Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Mod Pathol ; 31(1): 160-168, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28862263

RESUMEN

An increasing body of evidence supports the involvement of NF1 mutations, constitutional or somatic, in the pathogenesis of gastrointestinal stromal tumors (GISTs). Due to the large size of the NF1 locus, the existence of multiple pseudogenes and the wide spectrum of mechanisms of gene inactivation, the analysis of NF1 gene status is still challenging for most laboratories. Here we sought to assess the efficacy of a recently developed neurofibromin-specific antibody (NFC) in detecting NF1-inactivated GISTs. NFC reactivity was analyzed in a series of 98 GISTs. Of these, 29 were 'NF1-associated' (17 with ascertained NF1 mutations and 12 arising in the context of clinically diagnosed Neurofibromatosis type 1 syndrome and thus considered bona fine NF1 inactivated); 38 were 'NF1-unrelated' (either wild-type or carrying non-pathogenic variants of NF1). Thirty-one additional GISTs with no available information on NF1 gene status or with NF1 gene variants of uncertain pathogenic significance were also included in the analysis. Cases were scored as NFC negative when, in the presence of NFC positive internal controls, no cytoplasmic staining was detected in the neoplastic cells. NFC immunoreactivity was lost in 24/29 (83%) NF1-associated GISTs as opposed to only 2/38 (5%) NF1-unrelated GISTs (P=3e-11). NFC staining loss significantly correlated (P=0.007) with the presence of biallelic NF1 inactivation, due essentially to large deletions or truncating mutations. NFC reactivity was instead retained in two cases in which the NF1 alteration was heterozygous and in one case where the pathogenic NF1 variant, although homo/hemizygous, was a missense mutation predicted not to affect neurofibromin half-life. Overall this study provides evidence that NFC is a valuable tool for identifying NF1-inactivated GISTs, thus serving as a surrogate for molecular analysis.


Asunto(s)
Anticuerpos Monoclonales , Análisis Mutacional de ADN/métodos , Tumores del Estroma Gastrointestinal/genética , Neurofibromina 1/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mutación
2.
Pediatr Blood Cancer ; 64(6)2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-27862886

RESUMEN

Pediatric spinal oligodendrogliomas are rare and aggressive tumors. They do not share the same molecular features of adult oligodendroglioma, and no previous reports have examined the molecular features of pediatric spinal oligodendroglioma. We present the case of a child with a recurrent spinal anaplastic oligodendroglioma. We performed whole exome (paired tumor and germline DNA) and transcriptome (tumor RNA) sequencing, which revealed somatic mutations in NF1 and FGFR1. These data allowed us to explore potential personalized therapies for this patient and expose molecular drivers that may be involved in similar cases.


Asunto(s)
Eliminación de Gen , Proteínas de Neoplasias , Neurofibromina 1 , Oligodendroglioma , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos , Neoplasias de la Columna Vertebral , Preescolar , Exoma , Femenino , Humanos , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Neurofibromina 1/biosíntesis , Neurofibromina 1/genética , Oligodendroglioma/diagnóstico por imagen , Oligodendroglioma/genética , Oligodendroglioma/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/biosíntesis , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Neoplasias de la Columna Vertebral/diagnóstico por imagen , Neoplasias de la Columna Vertebral/genética , Neoplasias de la Columna Vertebral/metabolismo , Transcriptoma
3.
Int J Oncol ; 48(1): 130-44, 2016 01.
Artículo en Inglés | MEDLINE | ID: mdl-26530297

RESUMEN

Advances in genomic and transcriptome sequencing are revealing the massive scale of previously unrecognised alterations occurring during neoplastic transformation. Breast cancers are genetically and phenotypically heterogeneous. Each of the three major subtypes [ERBB2 amplified, estrogen receptor (ESR)-positive and triple-negative] poses diagnostic and therapeutic challenges. Here we show, using high-resolution next-generation transcriptome sequencing, that in all three breast cancer subtypes, but not matched controls, there was significant overexpression of transcripts from intronic and untranslated regions in addition to exons from specific genes, particularly amplified oncogenes and hormone receptors. For key genes ERBB2 and ESR1, we demonstrate that overexpression is linked to the production of highly modified and truncated splice variants in tumours, but not controls, correlated with tumour subtype. Translation of these tumour-specific splice variants generates truncated proteins with altered subcellular locations and functions, modifying the phenotype, affecting tumour biology, and targeted antitumour therapies. In contrast, tumour suppressors TP53, BRCA1/2 and NF1 did not show intronic overexpression or truncated splice variants in cancers. These findings emphasize the detection of intronic as well as exonic changes in the transcriptional landscapes of cancers have profound therapeutic implications.


Asunto(s)
Neoplasias de la Mama/genética , Receptor alfa de Estrógeno/biosíntesis , Receptor ErbB-2/biosíntesis , Transcripción Genética , Transcriptoma/genética , Empalme Alternativo/genética , Proteína BRCA1/biosíntesis , Proteína BRCA2/biosíntesis , Neoplasias de la Mama/clasificación , Neoplasias de la Mama/patología , Receptor alfa de Estrógeno/genética , Exones , Femenino , Regulación Neoplásica de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Intrones/genética , Mutación , Neurofibromina 1/biosíntesis , Receptor ErbB-2/genética , Proteína p53 Supresora de Tumor/biosíntesis
5.
Oncotarget ; 6(19): 17753-63, 2015 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-25980496

RESUMEN

To identify 'melanoma-specific' microRNAs (miRNAs) we used an unbiased microRNA profiling approach to comprehensively study cutaneous melanoma in relation to other solid malignancies, which revealed 233 differentially expressed (≥ 2 fold, p < 0.05) miRNAs. Among the top 20 most significantly different miRNAs was hsa-miR-514a-3p. miR-514a is a member of a cluster of miRNAs (miR-506-514) involved in initiating melanocyte transformation and promotion of melanoma growth. We found miR-514a was expressed in 38/55 (69%) melanoma cell lines but in only 1/34 (3%) other solid cancers. To identify miR-514a regulated targets we conducted a miR-514a-mRNA 'pull-down' experiment, which revealed hundreds of genes, including: CTNNB1, CDK2, MC1R, and NF1, previously associated with melanoma. NF1 was selected for functional validation because of its recent implication inacquired resistance to BRAFV600E-targeted therapy. Luciferase-reporter assays confirmed NF1 as a direct target of miR-514a and over-expression of miR-514a in melanoma cell lines inhibited NF1 expression, which correlated with increased survival of BRAFV600E cells treated with PLX4032. These data provide another mechanism for the dysregulation of the MAPK pathway which may contribute to the profound resistance associated with current RAF-targeted therapies.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/genética , Melanoma/genética , MicroARNs/genética , Neurofibromina 1/biosíntesis , Proteínas Proto-Oncogénicas B-raf/metabolismo , Antineoplásicos/farmacología , Western Blotting , Línea Celular Tumoral , Perfilación de la Expresión Génica , Genes Supresores de Tumor , Humanos , Indoles/farmacología , Melanoma/metabolismo , Melanoma/patología , Mutagénesis Sitio-Dirigida , Neurofibromina 1/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , Sulfonamidas/farmacología , Transfección , Vemurafenib
6.
Proc Natl Acad Sci U S A ; 111(46): E4920-8, 2014 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-25368158

RESUMEN

In cardiomyocytes, calcium is known to control gene expression at the level of transcription, whereas its role in regulating alternative splicing has not been explored. Here we report that, in mouse primary or embryonic stem cell-derived cardiomyocytes, increased calcium levels induce robust and reversible skipping of several alternative exons from endogenously expressed genes. Interestingly, we demonstrate a calcium-mediated splicing regulatory mechanism that depends on changes of histone modifications. Specifically, the regulation occurs through changes in calcium-responsive kinase activities that lead to alterations in histone modifications and subsequent changes in the transcriptional elongation rate and exon skipping. We demonstrate that increased intracellular calcium levels lead to histone hyperacetylation along the body of the genes containing calcium-responsive alternative exons by disrupting the histone deacetylase-to-histone acetyltransferase balance in the nucleus. Consequently, the RNA polymerase II elongation rate increases significantly on those genes, resulting in skipping of the alternative exons. These studies reveal a mechanism by which calcium-level changes in cardiomyocytes impact on the output of gene expression through altering alternative pre-mRNA splicing patterns.


Asunto(s)
Empalme Alternativo , Señalización del Calcio/fisiología , Histona Desacetilasas/fisiología , Histonas/metabolismo , Miocitos Cardíacos/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Acetilación , Empalme Alternativo/efectos de los fármacos , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Exones , Regulación de la Expresión Génica/fisiología , Genes de Neurofibromatosis 1 , Ratones , Miocitos Cardíacos/efectos de los fármacos , Neurofibromina 1/biosíntesis , Neurofibromina 1/genética , Cloruro de Potasio/farmacología , Inhibidores de Proteínas Quinasas/farmacología , ARN Polimerasa II/metabolismo , ARN Mensajero/biosíntesis , ARN Interferente Pequeño/farmacología , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Canales Catiónicos TRPP/fisiología , Elongación de la Transcripción Genética
7.
Int J Biol Macromol ; 57: 22-5, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23500436

RESUMEN

It was previously reported that an antitumor polysaccharide (PGPW1) was isolated from the root of Panax ginseng. To extend our study, we investigated here the anti-invasive and metastatic effects of PGPW1 on human gastric cancer cell line HGC-27 and tried to determine its possible mechanism of action. Both scratch wound-healing and Transwell assay identified that PGPW1 dose-dependently inhibited migration and invasiveness of HGC-27 cells. Furthermore, results of western blot showed that protein levels of Twist and AKR1C2 were inhibited by PGPW1, whereas an increase of NF1 was observed. Moreover, down-regulation of Twist expression by PGPW1 blocked epithelial-mesenchymal transition (EMT), characterized by a gain of epithelial cell markers, E-cadherin, and loss of the mesenchymal markers, vimentin and N-cadherin, at protein levels. Collectively, we confirmed that PGPW1 decreased migration and invasion of HGC-27 cells by regulation of Twist, AKR1C2, NF1, E-cadherin, vimentin and N-cadherin expression. In conclusion, PGPW1 may serve as a powerful chemopreventive agent against gastric cancer metastasis.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Proteínas Nucleares/biosíntesis , Panax/química , Polisacáridos/farmacología , Neoplasias Gástricas/metabolismo , Proteína 1 Relacionada con Twist/biosíntesis , Antígenos CD/biosíntesis , Antígenos CD/genética , Cadherinas/biosíntesis , Cadherinas/genética , Línea Celular Tumoral , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Transición Epitelial-Mesenquimal/efectos de los fármacos , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Hidroxiesteroide Deshidrogenasas/biosíntesis , Hidroxiesteroide Deshidrogenasas/genética , Metástasis de la Neoplasia , Neurofibromina 1/biosíntesis , Neurofibromina 1/genética , Proteínas Nucleares/genética , Polisacáridos/química , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Proteína 1 Relacionada con Twist/genética , Vimentina/biosíntesis , Vimentina/genética
8.
Neurochem Res ; 38(3): 564-72, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23242790

RESUMEN

Passive avoidance (PA) conditioning is a fear motivated task able to initiate a cascade of altered gene expression within the hippocampus, a structure critical to learning and memory. We have previously shown that neurofibromin (NF1) and amyloid precursor protein (APP), two genes implicated in cognitive function, are differentially expressed in brain of dopamine D3 receptor knock-out mice (D(3)R(-/-)), suggesting that the receptor might have a role in their trascriptional regulation. Here in this study, we hypothesized that during acquisition of PA conditioning the expression of NF1 and APP genes could be influenced by D(3)Rs. To address this issue, we analyzed the expression of NF1 and APP in the hippocampus of both wild-type (WT) and D(3)R(-/-) mice subjected to the single trial step-through PA paradigm. Our finding demonstrated that (1) D(3)R(-/-) mice exhibit increased cognitive performance as compared to WT mice in the step-through PA trial; (2) acquisition of PA increased D(3)R and NF1, but not APP expression in WT mice hippocampus; (3) PA-driven NF1 induction in WT was abrogated in D(3)R(-/-) mice and finally that (4) the heightened basal APP expression observed in naive D(3)R(-/-) mice was totally reversed by acquisition of PA. In conclusion, the present finding show for the first time that both D(3)R and NF1 genes are upregulated following PA conditioning and suggest that hippocampal D(3)Rs might be relevant to NF1 transcriptional regulation in the hippocampus.


Asunto(s)
Precursor de Proteína beta-Amiloide/biosíntesis , Reacción de Prevención/fisiología , Neurofibromina 1/biosíntesis , Receptores de Dopamina D3/deficiencia , Animales , Hipocampo/metabolismo , Masculino , Ratones , Ratones Noqueados
9.
PLoS One ; 6(9): e24454, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21931722

RESUMEN

Primary glioblastomas are subdivided into several molecular subtypes. There is an ongoing debate over the cell of origin for these tumor types where some suggest a progenitor while others argue for a stem cell origin. Even within the same molecular subgroup, and using lineage tracing in mouse models, different groups have reached different conclusions. We addressed this problem from a combined mathematical modeling and experimental standpoint. We designed a novel mathematical framework to identify the most likely cells of origin of two glioma subtypes. Our mathematical model of the unperturbed in vivo system predicts that if a genetic event contributing to tumor initiation imparts symmetric self-renewing cell division (such as PDGF overexpression), then the cell of origin is a transit amplifier. Otherwise, the initiating mutations arise in stem cells. The mathematical framework was validated with the RCAS/tv-a system of somatic gene transfer in mice. We demonstrated that PDGF-induced gliomas can be derived from GFAP-expressing cells of the subventricular zone or the cortex (reactive astrocytes), thus validating the predictions of our mathematical model. This interdisciplinary approach allowed us to determine the likelihood that individual cell types serve as the cells of origin of gliomas in an unperturbed system.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Regulación Neoplásica de la Expresión Génica , Genes de Neurofibromatosis 1 , Glioblastoma/metabolismo , Neurofibromina 1/biosíntesis , Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Animales , Encéfalo/patología , Neoplasias Encefálicas/genética , Glioblastoma/genética , Humanos , Inmunohistoquímica/métodos , Ratones , Ratones Transgénicos , Modelos Genéticos , Modelos Teóricos , Mutación , Células Madre/citología
10.
Neuropeptides ; 44(1): 45-51, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19919880

RESUMEN

In our previous study we have identified PACAP, VIP and their receptors in rat malignant peripheral nerve sheath tumor (MPNST) cells, thus showing anti-apoptotic roles. Recently it has been shown that the tumor suppressor neurofibromin, encoded by the Neurofibromatosis type I (NF1) gene, promotes MPNST cells sensitivity to apoptosis after serum withdrawal. In the present study we investigated whether PACAP or VIP negatively regulate NF1 expression under normal or serum-dependent pro-apoptotic culture conditions. Results indicated that serum itself significantly influenced gene and protein levels. In fact, the low NF1 levels of cells cultured in normal serum-containing medium were remarkably increased in cells switched to low- or no-serum after 24h and 48 h. Treatment with 100 nM PACAP or VIP did not affect NF1 expression when using normal amounts of serum, whereas it significantly inhibited transcript and protein levels both in low- or no-serum cultured cells. In particular, PACAP reduced NF1 levels already after 24h in low-serum cultured cells, while VIP showed a similar effect only after serum deprivation. However, both PACAP and VIP downregulated gene and protein levels within 48 h either in low-dose and serum-starved cells. Results were confirmed by fluorescence microscopy, showing that 100 nM PACAP or VIP attenuated neurofibromin cytoplasmic localization only in low- or no-serum cultured cells. The present study provides a comprehensive analysis of both neuropeptides effect on NF1 expression in normal, low- or serum-starved MPNST cells, ameliorating the hypothesis that resistance to apoptosis in serum-deprived cells might be correlated to PACAP-/VIP-induced NF1 inhibition.


Asunto(s)
Genes de Neurofibromatosis 1/efectos de los fármacos , Neoplasias de la Vaina del Nervio/genética , Neoplasias de la Vaina del Nervio/metabolismo , Neoplasias del Sistema Nervioso Periférico/genética , Neoplasias del Sistema Nervioso Periférico/metabolismo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Péptido Intestinal Vasoactivo/farmacología , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Línea Celular Tumoral , Medios de Cultivo , ADN Complementario/biosíntesis , ADN Complementario/genética , Técnica del Anticuerpo Fluorescente , Expresión Génica/efectos de los fármacos , Neurofibromina 1/biosíntesis , Neurofibromina 1/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
11.
Protein Expr Purif ; 65(1): 30-7, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19111619

RESUMEN

The protein giant neurofibromin (320kDa) is the protein product of the NF1 tumor suppressor gene, alterations of which are responsible for the pathogenesis of neurofibromatosis type 1 (NF1). Neurofibromin is a Ras-specific GTPase activating protein (RasGAP) that, 15 years after the cloning of the gene, remains the only clearly defined function of the protein. In a structural proteomics approach, we aimed at defining functions beyond RasGAP activity based on the discovery of structural modules. Given the poor outcome of domain prediction tools, we have undertaken a fragment solubility survey covering the full protein sequence, with the aim of defining new domain boundaries or fragments that could be investigated by biochemical methods including structural analysis. More than 200 constructs have been expressed and tested for solubility in small scale assays. Boundaries were chosen based upon secondary structure predictions, sequence conservation among neurofibromin orthologues and chemical properties of amino acids. Using this strategy we recently discovered a novel bipartite module in neurofibromin. We have expanded our study to include ESPRIT, a library-based construct screen, to perform fragment testing at a finer level with respect to the choice of terminal residues. Our study confirms earlier notions about the challenges neurofibromin presents to the biochemist and points to strategies whereby the success rate may be enhanced in the future.


Asunto(s)
Neurofibromatosis 1/enzimología , Neurofibromina 1/química , Humanos , Neurofibromina 1/biosíntesis , Neurofibromina 1/genética , Neurofibromina 1/aislamiento & purificación , Estabilidad Proteica , Estructura Terciaria de Proteína/fisiología , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Homología de Secuencia de Aminoácido , Solubilidad
12.
J Cell Sci ; 121(Pt 2): 167-77, 2008 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-18089649

RESUMEN

Patients with the genetic disease type I neurofibromatosis (NF1) exhibit characteristic pigmentary lesions associated with loss of a single allele of NF1, encoding the 260 kDa protein neurofibromin. To understand the basis for these pigmentary problems, the properties of melanocytes haploinsufficient for the murine gene Nf1 were studied using Nf1(+/-) knockout mice. We demonstrate that neurofibromin regulates the Kit-Mitf signaling axis in vivo during melanocyte development. Primary Nf1(+/-) melanocytes were purified by FACS to measure melanogenic gene expression. We found that Nf1(+/-) melanocytes exhibit higher levels of melanogenic gene expression than their wild-type counterparts. Both prior to and following Kit stimulation, Nf1(+/-) melanocytes also exhibit increased activation of the MAP kinase pathway compared with primary cells. The melanogenic response of primary melanocytes to Mek inhibition is consistent with the changes observed with Nf1 haploinsufficiency; however, these changes differ from those observed with their immortalized counterparts. The observation that reduction of neurofibromin, either from haploinsufficiency in the case of primary melanocytes or from neurofibromin knockdown in the case of melan-a cells, enhances melanogenic gene expression suggests that neurofibromin plays a dominant role to MEK activity in controlling melanogenic gene expression in murine melanocytes.


Asunto(s)
Regulación de la Expresión Génica , Melanocitos/citología , Neurofibromina 1/fisiología , Alelos , Animales , Diferenciación Celular , Proliferación Celular , Separación Celular , Citometría de Flujo , Heterocigoto , Sistema de Señalización de MAP Quinasas , Melanocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Neurofibromina 1/biosíntesis
13.
Biochem Biophys Res Commun ; 348(3): 971-80, 2006 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-16908010

RESUMEN

Schwann cells derived from peripheral nerve sheath tumors from individuals with Neurofibromatosis Type 1 (NF1) are deficient for the protein neurofibromin, which contains a GAP-related domain (NF1-GRD). Neurofibromin-deficient Schwann cells have increased Ras activation, increased proliferation in response to certain growth stimuli, increased angiogenic potential, and altered cell morphology. This study examined whether expression of functional NF1-GRD can reverse the transformed phenotype of neurofibromin-deficient Schwann cells from both benign and malignant peripheral nerve sheath tumors. We reconstituted the NF1-GRD using retroviral transduction and examined the effects on cell morphology, growth potential, and angiogenic potential. NF1-GRD reconstitution resulted in morphologic changes, a 16-33% reduction in Ras activation, and a 53% decrease in proliferation in neurofibromin-deficient Schwann cells. However, NF1-GRD reconstitution was not sufficient to decrease the in vitro angiogenic potential of the cells. This study demonstrates that reconstitution of the NF1-GRD can at least partially reverse the transformation of human NF1 tumor-derived Schwann cells.


Asunto(s)
Proteínas Activadoras de GTPasa/deficiencia , Proteínas Activadoras de GTPasa/genética , Neurofibromina 1/deficiencia , Neurofibromina 1/genética , Células de Schwann/metabolismo , Secuencia de Bases , Línea Celular , Células Cultivadas , Proteínas Activadoras de GTPasa/biosíntesis , Proteínas Activadoras de GTPasa/metabolismo , Humanos , Datos de Secuencia Molecular , Neurofibromina 1/biosíntesis , Neurofibromina 1/metabolismo , Estructura Terciaria de Proteína/genética , Retroviridae/genética , Transducción Genética
14.
Am J Surg Pathol ; 30(8): 1047-51, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16861979

RESUMEN

The tumorigenesis of sporadic endocrine tumors is still not fully understood. It is well known that patients with von Recklinghausen syndrome (NF-1) (OMIM 162200) carrying NF1 germline mutations are predisposed to endocrine tumors including pheochromocytomas and duodenal somatostatinomas. It is unclear, however, whether the rarely reported occurrence of pancreatic insulinomas in NF-1 patients represents a coincidental finding or whether insulinomas are a rare manifestation of the NF-1 syndrome. To determine the potential association between the NF-1 syndrome and pancreatic endocrine tumors, we analyzed a NF-1 patient with a well-differentiated pancreatic endocrine carcinoma for NF1 mutation, allelic loss of the NF1 gene and its expression in peripheral blood and tumor cells. The germline mutation c. 499 del TGTT known in the family was confirmed by polymerase chain reaction (PCR) and direct sequencing of exon 4 in DNA extracted from peripheral blood. Loss of heterozygosity (LOH) analysis of the NF1 gene was carried out using 3 intragenic microsatellite markers on 17q11.2. RNA expression was examined by reverse transcription and a consecutive PCR spanning intron 3 of the NF1 gene including the mutated site in exon 4. Immunohistochemistry was used to analyze NF-1 protein expression. Mutation analysis of peripheral blood leukocytes confirmed the 4 base pair deletion in exon 4 starting at codon 167 (499 del TGTT). LOH analysis of tumor tissue revealed retention of both NF1 alleles. While reverse transcriptase-PCR of peripheral blood showed bi-allelic expression of both the wild-type NF1 and the mutated form, reverse transcriptase-PCR of tumor extracts demonstrated expression of the mutated but not the wild-type NF1 allele. Additionally, neurofibromin, the NF1 gene product, was absent in the tumor tissue of the NF-1 patient. These results show that the wild-type NF1 transcrips and protein are reduced, in the reported insulinoma, supposedly by epigenetic mechanisms. This provides strong evidence that there is a relationship between von Recklinghausen disease and the patient's insulinoma. In this line, insulinomas may be viewed as a rare manifestation of the NF-1 syndrome. Furthermore, the NF1 gene must be considered as a candidate tumor suppressor gene for sporadic insulinomas and probably other pancreatic endocrine tumors.


Asunto(s)
Insulinoma/etiología , Neurofibromatosis 1/complicaciones , Neoplasias Pancreáticas/etiología , Secuencia de Bases , Genes de Neurofibromatosis 1 , Mutación de Línea Germinal , Humanos , Inmunohistoquímica , Pérdida de Heterocigocidad , Neurofibromina 1/biosíntesis , Neurofibromina 1/genética , Fenotipo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
15.
J Histochem Cytochem ; 54(3): 363-70, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16314442

RESUMEN

Neurofibromatosis type 1 (NF1) is an inherited disease with an incidence of about 1:3000 worldwide. Approximately half of all patients with NF1 present osseous manifestations, which can vary from mild to severely debilitating changes such as congenital pseudarthrosis. In the present study, fracture healing of mouse tibia was followed and specimens were collected 5, 9, 14, and 22 days postoperatively. Experimental pseudarthrosis of rat was followed up to 15 weeks postoperatively. In situ hybridization and immunohistochemistry were used to demonstrate expression of NF1 tumor suppressor and phosphorylated p44/42 mitogen-activated protein kinase (MAPK), an indicator of the Ras-MAPK pathway. The results showed that ossified callus was formed in mouse fracture 22 days after the operation. The final outcome of rat pseudarthrosis was detected 9 weeks after the operation, presenting abundant cartilaginous callus at the pseudarthrosis. NF1 gene expression was noted in the maturing and in the hypertrophic cartilages during normal mouse fracture healing, and in rat pseudarthrosis. Phosphorylated p44/42 MAPK was detected in a subpopulation of the hypertrophic chondrocytes in both models. Furthermore, positive labeling for NF1 mRNA and protein was detected in endothelium in both the pseudarthrosis and in the fracture. In conclusion, NF1 gene expression and function are needed for normal fracture healing, possibly restraining excessive Ras-MAPK pathway activation.


Asunto(s)
Huesos/metabolismo , Curación de Fractura , Fracturas Óseas/metabolismo , Neurofibromina 1/biosíntesis , Seudoartrosis/metabolismo , Animales , Callo Óseo/patología , Cartílago/metabolismo , Cartílago/patología , Endotelio/metabolismo , Fémur/metabolismo , Fémur/patología , Inmunohistoquímica , Hibridación in Situ , Ratones , Proteína Quinasa 1 Activada por Mitógenos/biosíntesis , Proteína Quinasa 3 Activada por Mitógenos/biosíntesis , Neurofibromina 1/genética , Fosforilación , ARN Mensajero/biosíntesis , Ratas , Tibia/metabolismo , Tibia/patología
16.
J Neurosci Res ; 79(3): 318-28, 2005 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-15602756

RESUMEN

Neurofibromatosis type 1 (NF1) is a genetic disease caused by the loss of neurofibromin, which can lead to formation of highly invasive malignant peripheral nerve sheath tumors (MPNST). We characterized platelet-derived growth factor-beta (PDGF-beta) receptor expression levels and signal transduction pathways in NF1 MPNST cell lines and compared them with the expression of PDGF-beta receptors in normal human Schwann cells (nhSC). As examined by Western blotting, PDGF-beta receptor expression levels were similar in nhSC and NF1 MPNST cell lines. MAPK and Akt also were phosphorylated in both cell types to a similar degree in response to PDGF B chains (PDGF-BB). However, increased intracellular calcium (Ca2+) levels in response to PDGF-BB were observed only in the NF1 MPNST cell lines; nhSC did not show any increase in intracellular calcium when stimulated with PDGF-BB. The calcium response in NF1 MPNST cell lines was blocked with thapsigargin, suggesting that the PDGF-BB-stimulated increases in intracellular calcium originated in the internal compartment of the cell rather than reflecting influx of calcium from the extracellular compartment. Calmodulin kinase II (CAMKII) is phosphorylated in response to PDGF-BB in the NF1 MPNST cell lines, whereas no phosphorylation of CAMKII was observed in nhSCs. The decreased growth of NF1 MPNST cell lines after treatment with a CAMKII inhibitor is consistent with the view that aberrant activation of the calcium-signaling pathway by PDGF-BB contributes to the formation of MPNST in NF1 patients.


Asunto(s)
Señalización del Calcio/fisiología , Neoplasias de la Vaina del Nervio/metabolismo , Neurofibromatosis 1/metabolismo , Factor de Crecimiento Derivado de Plaquetas/farmacología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Células de Schwann/metabolismo , Adulto , Becaplermina , Calcio/metabolismo , Señalización del Calcio/efectos de los fármacos , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina , Proteínas Quinasas Dependientes de Calcio-Calmodulina/antagonistas & inhibidores , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Línea Celular , Transformación Celular Neoplásica/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Masculino , Persona de Mediana Edad , Neoplasias de la Vaina del Nervio/tratamiento farmacológico , Neoplasias de la Vaina del Nervio/fisiopatología , Neurofibromatosis 1/fisiopatología , Neurofibromina 1/biosíntesis , Fosforilación/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt , Proteínas Proto-Oncogénicas c-sis , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/efectos de los fármacos , Células de Schwann/efectos de los fármacos , Células de Schwann/patología
17.
Trends Genet ; 19(4): 207-16, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12683974

RESUMEN

Alteration of mRNA sequence through base modification mRNA editing frequently generates protein diversity. Several proteins have been identified as being similar to C-to-U mRNA editing enzymes based on their structural domains and the occurrence of a catalytic domain characteristic of cytidine deaminases. In light of the hypothesis that these proteins might represent novel mRNA editing systems that could affect proteome diversity, we consider their structure, expression and relevance to biomedically significant processes or pathologies.


Asunto(s)
Apolipoproteínas B/genética , Citidina Desaminasa/fisiología , Edición de ARN/fisiología , ARN Mensajero/metabolismo , Desaminasas APOBEC-1 , Secuencia de Aminoácidos , Animales , Apolipoproteínas B/biosíntesis , Mamíferos/genética , Ratones , Datos de Secuencia Molecular , Neurofibromina 1/biosíntesis , Neurofibromina 1/genética , Conejos , Alineación de Secuencia , Homología de Secuencia de Aminoácido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA