families. Also, over the last few years, advanced molecular genetic techniques, such as comparative genomic hybridization (CGH) and loss of heterozygosity (LOH) analyses, have detected some differences in genomic aberrations among various types of NETs. Whether these chromosomic alterations have implications in the treatment of patients and the outcome of the disease is still unclear.


Asunto(s)
Neoplasias Gastrointestinales/genética , Tumores Neuroendocrinos/genética , Neoplasias Pancreáticas/genética , Adenoma de Células de los Islotes Pancreáticos/diagnóstico , Adenoma de Células de los Islotes Pancreáticos/genética , Tumor Carcinoide/diagnóstico , Tumor Carcinoide/genética , Neoplasias Gastrointestinales/diagnóstico , Pruebas Genéticas , Humanos , Síndromes Neoplásicos Hereditarios/diagnóstico , Síndromes Neoplásicos Hereditarios/genética , Tumores Neuroendocrinos/diagnóstico , Neoplasias Pancreáticas/diagnóstico
8.
Genes Chromosomes Cancer ; 47(1): 84-92, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17943967

RESUMEN

Well-differentiated neuroendocrine tumors (WDNT, carcinoid tumors) are uncommon indolent neoplasms. The genetic alterations of these tumors are not well characterized. We used genome-wide high-density single nucleotide polymorphism (SNP) array analysis to detect copy number alterations in 29 WDNTs, including seven lung, seven nonileal gastrointestinal, and 15 ileal tumors, and compared with allelic imbalances in 15 pancreatic endocrine tumors (PETs). Most frequent allelic imbalances in WDNTs were losses of chromosome 18 in 10 tumors (34%), chromosome 21 or 21q in six (21%), chromosome 13 or 13q in five (17%) and chromosome 16 or 16q in four (14%) tumors, and amplification of chromosome 20 or 20p in seven (24%) tumors. We also found one tumor with loss of heterozygosity of chromosomes 10 and 15 without copy number loss. These allelic imbalances were associated with primary site of tumor: loss of chromosome 18 was present exclusively in ileal WDNTs (P = 0.001), and loss of chromosome 21 or 21q was more frequent in nonileal gastrointestinal WDNTs (P = 0.02). The tumors with loss of chromosome 21 were larger compared to tumors without loss (P = 0.03). Chromosomal aberrations were less common in WDNTs from lung and gastrointestinal tract compared to PETs (P = 0.001). Our study shows that genome-wide allelotyping using SNP array is a powerful new tool for the analysis of allelic imbalances in WDNTs, and some of these alterations are tumor site-dependent and are different than in PETs.


Asunto(s)
Adenoma de Células de los Islotes Pancreáticos/genética , Desequilibrio Alélico , Tumor Carcinoide/genética , Diferenciación Celular/genética , Genoma Humano , Polimorfismo de Nucleótido Simple , Adenoma de Células de los Islotes Pancreáticos/patología , Adulto , Anciano , Tumor Carcinoide/patología , Aberraciones Cromosómicas , Femenino , Neoplasias Gastrointestinales/genética , Humanos , Neoplasias del Íleon/genética , Pérdida de Heterocigocidad/genética , Neoplasias Pulmonares/genética , Masculino , Persona de Mediana Edad
10.
Virchows Arch ; 450(5): 549-57, 2007 May.
Artículo en Inglés | MEDLINE | ID: mdl-17429687

RESUMEN

Altered expression of recently described claudins (CLDNs) as members of tight junction (TJ) transmembrane proteins was noted in several malignancies. We aimed to analyze protein and messenger RNA (mRNA) expressions of different CLDNs in human pancreatic endocrine tumors (PET) and ductal adenocarcinomas. A total of 45 formalin-fixed, paraffin-embedded samples were studied. Immunohistochemistry and real-time reverse transcriptase polymerase chain reaction analysis were carried out for quantification of CLDN 1, -2, -3, -4, and -7 expressions. Normal acini and ducts showed strong CLDNs 1, -3, -4, and -7 and scattered CLDN 2 protein expressions, while Langerhans islands revealed only CLDN 3 and -7 expressions. CLDN 2 expression was found in the half of ductal adenocarcinomas, while the vast majority of endocrine tumors were negative. CLDN 1, -4, and -7 immunohistochemistry was positive in all adenocarcinomas, whereas endocrine tumors were completely negative for CLDNs 1 and -4. CLDN 3 and -7 proteins were detected in all endocrine tumors, while CLDN 3 in ductal adenocarcinomas was negative. The mRNA expression of CLDNs showed differences between endocrine tumors and ductal adenocarcinomas, similar as found for protein expression. Our findings support that PET and ductal carcinomas are specifically characterized by different expression pattern of CLDNs. High expressions of CLDN 3 in endocrine tumors and CLDN 4 in ductal carcinomas might attract them as targets for adjuvant therapy.


Asunto(s)
Adenoma de Células de los Islotes Pancreáticos/metabolismo , Carcinoma de Células de los Islotes Pancreáticos/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Proteínas de la Membrana/metabolismo , Neoplasias Pancreáticas/metabolismo , Adenoma de Células de los Islotes Pancreáticos/genética , Adenoma de Células de los Islotes Pancreáticos/patología , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor , Carcinoma de Células de los Islotes Pancreáticos/genética , Carcinoma de Células de los Islotes Pancreáticos/patología , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/secundario , Femenino , Expresión Génica , Humanos , Técnicas para Inmunoenzimas , Masculino , Proteínas de la Membrana/genética , Persona de Mediana Edad , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , ARN Mensajero/metabolismo , Estudios Retrospectivos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
Oncogene ; 26(33): 4817-24, 2007 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-17297449

RESUMEN

Thymidylate synthase (TS) is an essential enzyme for DNA synthesis and repair and elevated levels of TS have been identified as an important prognostic biomarker for colorectal cancer and several other common human malignancies. In addition, TS gene expression has been linked with cell-cycle regulation and cell proliferation through the ability of retinoblastoma protein to repress the transcriptional activation of E2F target genes such as TS. Therefore, overproduction of TS could participate in the progression to a neoplastic phenotype. Consistent with this model, a recent study has suggested that ectopic TS expression can induce a transformed phenotype in mammalian cells. To investigate the role of deregulated TS activity in tumor development, we generated transgenic mice that express high levels of catalytically active human TS (hTS) exclusively in the pancreas and low levels of hTS in multiple other tissues. Analyses of pancreatic tissue in TS transgenic mice revealed abnormalities within the endocrine pancreas, ranging from pancreatic islet hyperplasia to the detection of islet cell tumors. Overexpression of hTS in murine islets provides a new model to study genetic alterations associated with the progression from normal cells to hyperplasia to islet cell tumors, and suggests that this mouse model may be useful for regulating TS activity in vivo for development of cancer prevention and new therapies.


Asunto(s)
Adenoma de Células de los Islotes Pancreáticos/patología , Islotes Pancreáticos/patología , Neoplasias Pancreáticas/patología , Timidilato Sintasa/metabolismo , Adenoma de Células de los Islotes Pancreáticos/enzimología , Adenoma de Células de los Islotes Pancreáticos/genética , Animales , Humanos , Hiperplasia , Immunoblotting , Inmunohistoquímica , Islotes Pancreáticos/enzimología , Islotes Pancreáticos/metabolismo , Ratones , Ratones Transgénicos , Células 3T3 NIH , Neoplasias Pancreáticas/enzimología , Neoplasias Pancreáticas/genética , Timidilato Sintasa/genética , Factores de Tiempo
12.
Clin Endocrinol (Oxf) ; 66(1): 110-4, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17201809

RESUMEN

OBJECTIVE: We and others have reported loss of heterozygosity (LOH) on chromosome 3p25 in sporadic malignant pancreatic endocrine tumours (PETs). A common region of deletion on chromosome 3p25 contains numerous genes, including VHL and PPARgamma, that have been excluded previously as candidate tumour suppressor genes by DNA sequencing analysis. We have analysed whether WNT7A or HDAC11 was biallelically inactivated in a group of well-characterized PETs. PATIENTS AND DESIGN: Ten PETs from eight patients were selected from a previous study, where LOH on chromosome 3p25 was found in 11 out of 22 sporadic PETs. These tumours were examined for inactivating mutations of WNT7A and HDAC11 by direct sequencing of all exons and intron-exon boundaries. Inactivation of WNT7A expression by aberrant CpG island methylation and WNT7A protein expression were evaluated by methylation-specific polymerase chain reaction (PCR) and immunohistochemistry, respectively. HDAC11 protein expression was also examined. RESULTS: No point mutations, deletion or insertions were detected in either WNT7A or HDAC11 in any of the PETs. Two polymorphisms were identified in the third exon of the WNT7A gene. CpG methylation of the WNT7A gene was not detected and the WNT7A and HDAC11 proteins were normally expressed. CONCLUSION: The absence of tumour-specific somatic events in WNT7A and HDAC11 suggests that these genes are unlikely to have a classical tumour suppressor gene role in sporadic malignant PETs. The putative 3p25 tumour suppressor remains to be identified.


Asunto(s)
Adenoma de Células de los Islotes Pancreáticos/genética , Genes Supresores de Tumor , Histona Desacetilasas/genética , Pérdida de Heterocigocidad , Neoplasias Pancreáticas/genética , Proteínas Wnt/genética , Islas de CpG , Metilación de ADN , Análisis Mutacional de ADN , Cartilla de ADN , Exones , Glucagonoma/genética , Histona Desacetilasas/análisis , Humanos , Inmunohistoquímica , Insulinoma/genética , Proteínas Wnt/análisis
14.
Am J Med Genet A ; 140(15): 1669-72, 2006 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-16835931

RESUMEN

Acrochordons are frequently encountered benign skin lesions that may occasionally represent underlying pathology. Pancreatic islet-cell tumors are rare neoplasms and few cases have been described in patients with tuberous sclerosis complex (TSC). A 39-year-old man presenting in acute renal failure was referred to us for further diagnostic evaluation of coincidentally noted dysmorphic features. Physical examination revealed over 1,000 acrochordons in addition to findings meeting criteria for TSC. The diagnosis was confirmed by disclosure of mutation in the TSC2 gene. Further evaluation revealed pancreatic islet cell tumors. Acrochordons are a common skin lesion, but when presenting in an atypical manner or unusual number may be a sign of TSC and underlying occult pathology thereby warranting evaluation of TSC2. Additionally, mutations in TSC2 gene may be a risk factor for developing pancreatic islet-cell tumors.


Asunto(s)
Adenoma de Células de los Islotes Pancreáticos/genética , Adenoma de Células de los Islotes Pancreáticos/patología , Mutación , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patología , Enfermedades de la Piel/genética , Esclerosis Tuberosa/genética , Proteínas Supresoras de Tumor/genética , Adenoma de Células de los Islotes Pancreáticos/complicaciones , Adulto , Femenino , Humanos , Masculino , Neoplasias Pancreáticas/complicaciones , Linaje , Insuficiencia Renal/diagnóstico , Insuficiencia Renal/genética , Enfermedades de la Piel/complicaciones , Esclerosis Tuberosa/complicaciones , Proteína 2 del Complejo de la Esclerosis Tuberosa
15.
J Clin Invest ; 116(3): 642-51, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16470244

RESUMEN

Previously we observed that neural cell adhesion molecule (NCAM) deficiency in beta tumor cells facilitates metastasis into distant organs and local lymph nodes. Here, we show that NCAM-deficient beta cell tumors grew leaky blood vessels with perturbed pericyte-endothelial cell-cell interactions and deficient perivascular deposition of ECM components. Conversely, tumor cell expression of NCAM in a fibrosarcoma model (T241) improved pericyte recruitment and increased perivascular deposition of ECM molecules. Together, these findings suggest that NCAM may limit tumor cell metastasis by stabilizing the microvessel wall. To directly address whether pericyte dysfunction increases the metastatic potential of solid tumors, we studied beta cell tumorigenesis in primary pericyte-deficient Pdgfb(ret/ret) mice. This resulted in beta tumor cell metastases in distant organs and local lymph nodes, demonstrating a role for pericytes in limiting tumor cell metastasis. These data support a new model for how tumor cells trigger metastasis by perturbing pericyte-endothelial cell-cell interactions.


Asunto(s)
Adenoma de Células de los Islotes Pancreáticos/patología , Metástasis de la Neoplasia/patología , Metástasis de la Neoplasia/prevención & control , Neoplasias Pancreáticas/patología , Pericitos/fisiología , Adenoma de Células de los Islotes Pancreáticos/irrigación sanguínea , Adenoma de Células de los Islotes Pancreáticos/genética , Adenoma de Células de los Islotes Pancreáticos/metabolismo , Animales , Comunicación Celular/genética , Endotelio Vascular/metabolismo , Endotelio Vascular/patología , Fibrosarcoma/irrigación sanguínea , Fibrosarcoma/genética , Fibrosarcoma/metabolismo , Fibrosarcoma/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Patológica/metabolismo , Moléculas de Adhesión de Célula Nerviosa/deficiencia , Moléculas de Adhesión de Célula Nerviosa/genética , Moléculas de Adhesión de Célula Nerviosa/fisiología , Neoplasias Pancreáticas/irrigación sanguínea , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Pericitos/metabolismo
16.
Curr Opin Oncol ; 18(1): 23-9, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16357560

RESUMEN

PURPOSE OF REVIEW: Neoplasms of the endocrine pancreas, commonly referenced as pancreatic islet cell tumors, are rare, often well differentiated endocrine neoplasms, whose biology remains poorly characterized. This article reviews the current clinical management of pancreatic islet cell tumors and describes the molecular events that have been studied to guide future therapies of these peculiar neoplasms. RECENT FINDINGS: While some islet cell tumors arise in association with the MEN-1 syndrome, the majority of these neoplasms are sporadic lesions whose underlying genetic and molecular events remain largely unknown. Recent work has identified changes in gene expression occurring in metastatic and non-metastatic islet cell tumors, which appear to correlate with the occurrence of lymph node and liver metastases. Epigenetic alterations of select tumor suppressor genes may influence patient survival, and the presence of gene promoter methylation may be used as a prognostic marker system. In addition, multiple molecular alterations, including changes in expression of cellular proteins with migratory, cell cycle or angiogenic functions, have been demonstrated to influence islet cell tumor growth, invasion and metastatic spread. SUMMARY: Understanding the molecular events underlying the biology of pancreatic islet cell tumors will aid the development of accurate prognostic markers and will guide improved therapeutic modalities in the future.


Asunto(s)
Adenoma de Células de los Islotes Pancreáticos/terapia , Carcinoma de Células de los Islotes Pancreáticos/terapia , Neoplasias Pancreáticas/terapia , Adenoma de Células de los Islotes Pancreáticos/diagnóstico , Adenoma de Células de los Islotes Pancreáticos/epidemiología , Adenoma de Células de los Islotes Pancreáticos/genética , Biomarcadores , Carcinoma de Células de los Islotes Pancreáticos/diagnóstico , Carcinoma de Células de los Islotes Pancreáticos/epidemiología , Carcinoma de Células de los Islotes Pancreáticos/genética , Humanos , Metástasis de la Neoplasia/fisiopatología , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/epidemiología , Neoplasias Pancreáticas/genética , Factores de Elongación Transcripcional/genética
17.
Arq. bras. endocrinol. metab ; 49(5): 735-746, out. 2005. tab
Artículo en Portugués | LILACS | ID: lil-419975

RESUMEN

As síndromes de neoplasias endócrinas múltiplas (NEM) incluem as do tipo 1 (MEN 1) e 2 (MEN 2), a síndrome de von Hippel-Lindau, neurofibromatose tipo 1 e o complexo de Carney. Estas são síndromes genéticas complexas decorrentes de ativação ou inativação de diferentes tipos de genes envolvidos na regulação da proliferação celular. Nesta revisão, discutiremos as manifestações clínicas e o acompanhamento da MEN 1, assim como o rastreamento genético de potenciais portadores de alterações no gene MEN 1. A MEN 1 inclui o desenvolvimento de hiperparatiroidismo primário multifocal, tumores de ilhotas pancreáticas e adenomas de hipófise. Além disso, alguns pacientes podem apresentar manifestações cutâneas como angiofibromas e colagenomas e ainda podem desenvolver outras neoplasias como tumores carcinóides, tumores de tiróide, adenomas de adrenal, lipomas, feocromocitomas e meningiomas. A MEN 1 é uma síndrome hereditária, transmitida de forma autossômica dominante e causada por mutação inativadora do gene MEN 1. O gene MEN 1 codifica uma proteína denominada "menin", que é um gene supressor tumoral. Vários estudos demonstraram sua importância na regulação da proliferação celular e confirmaram seu papel na patogênese da MEN 1. A identificação do gene MEN 1 e sua análise genética resultaram na possibilidade de monitoração de pacientes que ainda não apresentam manifestações clínicas associadas a esta síndrome e diagnóstico precoce e tratamento dos pacientes afetados. Tais medidas poderão implicar em sobrevida maior para estes pacientes. Estudos adicionais visando uma melhor compreensão da função e dos mecanismos de sinalização da proteína "menin" poderão propiciar alternativas terapêuticas para os pacientes que evoluem com malignização de tumores relacionados à MEN 1, podendo resultar em maior sobrevida.


Asunto(s)
Humanos , Neoplasia Endocrina Múltiple Tipo 1 , Mutación/genética , Proteínas Proto-Oncogénicas/genética , Adenoma de Células de los Islotes Pancreáticos/diagnóstico , Adenoma de Células de los Islotes Pancreáticos/genética , Adenoma de Células de los Islotes Pancreáticos/terapia , Pruebas Genéticas , Hiperparatiroidismo Primario/diagnóstico , Hiperparatiroidismo Primario/genética , Hiperparatiroidismo Primario/terapia , Neoplasia Endocrina Múltiple Tipo 1/diagnóstico , Neoplasia Endocrina Múltiple Tipo 1/genética , Neoplasia Endocrina Múltiple Tipo 1/terapia , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/terapia , Neoplasias Hipofisarias/diagnóstico , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/terapia
18.
Genet Med ; 7(2): 131-8, 2005 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-15714081

RESUMEN

PURPOSE: Based on results of diagnostic MEN1 testing, we have attempted to further define the mutational spectrum of the MEN1 gene and the clinical features most frequently associated with MEN1 mutations. METHODS: Mutation testing was performed on blood samples by PCR amplification and sequencing of exons 2 to 10 of the MEN1 gene and the corresponding intron-exon junctions. Pedigree phenotypic information was obtained by written questionnaire. RESULTS: Among 288 presumably unrelated pedigrees, 73 independent mutations were found in 89 families. Five mutations were found in 2 pedigrees, and 4 mutations were seen in more than 2 pedigrees. There were 17 nonsense mutations (23.3%), 2 in-frame deletions (2.7%), 18 frameshift-deletion mutations (24.7%), 10 frameshift-insertion or -duplication mutations (13.7%), 13 splice-site mutations (17.8%), and 13 presumptive missense mutations (17.8%). Thirty-nine of 56 pedigrees with parathyroid and pancreatic islet neoplasia tested positive, compared with 4/24 and 8/32 pedigrees affected with hyperparathyroidism or hyperparathyroidism and pituitary tumors. MEN1 mutations were found in 6/20 sporadic patients, all of whom had both parathyroid and pancreatic neoplasms. Of 14 mutation-negative sporadic patients, 10 exhibited hyperparathyroidism and pituitary tumors without islet cell neoplasia. Somatic mosaicism was detected in 1 sporadic patient. CONCLUSION: Patients from pedigrees with hyperparathyroidism and pancreatic islet tumors are most likely to test positive for MEN1 mutations. Mutations are less often detected in patients from pedigrees with hyperparathyroidism alone or in combination with pituitary tumors without pancreatic islet neoplasia. Sporadic cases are less likely to test positive than familial cases, in part due to somatic mosaicism.


Asunto(s)
Neoplasia Endocrina Múltiple Tipo 1/diagnóstico , Neoplasia Endocrina Múltiple Tipo 1/genética , Proteínas Proto-Oncogénicas/genética , Adenoma de Células de los Islotes Pancreáticos/etiología , Adenoma de Células de los Islotes Pancreáticos/genética , Análisis Mutacional de ADN , Genes Supresores de Tumor , Humanos , Hiperparatiroidismo/etiología , Hiperparatiroidismo/genética , Neoplasia Endocrina Múltiple Tipo 1/patología , Neoplasias Pancreáticas/etiología , Neoplasias Pancreáticas/genética , Linaje , Fenotipo , Reacción en Cadena de la Polimerasa
19.
Arq Bras Endocrinol Metabol ; 49(5): 735-46, 2005 Oct.
Artículo en Portugués | MEDLINE | ID: mdl-16444356

RESUMEN

Multiple endocrine neoplasia (MEN) syndromes include types 1 (MEN 1) and 2 (MEN 2), von Hippel-Lindau syndrome, neurofibromatosis type 1 and Carney complex. These are complex genetic syndromes caused by activation or inactivation of different types of genes known to be involved in the regulation of cell proliferation. In this review we will discuss the clinical manifestations and management of the MEN 1 syndrome as well as the genetic screening of potential MEN 1 gene carriers. MEN 1 is a hereditary syndrome, transmitted in an autosomic dominant fashion and caused by an inactivating mutation of the MEN 1 gene, characterized by the development of primary hyperparathyroidism, islet cell tumors and pituitary adenomas. In addition, these patients can present with cutaneous manifestations such as angiofibromas and collagenomas, and can develop other neoplastic manifestations including carcinoids, thyroid tumors, adrenal adenomas, lipomas, pheochromocytomas and meningiomas. The MEN 1 gene encodes a peptide which is a tumor suppressor gene called menin. Several studies have demonstrated its importance in regulation of cell proliferation and have confirmed its role in the pathogenesis of the MEN 1 syndrome. The discovery of the MEN 1 gene and the genetic analysis of MEN 1 patients have resulted in earlier diagnosis and treatment of asymptomatic carriers which can potentially result in a longer survival of these patients. Further investigation of the function and signaling pathways of the menin protein will hopefully offer therapeutic alternatives to patients with malignant progression of MEN 1-related tumors and also result in improved survival.


Asunto(s)
Neoplasia Endocrina Múltiple Tipo 1 , Mutación/genética , Proteínas Proto-Oncogénicas/genética , Adenoma de Células de los Islotes Pancreáticos/diagnóstico , Adenoma de Células de los Islotes Pancreáticos/genética , Adenoma de Células de los Islotes Pancreáticos/terapia , Pruebas Genéticas , Humanos , Hiperparatiroidismo Primario/diagnóstico , Hiperparatiroidismo Primario/genética , Hiperparatiroidismo Primario/terapia , Neoplasia Endocrina Múltiple Tipo 1/diagnóstico , Neoplasia Endocrina Múltiple Tipo 1/genética , Neoplasia Endocrina Múltiple Tipo 1/terapia , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/terapia , Neoplasias Hipofisarias/diagnóstico , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/terapia
20.
Surgery ; 134(6): 902-8; discussion 909, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14668721

RESUMEN

BACKGROUND: The aberrant promoter methylation of the mismatch repair gene, hMLH1, is associated with microsatellite instability (MSI) in cancer cells and often is associated with a favorable prognosis. METHODS: Pancreatic endocrine neoplasms (PENs) were obtained from 48 patients who underwent surgical resection. Methylation-specific polymerase chain reaction was used to detect methylation in the hMLH1 promoter. Tumor MSI at loci BAT26, BAT25, D2S123, D5S346, and D17S250 was determined with microsatellite polymerase chain reaction. RESULTS: Hypermethylation of the hMLH1 promoter was present in 11 of 48 PENs (23%). Five of the 11 hMLH1-methylated PENs were found to be microsatellite unstable, and MSI was restricted to PENs with hMLH1 hypermethylation. Tumor recurrence at 2 years after surgical resection was significantly less common among the hMLH1-methylated PENs (11%), compared with the unmethylated PENs (35%; P=.038). Patients with hMLH1-methylated PENs experienced improved 5-year survival (100%) compared with patients with unmethylated tumors (56%; P=.010). Likewise, MSI-positive PENs were associated with improved survival compared with MSI-negative tumors (100% vs 59%; P=.017) at 5 years. CONCLUSION: As in hereditary nonpolyposis colorectal cancer in which MSI is associated with improved survival, methylation of hMLH1 leads to MSI in PENs and affords a favorable prognosis.


Asunto(s)
Adenoma de Células de los Islotes Pancreáticos/genética , Metilación de ADN , Inestabilidad Genómica/genética , Repeticiones de Microsatélite/genética , Proteínas de Neoplasias/genética , Neoplasias Pancreáticas/genética , Proteínas Adaptadoras Transductoras de Señales , Proteínas Portadoras , Genes Supresores de Tumor/fisiología , Humanos , Homólogo 1 de la Proteína MutL , Proteínas Nucleares , Valor Predictivo de las Pruebas , Pronóstico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA