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.
Cell Death Dis ; 15(5): 311, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38697987

RESUMEN

Cancer cells are highly dependent on bioenergetic processes to support their growth and survival. Disruption of metabolic pathways, particularly by targeting the mitochondrial electron transport chain complexes (ETC-I to V) has become an attractive therapeutic strategy. As a result, the search for clinically effective new respiratory chain inhibitors with minimized adverse effects is a major goal. Here, we characterize a new OXPHOS inhibitor compound called MS-L6, which behaves as an inhibitor of ETC-I, combining inhibition of NADH oxidation and uncoupling effect. MS-L6 is effective on both intact and sub-mitochondrial particles, indicating that its efficacy does not depend on its accumulation within the mitochondria. MS-L6 reduces ATP synthesis and induces a metabolic shift with increased glucose consumption and lactate production in cancer cell lines. MS-L6 either dose-dependently inhibits cell proliferation or induces cell death in a variety of cancer cell lines, including B-cell and T-cell lymphomas as well as pediatric sarcoma. Ectopic expression of Saccharomyces cerevisiae NADH dehydrogenase (NDI-1) partially restores the viability of B-lymphoma cells treated with MS-L6, demonstrating that the inhibition of NADH oxidation is functionally linked to its cytotoxic effect. Furthermore, MS-L6 administration induces robust inhibition of lymphoma tumor growth in two murine xenograft models without toxicity. Thus, our data present MS-L6 as an inhibitor of OXPHOS, with a dual mechanism of action on the respiratory chain and with potent antitumor properties in preclinical models, positioning it as the pioneering member of a promising drug class to be evaluated for cancer therapy. MS-L6 exerts dual mitochondrial effects: ETC-I inhibition and uncoupling of OXPHOS. In cancer cells, MS-L6 inhibited ETC-I at least 5 times more than in isolated rat hepatocytes. These mitochondrial effects lead to energy collapse in cancer cells, resulting in proliferation arrest and cell death. In contrast, hepatocytes which completely and rapidly inactivated this molecule, restored their energy status and survived exposure to MS-L6 without apparent toxicity.


Asunto(s)
Antineoplásicos , Proliferación Celular , Complejo I de Transporte de Electrón , Mitocondrias , Proteínas de Saccharomyces cerevisiae , Animales , Humanos , Complejo I de Transporte de Electrón/metabolismo , Complejo I de Transporte de Electrón/antagonistas & inhibidores , Antineoplásicos/farmacología , Ratones , Línea Celular Tumoral , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Desacopladores/farmacología , Fosforilación Oxidativa/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/efectos de los fármacos , Ratas , NADH Deshidrogenasa/metabolismo , NADH Deshidrogenasa/antagonistas & inhibidores
2.
Cell Rep Med ; 4(12): 101339, 2023 12 19.
Artículo en Inglés | MEDLINE | ID: mdl-38118405

RESUMEN

Rhabdomyosarcoma (RMS) is the main form of pediatric soft-tissue sarcoma. Its cure rate has not notably improved in the last 20 years following relapse, and the lack of reliable preclinical models has hampered the design of new therapies. This is particularly true for highly heterogeneous fusion-negative RMS (FNRMS). Although methods have been proposed to establish FNRMS organoids, their efficiency remains limited to date, both in terms of derivation rate and ability to accurately mimic the original tumor. Here, we present the development of a next-generation 3D organoid model derived from relapsed adult and pediatric FNRMS. This model preserves the molecular features of the patients' tumors and is expandable for several months in 3D, reinforcing its interest to drug combination screening with longitudinal efficacy monitoring. As a proof-of-concept, we demonstrate its preclinical relevance by reevaluating the therapeutic opportunities of targeting apoptosis in FNRMS from a streamlined approach based on transcriptomic data exploitation.


Asunto(s)
Antineoplásicos , Rabdomiosarcoma , Adulto , Humanos , Niño , Recurrencia Local de Neoplasia/tratamiento farmacológico , Rabdomiosarcoma/tratamiento farmacológico , Rabdomiosarcoma/patología , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Organoides/patología , Muerte Celular
3.
Cell Death Discov ; 9(1): 230, 2023 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-37414800

RESUMEN

Toll-like receptor 3 (TLR3) is a pattern recognition receptor mainly known for its role in innate immune response to infection. Indeed, binding of double-stranded RNA (dsRNA) to TLR3 triggers a pro-inflammatory cascade leading to cytokine release and immune cell activation. Its anti-tumoral potential has emerged progressively, associated with a direct impact on tumor cell death induction and with an indirect action on immune system reactivation. Accordingly, TLR3 agonists are currently being tested in clinical trials for several adult cancers. Meanwhile, TLR3 variants have been linked to auto-immune disorders, and as risk factors of viral infection and cancers. However, aside from neuroblastoma, TLR3 role in childhood cancers has not been evaluated. Here, by integrating public transcriptomic data of pediatric tumors, we unveil that high TLR3 expression is largely associated with a better prognosis in childhood sarcomas. Using osteosarcomas and rhabdomyosarcomas as models, we show that TLR3 efficiently drives tumor cell death in vitro and induces tumor regression in vivo. Interestingly, this anti-tumoral effect was lost in cells expressing the homozygous TLR3 L412F polymorphism, which is enriched in a rhabdomyosarcomas cohort. Thus, our results demonstrate the therapeutic potential associated with the targeting of TLR3 in pediatric sarcomas, but also the need to stratify patients eligible for this clinical approach with respect to the TLR3 variants expressed.

4.
Med Sci (Paris) ; 34(8-9): 701-708, 2018.
Artículo en Francés | MEDLINE | ID: mdl-30230466

RESUMEN

During tumor development, malignant cells rewire their metabolism to meet the biosynthetic needs required to increase their biomass and to overcome their microenvironment constraints. The sustained activation of aerobic glycolysis, also called Warburg effect, is one of these adaptative mechanisms. The progresses in this area of research have revealed the flexibility of cancer cells that alternate between glycolytic and oxidative metabolism to cope with their conditions of development while sharing their energetic resources. In this survey, we review these recent breakthroughs and discuss a model that likens tumor to an evolutive metabolic ecosystem. We further emphasize the ensuing therapeutic applications that target metabolic weaknesses of neoplastic cells.


Asunto(s)
Metabolismo Energético/fisiología , Glucólisis/fisiología , Neoplasias/metabolismo , Neoplasias/patología , Microambiente Tumoral/fisiología , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Humanos , Mitocondrias/metabolismo , Oxidación-Reducción
5.
Mol Cancer Ther ; 17(1): 60-72, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29051320

RESUMEN

Streptozotocin-based chemotherapy is the first-line chemotherapy recommended for advanced pancreatic neuroendocrine tumors (pNETs), whereas targeted therapies, including mTOR inhibitors, are available in second-line treatment. Unfortunately, objective response rates to both treatments are limited. Because mTOR pathway activation, commonly observed in pNETs, has been reported as one of the major mechanisms accounting for chemoresistance, we investigated the potential benefit of mTOR inhibition combined with streptozotocin treatment in a subset of pNETs, namely insulinomas. To evaluate the potential of mTOR inhibition in combination with streptozotocin, we selected four different inhibitors acting at various levels of the pathway (everolimus: inhibition of mTORC1; MK-2206: inhibition of AKT; BKM120: inhibition of PI3K, mTORC1, and mTORC2; and BEZ235: inhibition of mTORC1 and mTORC2). Effects on cell viability and apoptosis were assessed in insulinoma cell lines INS-1E (rat) and MIN6 (mouse) in vitro and were confirmed in vivo by using a mouse model of hepatic tumor dissemination after intrasplenic xenograft. In vitro, all four combinations display synergistic effects. These combinations lead to heterogeneous mTOR pathway inhibition, in agreement with their respective target, and increased apoptosis. In vivo, tumor growth in the liver was significantly inhibited by combining streptozotocin with everolimus (P = 0.0014), BKM120 (P = 0.0092), or BEZ235 (P = 0.008) as compared to each agent alone. These results suggest that targeting the mTOR pathway in combination with streptozotocin could be of potential benefit for insulinomas and pNET patients and thus support further clinical investigations. Mol Cancer Ther; 17(1); 60-72. ©2017 AACR.


Asunto(s)
Insulinoma/tratamiento farmacológico , Estreptozocina/uso terapéutico , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/uso terapéutico , Animales , Femenino , Humanos , Insulinoma/patología , Ratones , Ratones Desnudos , Estreptozocina/farmacología , Serina-Treonina Quinasas TOR/farmacología
6.
Biochim Biophys Acta Gene Regul Mech ; 1860(4): 427-437, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-28215965

RESUMEN

Although Men1 is a well-known tumour suppressor gene, little is known about the functions of Menin, the protein it encodes for. Since few years, numerous publications support a major role of Menin in the control of epigenetics gene regulation. While Menin interaction with MLL complex favours transcriptional activation of target genes through H3K4me3 marks, Menin also represses gene expression via mechanisms involving the Polycomb repressing complex (PRC). Interestingly, Ezh2, the PRC-methyltransferase that catalyses H3K27me3 repressive marks and Menin have been shown to co-occupy a large number of promoters. However, lack of binding between Menin and Ezh2 suggests that another member of the PRC complex is mediating this indirect interaction. Having found that ActivinB - a TGFß superfamily member encoded by the Inhbb gene - is upregulated in insulinoma tumours caused by Men1 invalidation, we hypothesize that Menin could directly participate in the epigenetic-repression of Inhbb gene expression. Using Animal model and cell lines, we report that loss of Menin is directly associated with ActivinB-induced expression both in vivo and in vitro. Our work further reveals that ActivinB expression is mediated through a direct modulation of H3K27me3 marks on the Inhbb locus in Menin-KO cell lines. More importantly, we show that Menin binds on the promoter of Inhbb gene where it favours the recruitment of Ezh2 via an indirect mechanism involving Akt-phosphorylation. Our data suggests therefore that Menin could take an important part to the Ezh2-epigenetic repressive landscape in many cells and tissues through its capacity to modulate Akt phosphorylation.


Asunto(s)
Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Regulación de la Expresión Génica , Histonas/metabolismo , Subunidades beta de Inhibinas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Animales , Línea Celular Tumoral , Embrión de Mamíferos/citología , Fibroblastos/metabolismo , Sitios Genéticos , Subunidades beta de Inhibinas/metabolismo , Lisina , Metilación , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , Complejo Represivo Polycomb 1/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Regiones Promotoras Genéticas/genética , Unión Proteica , Transducción de Señal
7.
J Pathol ; 242(1): 90-101, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28188614

RESUMEN

Foxa2, known as one of the pioneer factors, plays a crucial role in islet development and endocrine functions. Its expression and biological functions are regulated by various factors, including, in particular, insulin and glucagon. However, its expression and biological role in adult pancreatic α-cells remain elusive. In the current study, we showed that Foxa2 was overexpressed in islets from α-cell-specific Men1 mutant mice, at both the transcriptional level and the protein level. More importantly, immunostaining analyses showed its prominent nuclear accumulation, specifically in α-cells, at a very early stage after Men1 disruption. Similar nuclear FOXA2 expression was also detected in a substantial proportion (12/19) of human multiple endocrine neoplasia type 1 (MEN1) glucagonomas. Interestingly, our data revealed an interaction between Foxa2 and menin encoded by the Men1 gene. Furthermore, using several approaches, we demonstrated the relevance of this interaction in the regulation of two tested Foxa2 target genes, including the autoregulation of the Foxa2 promoter by Foxa2 itself. The current study establishes menin, a novel protein partner of Foxa2, as a regulator of Foxa2, the biological functions of which extend beyond the pancreatic endocrine cells. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Glucagonoma/metabolismo , Factor Nuclear 3-beta del Hepatocito/biosíntesis , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Neoplasias Pancreáticas/metabolismo , Animales , Regulación Neoplásica de la Expresión Génica , Glucagonoma/genética , Factor Nuclear 3-beta del Hepatocito/genética , Humanos , Ratones Transgénicos , Neoplasia Endocrina Múltiple Tipo 1/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias Pancreáticas/genética , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Transfección , Células Tumorales Cultivadas
8.
Mol Cell Biol ; 36(5): 756-64, 2015 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-26711255

RESUMEN

Loss of pancreatic ß-cell maturity occurs in diabetes and insulinomas. Although both physiological and pathological stresses are known to promote ß-cell dedifferentiation, little is known about the molecules involved in this process. Here we demonstrate that activinB, a transforming growth factor ß (TGF-ß)-related ligand, is upregulated during tumorigenesis and drives the loss of insulin expression and ß-cell maturity in a mouse insulinoma model. Our data further identify Pax4 as a previously unknown activinB target and potent contributor to the observed ß-cell dedifferentiation. More importantly, using compound mutant mice, we found that deleting activinB expression abolishes tumor ß-cell dedifferentiation and, surprisingly, increases survival without significantly affecting tumor growth. Hence, this work reveals an unexpected role for activinB in the loss of ß-cell maturity, islet plasticity, and progression of insulinoma through its participation in ß-cell dedifferentiation.


Asunto(s)
Activinas/metabolismo , Desdiferenciación Celular , Células Secretoras de Insulina/patología , Insulinoma/patología , Páncreas/patología , Neoplasias Pancreáticas/patología , Activinas/genética , Animales , Regulación Neoplásica de la Expresión Génica , Insulina/genética , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Insulinoma/genética , Insulinoma/metabolismo , Ratones Endogámicos C57BL , Páncreas/metabolismo , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo
9.
Ann Endocrinol (Paris) ; 75(3): 133-40, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24997771

RESUMEN

CONTEXT: Multiple Endocrine Neoplasia Type 1 (MEN1) is an autosomal dominant inherited syndrome, related to mutations in the MEN1 gene. Controversial data suggest that the nonsynonymous p.Ala541Thr variant, usually considered as a non-pathogenic polymorphism, may be associated with an increased risk of MEN1-related lesions in carriers. OBJECTIVE: The aim of this study was to evaluate the pathogenic influence of the p.Ala541Thr variant on clinical and functional outcomes. PATIENTS AND METHODS: We analysed a series of 55 index patients carrying the p.Ala541Thr variant. Their clinical profile was compared to that of 117 MEN1 patients. The biological impact of the p.Ala541Thr variant on cell growth was additionally investigated on menin-deficient Leydig cell tumour (LCT)10 cells generated from Men1+/Men1- heterozygous knock-out mice, and compared with wild type (WT). RESULTS: The mean age at first appearance of endocrine lesions was similar in both p.Ala541Thr carriers and MEN1 patients, but no p.Ala541Thr patient had more than one cardinal MEN1 lesion at initial diagnosis. A second MEN1 lesion was diagnosed in 13% of MEN1 patients and in 7% of p.Ala541Thr carriers in the year following preliminary diagnosis. Functional studies on LCT10 cells showed that overexpression of the p.Ala541Thr variant did not inhibit cell growth, which is in direct contrast to results obtained from investigation of WT menin protein. CONCLUSION: Taken together, these data raise the question of a potential pathogenicity of the p.Ala541Thr missense variant of menin that commonly occurs within the general population. Additional studies are required to investigate whether it may be involved in a low-penetrance MEN1 phenotype.


Asunto(s)
Neoplasia Endocrina Múltiple Tipo 1/genética , Mutación , Polimorfismo Genético/genética , Proteínas Proto-Oncogénicas/genética , Adenoma/genética , Adulto , Animales , Línea Celular Tumoral , Femenino , Heterocigoto , Humanos , Hiperparatiroidismo/genética , Tumor de Células de Leydig/genética , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Fenotipo , Neoplasias Hipofisarias/genética , Proteínas Proto-Oncogénicas/deficiencia , Transfección
10.
J Pharm Biomed Anal ; 93: 118-24, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24183932

RESUMEN

Functional consequences of mutations in predisposition genes for familial cancer syndromes remain often elusive, especially when the corresponding gene products play pleiotropic functions and interact with numerous partners. Understanding the consequences of these genetic alterations requires access to their functional effects at the phenotypic level. Nuclear magnetic resonance (NMR) has emerged as a promising functional genomics probe, through its ability to monitor the consequences of genetic variations at the biochemical level. Here, we determine by NMR the metabolic perturbations associated with different disease-related mutations in the MEN1 gene, responsible for the multiple endocrine neoplasia syndrome, type 1 (MEN1), an example of hereditary cancer. The MEN1 gene encodes the Menin protein. Based on a cellular model that allows exogenous overexpression of either the wild type (WT) Menin protein or disease-related variant forms, we evaluate the feasibility of using metabolic profiles to discriminate cells with WT versus variant Menin overexpression. High-resolution magic angle spinning (HRMAS) NMR of whole cells allows to determine the metabolic features associated with overexpression of WT Menin as compared to the one of six different missense variants observed in MEN1 patients. We then identify several statistically significant individual metabolites associated with the metabolic signature of pathogenic versus WT variants. Whether such a metabolic phenotyping approach using cell lines could be exploited as a functional test in a human genetic cancer syndrome is further discussed.


Asunto(s)
Espectroscopía de Resonancia Magnética/métodos , Modelos Biológicos , Neoplasia Endocrina Múltiple Tipo 1/genética , Proteínas Proto-Oncogénicas/genética , Animales , Estudios de Factibilidad , Regulación Neoplásica de la Expresión Génica , Humanos , Mutación Missense , Ratas
11.
PLoS One ; 8(8): e72194, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24013263

RESUMEN

Pax4 and MafA (v-maf musculoaponeurotic fibrosarcoma oncogene homolog A) are two transcription factors crucial for normal functions of islet beta cells in the mouse. Intriguingly, recent studies indicate the existence of notable difference between human and rodent islet in terms of gene expression and functions. To better understand the biological role of human PAX4 and MAFA, we investigated their expression in normal and diseased human islets, using validated antibodies. PAX4 was detected in 43.0±5.0% and 39.1±4.0% of normal human alpha and beta cells respectively. We found that MAFA, detected in 88.3±6.3% insulin(+)cells as in the mouse, turned out to be also expressed in 61.2±6.4% of human glucagons(+) cells with less intensity than in insulin(+) cells, whereas MAFB expression was found not only in the majority of glucagon(+) cells (67.2±7.6%), but also in 53.6±10.5% of human insulin(+) cells. Interestingly, MAFA nuclear expression in both alpha and beta cells, and the percentage of alpha cells expressing PAX4 were found altered in a substantial proportion of patients with type 2 diabetes. Both MAFA and PAX4 display, therefore, a distinct expression pattern in human islet cells, suggesting more potential plasticity of human islets as compared with rodent islets.


Asunto(s)
Diabetes Mellitus Tipo 2/metabolismo , Células Secretoras de Glucagón/metabolismo , Proteínas de Homeodominio/metabolismo , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Factores de Transcripción Paired Box/metabolismo , Adolescente , Adulto , Anciano , Animales , Estudios de Casos y Controles , Núcleo Celular/metabolismo , Células Cultivadas , Diabetes Mellitus Tipo 2/patología , Femenino , Expresión Génica , Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Humanos , Células Secretoras de Insulina/metabolismo , Factores de Transcripción Maf de Gran Tamaño/genética , Masculino , Ratones , Persona de Mediana Edad , Obesidad/metabolismo , Factores de Transcripción Paired Box/genética , Páncreas/metabolismo , Páncreas/patología
12.
Am J Pathol ; 178(1): 336-44, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21224070

RESUMEN

The mammalian target of rapamycin (mTOR) inhibitors, such as rapalogues, are a promising new tool for the treatment of metastatic gastroenteropancreatic endocrine tumors. However, their mechanisms of action remain to be established. We used two murine intestinal endocrine tumoral cell lines, STC-1 and GLUTag, to evaluate the antitumor effects of rapamycin in vitro and in vivo in a preclinical model of liver endocrine metastases. In vitro, rapamycin inhibited the proliferation of cells in the basal state and after stimulation by insulin-like growth factor-1. Simultaneously, p70S6 kinase and 4EBP1 phosphorylation was inhibited. In vivo, rapamycin substantially inhibited the intrahepatic growth of STC-1 cells, irrespectively of the timing of its administration and even when the treatment was administered after cell intrahepatic engraftment. In addition, treated animals had significantly prolonged survival (mean survival time: 47.7 days in treated animals versus 31.8 days in controls) and better clinical status. Rapamycin treatment was associated with a significant decrease in mitotic index and in intratumoral vascular density within STC-1 tumors. Furthermore, the antitumoral effect obtained after treatment with a combination of rapamycin and phosphatidylinositol 3-kinase (PI3K) inhibitor LY294002 was more significant than with rapamycin alone in both cell lines. Our results suggest that the antitumor efficacy of rapamycin in neuroendocrine tumors results from a combination of antiproliferative and antiangiogenic effects. Interestingly, a more potent antitumor efficiency could be obtained by simultaneously targeting several levels of the PI3K/mTOR pathway.


Asunto(s)
Antibióticos Antineoplásicos/uso terapéutico , Carcinoma Neuroendocrino/tratamiento farmacológico , Cromonas/uso terapéutico , Neoplasias Intestinales/tratamiento farmacológico , Morfolinas/uso terapéutico , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/uso terapéutico , Sirolimus/uso terapéutico , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Animales , Protocolos de Quimioterapia Combinada Antineoplásica , Carcinoma Neuroendocrino/enzimología , Carcinoma Neuroendocrino/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias Intestinales/enzimología , Neoplasias Intestinales/patología , Ratones , Fosforilación/efectos de los fármacos
13.
Neuroendocrinology ; 91(3): 268-78, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20389030

RESUMEN

Gastroenteropancreatic (GEP) endocrine tumors are hypervascular tumors able to synthesize and secrete high amounts of VEGF. We aimed to study the regulation of VEGF production in GEP endocrine tumors and to test whether some of the drugs currently used in their treatment, such as somatostatin analogues and mTOR inhibitors, may interfere with VEGF secretion. We therefore analyzed the effects of the somatostatin analogue octreotide, the mTOR inhibitor rapamycin, the PI3K inhibitor LY294002, the MEK1 inhibitor PD98059 and the p38 inhibitor SB203850 on VEGF secretion, assessed by ELISA and Western blotting, in three murine endocrine cell lines, STC-1, INS-r3 and INS-r9. Octreotide and rapamycin induced a significant decrease in VEGF production by all three cell lines; LY294002 significantly inhibited VEGF production by STC-1 and INS-r3 only. We detected no effect of PD98059 whereas SB203850 significantly inhibited VEGF secretion in INS-r3 and INS-r9 cells only. By Western blotting analysis, we observed decreased intracellular levels of VEGF and HIF-1alpha under octreotide, rapamycin and LY294002. For rapamycin and LY294002, this effect was likely mediated by the inhibition of the mTOR/HIF-1/VEGF pathway. In addition to its well-known anti-secretory effects, octreotide may also act through the inhibition of the PI3K/Akt pathway, as suggested by the decrease in Akt phosphorylation detected in all three cell lines. In conclusion, our study points out to the complex regulation of VEGF synthesis and secretion in neoplastic GEP endocrine cells and suggests that the inhibition of VEGF production by octreotide and rapamycin may contribute to their therapeutic effects.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Tumores Neuroendocrinos/enzimología , Tumores Neuroendocrinos/metabolismo , Octreótido/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Cromonas/farmacología , Sinergismo Farmacológico , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Flavonoides/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/antagonistas & inhibidores , Imidazoles/farmacología , Insulina/metabolismo , Secreción de Insulina , MAP Quinasa Quinasa 1/antagonistas & inhibidores , Ratones , Morfolinas/farmacología , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Piridinas/farmacología , Ratas , Sirolimus/farmacología , Somatostatina/efectos de los fármacos , Somatostatina/metabolismo , Serina-Treonina Quinasas TOR , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
14.
J Surg Res ; 154(1): 68-77, 2009 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-18708196

RESUMEN

BACKGROUND: Clinical observations suggest that in neuroendocrine digestive tumors a high intratumoral microvascular density is associated with good prognosis. We used an experimental orthotopic xenograft model to analyze the relations between angiogenic activity and tumor progression in this tumor subset. MATERIAL AND METHODS: We compared 2 endocrine cell lines: STC-1, a low vascular endothelial growth factor (VEGF)-producing cell line, and INS-r3, a high VEGF-producing cell line. Tumor cells were grafted in the adventitial layer of the caecal wall of nude mice, sacrificed after 8 wk. RESULTS: At 8 wk, "primary" tumors were present in all animals. STC-1 derived tumors were morphologically moderately differentiated, with high proliferative and apoptotic activities; in contrast, INS-r3 derived tumors were well differentiated, with low proliferative and apoptotic activities. VEGF was expressed in <50% grafted STC-1 cells but in >90% of grafted INS-r3 cells. Microvascular density was significantly higher in INS-r3 derived tumors than in STC-1 derived tumors. All STC-1 derived tumors (n = 8) have invaded the mucosa, in contrast to none of the INS-r3 derived tumors (n = 8); liver metastases were detected in 7/8 animals bearing STC-1 derived tumors and in 0/8 animals with INS-r3 derived tumors, despite the presence of lymph node metastases. CONCLUSIONS: Our experimental data concur with clinical findings to suggest that in well differentiated digestive neuroendocrine tumors angiogenesis is disconnected from tumor progression: the development of a highly vascular tumor microenvironment is correlated with VEGF secretion but is not associated with invasive and metastatic properties; it must therefore be regarded as an indirect marker of differentiation.


Asunto(s)
Carcinoma Neuroendocrino/patología , Neoplasias del Sistema Digestivo/patología , Neoplasias Intestinales/patología , Neovascularización Patológica/patología , Animales , Antígenos Virales de Tumores/genética , Carcinoma Neuroendocrino/irrigación sanguínea , Línea Celular Tumoral , Neoplasias del Sistema Digestivo/irrigación sanguínea , Progresión de la Enfermedad , Glucagón/genética , Insulina/genética , Neoplasias Intestinales/irrigación sanguínea , Metástasis Linfática/patología , Ratones , Ratones Desnudos , Microcirculación , Regiones Promotoras Genéticas , Ratas , Virus 40 de los Simios/inmunología , Trasplante Heterólogo , Factor A de Crecimiento Endotelial Vascular/genética
15.
Gastroenterology ; 135(5): 1698-1709.e2, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18775714

RESUMEN

BACKGROUND & AIMS: Missense mutations account for 30% of mutations identified in patients with the multiple endocrine neoplasia type 1 (MEN1) syndrome. They raise several issues: the distinction between pathogenic mutations and polymorphisms is sometimes difficult and the functional effects of missense mutations are unclear. We aimed to evaluate the functional consequences of missense MEN1 mutations in an appropriate endocrine cellular context. METHODS: From the INS-1 insulinoma cell line, we established clones conditionally over expressing wild-type (WT) menin or its A160T, H317Y, and A541T variants. We compared the consequences of WT or variant menin over expression on apoptotic response after gamma-irradiation and analyzed the interactions of these proteins with p53. RESULTS: WT menin over expression sensitized INS-r3 cells to apoptosis through amplification of caspase-3 activation, increased p53 acetylation, and accelerated p21 activation; moreover, over expressed WT menin could be recovered in p53-containing complexes. For all 3 missense mutations tested, the functional effects observed with WT were impaired significantly and only low amounts of variant menin proteins were recovered in p53-containing complexes. CONCLUSIONS: Taking advantage of a new endocrine cellular model, we show a loss of function for 2 missense disease-related menin mutants and for a controversial variant as well. Furthermore, our results suggest the existence of functional interactions between p53 and menin for the control of apoptosis, which may cast new light on the mechanisms of endocrine tumorigenesis.


Asunto(s)
Apoptosis/efectos de los fármacos , ADN de Neoplasias/genética , Neoplasia Endocrina Múltiple Tipo 1/genética , Mutación Missense , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogénicas/genética , Recuento de Células , Proliferación Celular , Técnica del Anticuerpo Fluorescente Indirecta , Regulación Neoplásica de la Expresión Génica , Genes p53/genética , Humanos , Immunoblotting , Neoplasia Endocrina Múltiple Tipo 1/metabolismo , Neoplasia Endocrina Múltiple Tipo 1/patología , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Reacción en Cadena de la Polimerasa , Proteínas Proto-Oncogénicas/metabolismo , Células Tumorales Cultivadas
16.
Mol Cell Endocrinol ; 291(1-2): 109-15, 2008 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-18590796

RESUMEN

In a previous study, we demonstrated that the Men1 gene is mainly expressed in the proliferative crypt compartment of the small intestine and that a reduction of menin expression in the crypt-like IEC-17 cell line induces an increase in proliferation rate concomitant with an increase in cyclin D1 expression. The aim of the present study was to test the hypothesis that the NF-kappaB pathway may be involved in cyclin D1 overexpression. Transcriptional activity of the cyclin D1 gene promoter was increased upon reduction of menin expression. Blockade of the NF-kappaB pathway restored proliferation, cell cycle, cyclin D1 gene transcription and cyclin D1 expression levels to those observed in the presence of menin. These data support a correlation between cyclin D1 expression, NF-kappaB activity and menin expression in this epithelial cell line and are relevant to the physiological function of menin in regulating proliferation in the intestinal epithelium.


Asunto(s)
Células Epiteliales/fisiología , Mucosa Intestinal/citología , Proteínas Proto-Oncogénicas/metabolismo , Factor de Transcripción ReIA/metabolismo , Factores de Transcripción/metabolismo , Animales , Ciclo Celular/fisiología , Línea Celular , Ciclina D1/genética , Ciclina D1/metabolismo , Duodeno/citología , Duodeno/fisiología , Células Epiteliales/citología , Regulación de la Expresión Génica , Humanos , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/genética , Ratas , Factor de Transcripción ReIA/antagonistas & inhibidores , Factor de Transcripción ReIA/genética , Factores de Transcripción/genética , Transcripción Genética
17.
J Mol Biol ; 333(1): 87-102, 2003 Oct 10.
Artículo en Inglés | MEDLINE | ID: mdl-14516745

RESUMEN

Our previous studies on the human MEN1 (multiple endocrine neoplasia type 1) gene revealed heterogeneity of MEN1 2.8 kb transcripts related to variation in their 5' UTR only. Six distinct exons 1 (e1A-e1F) were isolated that suggested the existence of multiple but not already identified transcriptional start sites (TSS) and of a complex transcriptional control. Identification of a minimal promoter region and its adjacent regulatory regions appears an inescapable step to the understanding of MEN1 gene transcriptional regulation in normal and pathological situations. For this purpose, we subcloned the approximately 2000 bp region situated directly upstream of the exon 2 in front of a luciferase reporter gene, and we analyzed functional consequences of 5' and 3' serial deletions, comparatively in a series of endocrine versus non-endocrine cell lines. Primer extension and RPA experiments demonstrate that in HEK293 cells transcription initiated simultaneously at several points in endogenous MEN1 promoter as well as in transfected promoter fragments in reporter plasmids, mainly in Inr elements that are efficiently employed to synthetize previously described exons e1A-e1D. Functional consequences of TSS deletion are directly related to cellular context. The minimal promoter region is localized between -135 and -36. Five large adjacent cis-regulatory regions (UR1-UR5) exist upstream of this minimal promoter region, whose activity depend not only on the cellular context but also on the presence of a downstream sequence DR1. Five small cis-regulatory elements (C1-C5) are localized between -325 and -107. Overexpression of exogenous menin, the MEN1 gene's product, in mouse embryonic fibroblasts from Men1(-/-) knock-out mice dose-dependently decreases MEN1 promoter activity, through sequences surrounding the minimal promoter. Our data highlight the existence of a complex transcriptional regulation of the MEN1 gene, whose activity is clearly modulated depending not only on the cellular context but also on menin intracellular levels. They are the molecular bases required for a future understanding of a potential specific transcription control in endocrine cells.


Asunto(s)
Regulación de la Expresión Génica , Proteínas de Neoplasias/genética , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas , Secuencia de Bases , Humanos , Datos de Secuencia Molecular , Análisis de Secuencia de ADN , Eliminación de Secuencia
18.
J Biol Chem ; 277(25): 22407-13, 2002 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-11950843

RESUMEN

Little is known about the mechanisms by which protein-derived nutrients regulate hormone gene expression in the intestine. We have previously reported that protein hydrolysates (i.e. peptones), which are representative of the protein fraction in the lumen, increased cholecystokinin (CCK) gene transcription in the STC-1 enteroendocrine cell line. In the present work, we examined the intracellular events evoked by peptones to stimulate CCK gene transcription. In STC-1 cells, peptones stimulated cyclic AMP production and protein kinase A (PKA) activity. This was associated with a nuclear translocation of the PKA catalytic subunit and with a PKA-dependent phosphorylation of the CRE-binding protein (CREB) at Ser(133). Using transient transfection experiments and reporter luciferase assays, we show that peptone-stimulated transcriptional activity of the CCK gene promoter was significantly decreased when the PKA pathway was inhibited. Furthermore, the intracellular calcium chelator 1,2-bis-(O-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid-tetra(acetoxymethyl)ester completely inhibited peptone-induced stimulation of the CCK gene promoter activity, phosphorylation of CREB, and PKA activity. Peptones increased, in a calcium-dependent manner, the phosphorylation of extracellular signal-regulated protein kinases 1 and 2 (ERK1/2) and the MEK inhibitor PD98059 decreased the peptone-induced stimulation of CCK gene promoter activity. This stimulation was also reduced by 30% in the presence of the calcium/calmodulin-dependent protein kinase (CaMK) inhibitor KN-93. Total inhibition was obtained when the PKA, ERK, and CaMK pathways were simultaneously blocked with appropriate inhibitors to these pathways. These results demonstrate the simultaneous involvement of cAMP- and calcium-dependent protein kinases in the stimulation of intestinal CCK gene transcription by protein-derived nutrients.


Asunto(s)
Colecistoquinina/genética , Colecistoquinina/metabolismo , AMP Cíclico/metabolismo , Activación Transcripcional , Secuencias de Aminoácidos , Bencilaminas/farmacología , Western Blotting , Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Línea Celular , Núcleo Celular/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/química , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Genes Dominantes , Humanos , Luciferasas/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Peptonas/química , Peptonas/farmacología , Fosforilación , Plásmidos/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Estructura Terciaria de Proteína , Serina/metabolismo , Transducción de Señal , Sulfonamidas/farmacología , Factores de Tiempo , Transcripción Genética , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...