Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Cell Tissue Res ; 372(2): 379-392, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29427052

RESUMEN

Paragangliomas and pheochromocytomas are rare neuroendocrine tumors characterized by a large spectrum of hereditary predisposition. Based on gene expression profiling classification, they can be classically assigned to either a hypoxic/angiogenic cluster (cluster 1 including tumors with mutations in SDHx, VHL and FH genes) or a kinase-signaling cluster (cluster 2 consisting in tumors related to RET, NF1, TMEM127 and MAX genes mutations, as well as most of the sporadic tumors). The past 15 years have seen the emergence of an increasing number of genetically engineered and grafted models to investigate tumorigenesis and develop new therapeutic strategies. Among them, only cluster 2-related predisposed models have been successful but grafted models are however available to study cluster 1-related tumors. In this review, we present an overview of existing rodent models targeting predisposition genes involved or not in human pheochromocytoma/paraganglioma susceptibility and their contribution to the improvement of pheochromocytoma experimental research.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales/patología , Carcinogénesis/patología , Modelos Animales de Enfermedad , Feocromocitoma/patología , Neoplasias de las Glándulas Suprarrenales/genética , Animales , Carcinogénesis/genética , Predisposición Genética a la Enfermedad , Humanos , Feocromocitoma/genética , Roedores
2.
Hum Mol Genet ; 23(9): 2440-6, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24334767

RESUMEN

Malignant pheochromocytoma (PCC) and paraganglioma (PGL) are mostly caused by germline mutations of SDHB, encoding a subunit of succinate dehydrogenase. Using whole-exome sequencing, we recently identified a mutation in the FH gene encoding fumarate hydratase, in a PCC with an 'SDH-like' molecular phenotype. Here, we investigated the role of FH in PCC/PGL predisposition, by screening for germline FH mutations in a large international cohort of patients. We screened 598 patients with PCC/PGL without mutations in known PCC/PGL susceptibility genes. We searched for FH germline mutations and large deletions, by direct sequencing and multiplex ligation-dependent probe amplification methods. Global alterations in DNA methylation and protein succination were assessed by immunohistochemical staining for 5-hydroxymethylcytosine (5-hmC) and S-(2-succinyl) cysteine (2SC), respectively. We identified five pathogenic germline FH mutations (four missense and one splice mutation) in five patients. Somatic inactivation of the second allele, resulting in a loss of fumarate hydratase activity, was demonstrated in tumors with FH mutations. Low tumor levels of 5-hmC, resembling those in SDHB-deficient tumors, and positive 2SC staining were detected in tumors with FH mutations. Clinically, metastatic phenotype (P = 0.007) and multiple tumors (P = 0.02) were significantly more frequent in patients with FH mutations than those without such mutations. This study reveals a new role for FH in susceptibility to malignant and/or multiple PCC/PGL. Remarkably, FH-deficient PCC/PGLs display the same pattern of epigenetic deregulation as SDHB-mutated malignant PCC/PGL. Therefore, we propose that mutation screening for FH should be included in PCC/PGL genetic testing, at least for tumors with malignant behavior.


Asunto(s)
Fumarato Hidratasa/genética , Mutación de Línea Germinal/genética , Paraganglioma/genética , Feocromocitoma/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Niño , Exones/genética , Femenino , Predisposición Genética a la Enfermedad/genética , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven
3.
PLoS Genet ; 8(6): e1002731, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22761580

RESUMEN

Evolutionary forces that shape regulatory networks remain poorly understood. In mammals, the Rb pathway is a classic example of species-specific gene regulation, as a germline mutation in one Rb allele promotes retinoblastoma in humans, but not in mice. Here we show that p53 transactivates the Retinoblastoma-like 2 (Rbl2) gene to produce p130 in murine, but not human, cells. We found intronic fuzzy tandem repeats containing perfect p53 response elements to be important for this regulation. We next identified two other murine genes regulated by p53 via fuzzy tandem repeats: Ncoa1 and Klhl26. The repeats are poorly conserved in evolution, and the p53-dependent regulation of the murine genes is lost in humans. Our results indicate a role for the rapid evolution of tandem repeats in shaping differences in p53 regulatory networks between mammalian species.


Asunto(s)
Regulación de la Expresión Génica , Proteína p130 Similar a la del Retinoblastoma , Retinoblastoma/genética , Secuencias Repetidas en Tándem/genética , Proteína p53 Supresora de Tumor , Animales , Células Cultivadas , Evolución Molecular , Fibroblastos/citología , Fibroblastos/metabolismo , Humanos , Intrones/genética , Ratones , Mutación , Coactivador 1 de Receptor Nuclear/genética , Coactivador 1 de Receptor Nuclear/metabolismo , Elementos de Respuesta/genética , Proteína p130 Similar a la del Retinoblastoma/genética , Proteína p130 Similar a la del Retinoblastoma/metabolismo , Especificidad de la Especie , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
4.
Int J Cancer ; 135(10): 2237-48, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25124653

RESUMEN

Although the alteration of cellular metabolism in cancer was reported by Warburg in the early 1930s, a regain of interest in cancer metabolism has more recently followed the discovery of germline or somatic mutations in genes coding for metabolic enzymes (succinate dehydrogenase, fumarate hydratase and isocitrate dehydrogenase) that are associated with tumor susceptibility. Mutations in these genes are found in numerous tumor types including paragangliomas, kidney cancers, leiomyomas, glioblastomas and acute myeloid leukemia. They lead to the accumulation of so-called oncometabolites that behave as competitors of 2-oxoglutarate-dependent dioxygenases, involved in a broad spectrum of pathways such as hypoxic response and epigenetic reprogramming. Here, we review the diverse pathways affected by oncometabolites, their potential role in cancer formation, maintenance, metastasis and sensitivity to chemotherapies, as well as emerging new therapeutic strategies.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Metaboloma , Neoplasias/genética , Neoplasias/terapia , Humanos , Neoplasias/metabolismo
5.
FASEB J ; 26(1): 460-7, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21982950

RESUMEN

Markers of prostate tumor recurrence after radical prostatectomy are lacking and highly demanded. The androgen receptor (AR) is a nuclear receptor that plays a pivotal role in normal and cancerous prostate tissue. AR interacts with a number of proteins modulating its stability, localization, and activity. To test the hypothesis that an increased expression of AR partners might foster tumor development, we immunopurified AR partners in human tumors xenografted into mice. One of the identified AR partners was the multifunctional enzyme carbamoyl-phosphate synthetase II, aspartate transcarbamylase, and dihydroorotase (CAD), which catalyzes the 3 initial steps of pyrimidine biosynthesis. We combined experiments in C4-2, LNCaP, 22RV1, and PC3 human prostate cell lines and analysis of frozen radical prostatectomy samples to study the CAD-AR interaction. We show here that in prostate tumor cells, CAD fosters AR translocation into the nucleus and stimulates its transcriptional activity. Notably, in radical prostatectomy specimens, CAD expression was not correlated with proliferation markers, but a higher CAD mRNA level was associated with local tumor extension (P=0.049) and cancer relapse (P=0.017). These results demonstrate an unsuspected function for a key metabolic enzyme and identify CAD as a potential predictive marker of cancer relapse.


Asunto(s)
Aspartato Carbamoiltransferasa/metabolismo , Biomarcadores de Tumor/metabolismo , Carbamoil-Fosfato Sintasa (Glutamina-Hidrolizante)/metabolismo , Dihidroorotasa/metabolismo , Recurrencia Local de Neoplasia/diagnóstico , Neoplasias de la Próstata/diagnóstico , Receptores Androgénicos/metabolismo , Andrógenos/metabolismo , Animales , Aspartato Carbamoiltransferasa/genética , Carbamoil-Fosfato Sintasa (Glutamina-Hidrolizante)/genética , Línea Celular Tumoral , Núcleo Celular/metabolismo , Citosol/metabolismo , Dihidroorotasa/genética , Humanos , Masculino , Ratones , Recurrencia Local de Neoplasia/metabolismo , Trasplante de Neoplasias , Valor Predictivo de las Pruebas , Neoplasias de la Próstata/metabolismo , Pirimidinas/biosíntesis , ARN Interferente Pequeño/farmacología , Receptores Androgénicos/genética , Transcripción Genética/fisiología , Trasplante Heterólogo
6.
Chemistry ; 18(40): 12834-44, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22907863

RESUMEN

This work reveals the preponderance of an intramolecular metal chelation phenomenon in a controlled radical polymerization system involving the reversible trapping of the radical chains by a cobalt complex bis(acetylacetonato)cobalt(II). The cobalt-mediated radical polymerization (CMRP) of a series of N-vinyl amides was considered with the aim of studying the effect of the cobalt chelation by the amide moiety of the last monomer unit of the chain. The latter reinforces the cobalt-polymer bond in the order N-vinylpyrrolidone

7.
Pharmaceutics ; 14(5)2022 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-35631574

RESUMEN

Despite the development of new therapeutic strategies, cancer remains one of the leading causes of mortality worldwide. One of the current major challenges is the resistance of cancers to chemotherapy treatments inducing metastases and relapse of the tumor. The Hedgehog receptor Patched (Ptch1) is overexpressed in many types of cancers. We showed that Ptch1 contributes to the efflux of doxorubicin and plays an important role in the resistance to chemotherapy in adrenocortical carcinoma (ACC), a rare cancer which presents strong resistance to the standard of care chemotherapy treatment. In the present study, we isolated and characterized a subpopulation of the ACC cell line H295R in which Ptch1 is overexpressed and more present at the cell surface. This cell subpopulation is more resistant to doxorubicin, grows as spheroids, and has a greater capability of clonogenicity, migration, and invasion than the parental cells. Xenograft experiments performed in mice and in ovo showed that this cell subpopulation is more tumorigenic and metastatic than the parental cells. These results suggest that this cell subpopulation has cancer stem-like or persistent cell properties which were strengthened by RNA-seq. If present in tumors from ACC patients, these cells could be responsible for therapy resistance, relapse, and metastases.

8.
Cancer Res ; 81(13): 3480-3494, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34127497

RESUMEN

Succinate dehydrogenase is a key enzyme in the tricarboxylic acid cycle and the electron transport chain. All four subunits of succinate dehydrogenase are tumor suppressor genes predisposing to paraganglioma, but only mutations in the SDHB subunit are associated with increased risk of metastasis. Here we generated an Sdhd knockout chromaffin cell line and compared it with Sdhb-deficient cells. Both cell types exhibited similar SDH loss of function, metabolic adaptation, and succinate accumulation. In contrast, Sdhb-/- cells showed hallmarks of mesenchymal transition associated with increased DNA hypermethylation and a stronger pseudo-hypoxic phenotype compared with Sdhd-/- cells. Loss of SDHB specifically led to increased oxidative stress associated with dysregulated iron and copper homeostasis in the absence of NRF2 activation. High-dose ascorbate exacerbated the increase in mitochondrial reactive oxygen species, leading to cell death in Sdhb-/- cells. These data establish a mechanism linking oxidative stress to iron homeostasis that specifically occurs in Sdhb-deficient cells and may promote metastasis. They also highlight high-dose ascorbate as a promising therapeutic strategy for SDHB-related cancers. SIGNIFICANCE: Loss of different succinate dehydrogenase subunits can lead to different cell and tumor phenotypes, linking stronger 2-OG-dependent dioxygenases inhibition, iron overload, and ROS accumulation following SDHB mutation.


Asunto(s)
Ácido Ascórbico/farmacología , Homeostasis , Hierro/metabolismo , Mutación , Estrés Oxidativo , Succinato Deshidrogenasa/fisiología , Animales , Antioxidantes/farmacología , Dioxigenasas/antagonistas & inhibidores , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo , Mitocondrias/patología , Fenotipo , Especies Reactivas de Oxígeno
9.
Cell Rep ; 30(13): 4551-4566.e7, 2020 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-32234487

RESUMEN

Loss-of-function mutations in the SDHB subunit of succinate dehydrogenase predispose patients to aggressive tumors characterized by pseudohypoxic and hypermethylator phenotypes. The mechanisms leading to DNA hypermethylation and its contribution to SDH-deficient cancers remain undemonstrated. We examine the genome-wide distribution of 5-methylcytosine and 5-hydroxymethylcytosine and their correlation with RNA expression in SDHB-deficient tumors and murine Sdhb-/- cells. We report that DNA hypermethylation results from TET inhibition. Although it preferentially affects PRC2 targets and known developmental genes, PRC2 activity does not contribute to the DNA hypermethylator phenotype. We also prove, in vitro and in vivo, that TET silencing, although recapitulating the methylation profile of Sdhb-/- cells, is not sufficient to drive their EMT-like phenotype, which requires additional HIF2α activation. Altogether, our findings reveal synergistic roles of TET repression and pseudohypoxia in the acquisition of metastatic traits, providing a rationale for targeting HIF2α and DNA methylation in SDH-associated malignancies.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Metilación de ADN/genética , Proteínas de Unión al ADN/metabolismo , Mesodermo/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Succinato Deshidrogenasa/genética , 5-Metilcitosina/análogos & derivados , 5-Metilcitosina/metabolismo , Adulto , Anciano , Animales , Hipoxia de la Célula , Línea Celular , Línea Celular Tumoral , Dioxigenasas , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Genoma Humano , Humanos , Masculino , Ratones Desnudos , Persona de Mediana Edad , Mutación/genética , Metástasis de la Neoplasia , Fenotipo , Complejo Represivo Polycomb 2/metabolismo , Succinato Deshidrogenasa/deficiencia
10.
Cancer Res ; 78(8): 1914-1922, 2018 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-29431636

RESUMEN

Comprehensive genetic analyses have identified germline SDHB and FH gene mutations as predominant causes of metastatic paraganglioma and pheochromocytoma. However, some suspicious cases remain unexplained. In this study, we performed whole-exome sequencing of a paraganglioma exhibiting an SDHx-like molecular profile in the absence of SDHx or FH mutations and identified a germline mutation in the SLC25A11 gene, which encodes the mitochondrial 2-oxoglutarate/malate carrier. Germline SLC25A11 mutations were identified in six other patients, five of whom had metastatic disease. These mutations were associated with loss of heterozygosity, suggesting that SLC25A11 acts as a tumor-suppressor gene. Pseudohypoxic and hypermethylator phenotypes comparable with those described in SDHx- and FH-related tumors were observed both in tumors with mutated SLC25A11 and in Slc25a11Δ/Δ immortalized mouse chromaffin knockout cells generated by CRISPR-Cas9 technology. These data show that SLC25A11 is a novel paraganglioma susceptibility gene for which loss of function correlates with metastatic presentation.Significance: A gene encoding a mitochondrial carrier is implicated in a hereditary cancer predisposition syndrome, expanding the role of mitochondrial dysfunction in paraganglioma. Cancer Res; 78(8); 1914-22. ©2018 AACR.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales/genética , Neoplasias de las Glándulas Suprarrenales/patología , Predisposición Genética a la Enfermedad , Mutación de Línea Germinal , Proteínas de Transporte de Membrana/genética , Paraganglioma/secundario , Feocromocitoma/genética , Animales , Sistemas CRISPR-Cas , Estudios de Cohortes , Humanos , Pérdida de Heterocigocidad , Ratones , Ratones Noqueados , Mutación , Metástasis de la Neoplasia , Paraganglioma/genética , Fenotipo , Feocromocitoma/secundario
11.
FASEB J ; 20(3): 476-8, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16423876

RESUMEN

Spermatogenesis is a complex process involving cell proliferation, differentiation, and apoptosis. Fibroblast growth factor 2 (FGF-2) is involved in testicular function, but its role in spermatogenesis has not been fully documented. The control of FGF-2 expression particularly occurs at the translational level, by an internal ribosome entry site (IRES)-dependent mechanism driving the use of alternative initiation codons. To study IRES activity regulation in vivo, we have developed transgenic mice expressing a bicistronic construct coding for two luciferase genes. Here, we show that the FGF-2 IRES is age-dependently activated in mouse testis, whereas EMCV and c-myc IRESs are not. Real-time PCR confirms that this regulation is translational. By using immunohistological techniques, we demonstrate that FGF-2 IRES stimulation occurs in adult, but not in immature, type-A spermatogonias. This is correlated with activation of endogenous FGF-2 expression in spermatogonia; whereas FGF-2 mRNA transcription is known to decrease in adult testis. Interestingly, the FGF-2 IRES activation is triggered by testosterone and is partially inhibited by siRNA directed against the androgen receptor. Two-dimensional analysis of proteins bound to the FGF-2 mRNA 5'UTR after UV cross-linking reveals that testosterone treatment correlates with the binding of several proteins. These data suggest a paracrine loop where IRES-dependent FGF-2 expression, stimulated by Sertoli cells in response to testosterone produced by Leydig cells, would in turn activate Leydig function and testosterone production. In addition, nuclear FGF-2 isoforms could be involved in an intracrine function of FGF-2 in the start of spermatogenesis, mitosis, or meiosis initiation. This report demonstrates that mRNA translation regulation by an IRES-dependent mechanism participates in a physiological process.


Asunto(s)
Factor 2 de Crecimiento de Fibroblastos/fisiología , Células Intersticiales del Testículo/fisiología , Biosíntesis de Proteínas , ARN Mensajero/genética , Secuencias Reguladoras de Ácidos Nucleicos , Células de Sertoli/fisiología , Espermatogénesis/fisiología , Testículo/fisiología , Testosterona/fisiología , Regiones no Traducidas 5' , Factores de Edad , Antagonistas de Receptores Androgénicos , Animales , Codón , Factor 2 de Crecimiento de Fibroblastos/biosíntesis , Factor 2 de Crecimiento de Fibroblastos/genética , Genes Reporteros , Genes Sintéticos , Luciferasas de Renilla/genética , Masculino , Meiosis , Ratones , Ratones Transgénicos , Mitosis , Comunicación Paracrina , Iniciación de la Cadena Peptídica Traduccional/fisiología , Isoformas de Proteínas/fisiología , ARN Mensajero/efectos de la radiación , ARN Interferente Pequeño/farmacología , Receptores Androgénicos/genética , Proteínas Recombinantes de Fusión/fisiología , Ribosomas/metabolismo , Testículo/crecimiento & desarrollo , Testículo/metabolismo , Testosterona/metabolismo , Testosterona/farmacología , Rayos Ultravioleta
12.
Oncotarget ; 8(31): 51050-51057, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28881628

RESUMEN

Adrenocortical carcinoma is a rare neoplasm with a poor prognosis. Very important advances have been made in the identification of the genetic determinants of adrenocortical carcinoma pathogenesis but our understanding is still limited about the mechanisms that determine cancer spread and metastasis. One major problem hindering preclinical experimentation for new therapies for adrenocortical carcinoma is represented by the lack of suitable animal models for metastatic disease. With the aim to overcome these limitations, in this study we tested several protocols in order to establish a mouse xenograft model of metastatic adrenocortical carcinoma. The most efficient method, based upon intrasplenic injection followed by splenectomy, produced metastases with high efficiency, whose development could be followed over time by bioluminescence measurements. We expect that the availability of this model will greatly improve the possibilities for preclinical testing of new treatments for advanced-stage disease.

13.
Sci Signal ; 10(469)2017 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-28270555

RESUMEN

Adrenocortical carcinoma (ACC) is a rare endocrine malignancy with a dismal prognosis. Genomic studies have enabled progress in our understanding of the molecular bases of ACC, but factors that influence its prognosis are lacking. Amplification of the gene encoding the transcription factor steroidogenic factor-1 (SF-1; also known as NR5A1) is one of the genetic alterations common in ACC. We identified a transcriptional regulatory mechanism involving increased abundance of VAV2, a guanine nucleotide exchange factor for small GTPases that control the cytoskeleton, driven by increased expression of the gene encoding SF-1 in ACC. Manipulating SF-1 and VAV2 abundance in cultured ACC cells revealed that VAV2 was a critical factor for SF-1-induced cytoskeletal remodeling and invasion in culture (Matrigel) and in vivo (chicken chorioallantoic membrane) models. Analysis of ACC patient cohorts indicated that greater VAV2 abundance robustly correlated with poor prognosis in ACC patients. Because VAV2 is a druggable target, our findings suggest that blocking VAV2 may be a new therapeutic approach to inhibit metastatic progression in ACC patients.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/genética , Carcinoma Corticosuprarrenal/genética , Regulación Neoplásica de la Expresión Génica , Proteínas Proto-Oncogénicas c-vav/genética , Factor Esteroidogénico 1/genética , Adolescente , Neoplasias de la Corteza Suprarrenal/metabolismo , Neoplasias de la Corteza Suprarrenal/patología , Carcinoma Corticosuprarrenal/metabolismo , Carcinoma Corticosuprarrenal/patología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Línea Celular Tumoral , Embrión de Pollo , Membrana Corioalantoides/metabolismo , Citoesqueleto/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Invasividad Neoplásica , Proteínas Proto-Oncogénicas c-vav/metabolismo , Factor Esteroidogénico 1/metabolismo , Análisis de Supervivencia , Adulto Joven
14.
Mol Cell Endocrinol ; 421: 40-8, 2016 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-26123588

RESUMEN

Pheochromocytomas and paragangliomas (PPGL) are rare neuroendocrine tumors characterized by a high frequency of hereditary forms. Based on transcriptome classification, PPGL can be classified in two different clusters. Cluster 1 tumors are caused by mutations in SDHx, VHL and FH genes and are characterized by a pseudohypoxic signature. Cluster 2 PPGL carry mutations in RET, NF1, MAX or TMEM127 genes and display an activation of the MAPK and mTOR signaling pathways. Many genetically engineered and allografted mouse models have been generated these past 30 years to investigate the mechanisms of PPGL tumorigenesis and test new therapeutic strategies. Among them, only Cluster 2-related models have been successful while no Cluster 1-related knockout mouse was so far reported to develop a PPGL. In this review, we present an overview of existing, successful or not, PPGL models, and a description of our own experience on the quest of Sdhb knockout mouse models of PPGL.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales/genética , Neurofibromina 1/genética , Feocromocitoma/genética , Succinato Deshidrogenasa/genética , Neoplasias de las Glándulas Suprarrenales/patología , Animales , Humanos , Sistema de Señalización de MAP Quinasas , Ratones , Ratones Noqueados , Mutación , Feocromocitoma/patología
15.
Clin Cancer Res ; 22(5): 1120-9, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26490314

RESUMEN

PURPOSE: Germline mutations in genes encoding mitochondrial succinate dehydrogenase (SDH) are found in patients with paragangliomas, pheochromocytomas, gastrointestinal stromal tumors, and renal cancers. SDH inactivation leads to a massive accumulation of succinate, acting as an oncometabolite and which levels, assessed on surgically resected tissue are a highly specific biomarker of SDHx-mutated tumors. The aim of this study was to address the feasibility of detecting succinate in vivo by magnetic resonance spectroscopy. EXPERIMENTAL DESIGN: A pulsed proton magnetic resonance spectroscopy ((1)H-MRS) sequence was developed, optimized, and applied to image nude mice grafted with Sdhb(-/-) or wild-type chromaffin cells. The method was then applied to patients with paraganglioma carrying (n = 5) or not (n = 4) an SDHx gene mutation. Following surgery, succinate was measured using gas chromatography/mass spectrometry, and SDH protein expression was assessed by immunohistochemistry in resected tumors. RESULTS: A succinate peak was observed at 2.44 ppm by (1)H-MRS in all Sdhb(-/-)-derived tumors in mice and in all paragangliomas of patients carrying an SDHx gene mutation, but neither in wild-type mouse tumors nor in patients exempt of SDHx mutation. In one patient, (1)H-MRS results led to the identification of an unsuspected SDHA gene mutation. In another case, it helped define the pathogenicity of a variant of unknown significance in the SDHB gene. CONCLUSIONS: Detection of succinate by (1)H-MRS is a highly specific and sensitive hallmark of SDHx mutations. This noninvasive approach is a simple and robust method allowing in vivo detection of the major biomarker of SDHx-mutated tumors.


Asunto(s)
Complejo II de Transporte de Electrones/genética , Proteínas de la Membrana/genética , Paraganglioma/genética , Feocromocitoma/genética , Succinato Deshidrogenasa/genética , Animales , Predisposición Genética a la Enfermedad , Mutación de Línea Germinal/genética , Humanos , Espectroscopía de Resonancia Magnética , Masculino , Ratones , Paraganglioma/metabolismo , Paraganglioma/patología , Feocromocitoma/patología , Ácido Succínico/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
16.
Oncotarget ; 6(32): 32955-65, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26460615

RESUMEN

Metastatic pheochromocytomas and paragangliomas (PPGL) are malignant neuroendocrine tumors frequently associated with germline mutations in the SDHB gene. SDHB-mutated PPGL display a hypermethylator phenotype associated with hallmarks of epithelial-to-mesenchymal transition (EMT). In the present study, we report the characterization of a unique model of Sdhb knockout in mouse chromaffin cells. Sdhb deficient cells exhibit a metastatic phenotype as highlighted by increased individual cell migration (characterized by faster motility and increased persistence) as well as high invasive and adhesion abilities. This phenotype is associated with the modulation of Twist1, Twist2, Tcf3, Snai1, N-cadherin or Krt19 expression, reflecting an EMT-like reprogramming of cells. Krt19 is epigenetically silenced in Sdhb-deficient cells and re-expressed after treatment by the demethylating agent decitabine. Krt19 rescue by lentiviral transduction in Sdhb-deficient cells and Krt19 inhibition by RNA interference in wild-type cells were performed. Both studies revealed the involvement of KRT19 in the invasive phenotype by modulating collective and individual migration and cell/extra-cellular matrix adhesion properties. These findings underline the role of hypermethylation and EMT in the in vitro acquisition of metastatic properties, following SDHB loss of function.


Asunto(s)
Succinato Deshidrogenasa/deficiencia , Neoplasias de las Glándulas Suprarrenales/genética , Neoplasias de las Glándulas Suprarrenales/metabolismo , Animales , Adhesión Celular/fisiología , Línea Celular Tumoral , Células Cromafines/metabolismo , Células Cromafines/patología , Transición Epitelial-Mesenquimal , Humanos , Ratones , Ratones Noqueados , Invasividad Neoplásica , Metástasis de la Neoplasia , Paraganglioma/genética , Paraganglioma/metabolismo , Feocromocitoma/genética , Feocromocitoma/metabolismo , Succinato Deshidrogenasa/genética , Succinato Deshidrogenasa/metabolismo , Transcriptoma
17.
Nat Commun ; 6: 8784, 2015 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-26522426

RESUMEN

The tricarboxylic acid (TCA) cycle is a central metabolic pathway responsible for supplying reducing potential for oxidative phosphorylation and anabolic substrates for cell growth, repair and proliferation. As such it thought to be essential for cell proliferation and tissue homeostasis. However, since the initial report of an inactivating mutation in the TCA cycle enzyme complex, succinate dehydrogenase (SDH) in paraganglioma (PGL), it has become clear that some cells and tissues are not only able to survive with a truncated TCA cycle, but that they are also able of supporting proliferative phenotype observed in tumours. Here, we show that loss of SDH activity leads to changes in the metabolism of non-essential amino acids. In particular, we demonstrate that pyruvate carboxylase is essential to re-supply the depleted pool of aspartate in SDH-deficient cells. Our results demonstrate that the loss of SDH reduces the metabolic plasticity of cells, suggesting vulnerabilities that can be targeted therapeutically.


Asunto(s)
Complejo II de Transporte de Electrones/metabolismo , Proteínas de la Membrana/metabolismo , Tumores Neuroendocrinos/enzimología , Paraganglioma/enzimología , Ácido Pirúvico/metabolismo , Succinato Deshidrogenasa/metabolismo , Animales , Ácido Aspártico/metabolismo , Complejo II de Transporte de Electrones/genética , Humanos , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Tumores Neuroendocrinos/genética , Tumores Neuroendocrinos/metabolismo , Fosforilación Oxidativa , Paraganglioma/genética , Paraganglioma/metabolismo , Succinato Deshidrogenasa/genética
18.
Nat Commun ; 6: 6044, 2015 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-25625332

RESUMEN

Pheochromocytomas and paragangliomas (PCCs/PGLs) are neural crest-derived tumours with a very strong genetic component. Here we report the first integrated genomic examination of a large collection of PCC/PGL. SNP array analysis reveals distinct copy-number patterns associated with genetic background. Whole-exome sequencing shows a low mutation rate of 0.3 mutations per megabase, with few recurrent somatic mutations in genes not previously associated with PCC/PGL. DNA methylation arrays and miRNA sequencing identify DNA methylation changes and miRNA expression clusters strongly associated with messenger RNA expression profiling. Overexpression of the miRNA cluster 182/96/183 is specific in SDHB-mutated tumours and induces malignant traits, whereas silencing of the imprinted DLK1-MEG3 miRNA cluster appears as a potential driver in a subgroup of sporadic tumours. Altogether, the complete genomic landscape of PCC/PGL is mainly driven by distinct germline and/or somatic mutations in susceptibility genes and reveals different molecular entities, characterized by a set of unique genomic alterations.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales/genética , Predisposición Genética a la Enfermedad , Genoma Humano/genética , Genómica/métodos , Mutación/genética , Paraganglioma/genética , Feocromocitoma/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Niño , Estudios de Cohortes , Variaciones en el Número de Copia de ADN , Metilación de ADN/genética , Exoma/genética , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , MicroARNs/genética , MicroARNs/metabolismo , Persona de Mediana Edad , Análisis de Secuencia de ADN , Adulto Joven
19.
Transcription ; 4(2): 67-71, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23412358

RESUMEN

The clinical importance of tumor suppressor p53 makes it one of the most studied transcription factors. A comparison of mammalian p53 transcriptional repertoires may help identify fundamental principles in genome evolution and better understand cancer processes. Here we summarize mechanisms underlying the divergence of mammalian p53 transcriptional repertoires, with an emphasis on the rapid evolution of fuzzy tandem repeats containing p53 response elements.


Asunto(s)
Proteína p53 Supresora de Tumor/metabolismo , Animales , Evolución Molecular , Humanos , Ratones , Motivos de Nucleótidos , Regiones Promotoras Genéticas , Elementos de Respuesta , Proteína p130 Similar a la del Retinoblastoma/metabolismo , Secuencias Repetidas en Tándem , Proteína p53 Supresora de Tumor/genética
20.
PLoS One ; 7(8): e42252, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22879924

RESUMEN

Almost all prostate cancers respond to androgen deprivation treatment but many recur. We postulated that risk of hormone escape--frequency and delay--are influenced by hormone therapy modalities. More, hormone therapies induce crucial biological changes involving androgen receptors; some might be targets for escape prevention. We investigated the relationship between the androgen deprivation treatment and the risk of recurrence using nude mice bearing the high grade, hormone-dependent human prostate cancer xenograft PAC120. Tumor-bearing mice were treated by Luteinizing-Hormone Releasing Hormone (LHRH) antagonist alone, continuous or intermittent regimen, or combined with androgen receptor (AR) antagonists (bicalutamide or flutamide). Tumor growth was monitored. Biological changes were studied as for genomic alterations, AR mutations and protein expression in a large series of recurrent tumors according to hormone therapy modalities. Therapies targeting Her-2 or AKT were tested in combination with castration. All statistical tests were two-sided. Tumor growth was inhibited by continuous administration of the LH-RH antagonist degarelix (castration), but 40% of tumors recurred. Intermittent castration or complete blockade induced by degarelix and antiandrogens combination, inhibited tumor growth but increased the risk of recurrence (RR) as compared to continuous castration (RR(intermittent): 14.5, RR(complete blockade): 6.5 and 1.35). All recurrent tumors displayed new quantitative genetic alterations and AR mutations, whatever the treatment modalities. AR amplification was found after complete blockade. Increased expression of Her-2/neu with frequent ERK/AKT activation was detected in all variants. Combination of castration with a Her-2/neu inhibitor decreased recurrence risk (0.17) and combination with an mTOR inhibitor prevented it. Anti-hormone treatments influence risk of recurrence although tumor growth inhibition was initially similar. Recurrent tumors displayed genetic instability, AR mutations, and alterations of phosphorylation pathways. We postulated that Her-2/AKT pathways allowed salvage of tumor cells under castration and we demonstrated that their inhibition prevented tumor recurrence in our model.


Asunto(s)
Andrógenos/uso terapéutico , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/enzimología , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto , Andrógenos/deficiencia , Animales , Secuencia de Bases , Castración , Análisis por Conglomerados , Terapia Combinada , Supervivencia sin Enfermedad , Dosificación de Gen/genética , Humanos , Masculino , Ratones , Mutación/genética , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Proteínas Tirosina Quinasas/metabolismo , Receptor ErbB-2/genética , Receptores Androgénicos/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA