Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Mol Oncol ; 13(7): 1534-1547, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31099456

RESUMEN

Estrogens play a pivotal role in breast cancer etiology, and endocrine therapy remains the main first line treatment for estrogen receptor-alpha (ERα)-positive breast cancer. ER are transcription factors whose activity is finely regulated by various regulatory complexes, including histone deacetylases (HDACs). Here, we investigated the role of HDAC9 in ERα signaling and response to antiestrogens in breast cancer cells. Various Michigan Cancer Foundation-7 (MCF7) breast cancer cell lines that overexpress class IIa HDAC9 or that are resistant to the partial antiestrogen 4-hydroxy-tamoxifen (OHTam) were used to study phenotypic changes in response to ER ligands by using transcriptomic and gene set enrichment analyses. Kaplan-Meier survival analyses were performed using public transcriptomic datasets from human breast cancer biopsies. In MCF7 breast cancer cells, HDAC9 decreased ERα mRNA and protein expression and inhibited its transcriptional activity. Conversely, HDAC9 mRNA was strongly overexpressed in OHTam-resistant MCF7 cells and in ERα-negative breast tumor cell lines. Moreover, HDAC9-overexpressing cells were less sensitive to OHTam antiproliferative effects compared with parental MCF7 cells. Several genes (including MUC1, SMC3 and S100P) were similarly deregulated in OHTam-resistant and in HDAC9-overexpressing MCF7 cells. Finally, HDAC9 expression was positively associated with genes upregulated in endocrine therapy-resistant breast cancers and high HDAC9 levels were associated with worse prognosis in patients treated with OHTam. These results demonstrate the complex interactions of class IIa HDAC9 with ERα signaling in breast cancer cells and its effect on the response to hormone therapy.


Asunto(s)
Antineoplásicos Hormonales/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Resistencia a Antineoplásicos , Antagonistas de Estrógenos/farmacología , Histona Desacetilasas/genética , Proteínas Represoras/genética , Neoplasias de la Mama/genética , Receptor alfa de Estrógeno/genética , Femenino , Humanos , Células MCF-7 , Transcriptoma/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
2.
Cell Mol Life Sci ; 76(23): 4769-4781, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31127318

RESUMEN

The estrogen-related receptor γ (ERRγ, NR3B3) is a constitutively active nuclear receptor which has been proposed to act as a mediator of the low-dose effects of a number of environmental endocrine-disrupting chemicals (EDCs) such as the xenoestrogen bisphenol-A (BPA). To better characterize the ability of exogenous compounds to bind and activate ERRγ, we used a combination of cell-based, biochemical, structural and computational approaches. A purposely created stable cell line allowed for the determination of the EC50s for over 30 environmental ERRγ ligands, including previously unknown ones. Interestingly, affinity constants (Kds) of the most potent compounds measured by isothermal titration calorimetry were in the 50-500 nM range, in agreement with their receptor activation potencies. Crystallographic analysis of the interaction between the ERRγ ligand-binding domain (LBD) and compounds of the bisphenol, alkylphenol and naphthol families revealed a partially shared binding mode and minimal alterations of the receptor conformation upon ligand binding. Further biophysical characterizations coupled to molecular dynamics simulations suggested a mechanism through which ERRγ ligands would exhibit their agonistic properties by preserving the transcriptionally active form of the receptor while rigidifying some loop regions with associated functions. This unique mechanism contrasts with the classical one involving a ligand-induced repositioning and stabilization of the C-terminal activation helix H12.


Asunto(s)
Disruptores Endocrinos/química , Receptores de Estrógenos/metabolismo , Compuestos de Bencidrilo/química , Compuestos de Bencidrilo/metabolismo , Compuestos de Bencidrilo/farmacología , Sitios de Unión , Línea Celular Tumoral , Cristalografía por Rayos X , Disruptores Endocrinos/metabolismo , Disruptores Endocrinos/farmacología , Humanos , Ligandos , Simulación de Dinámica Molecular , Fenoles/química , Fenoles/metabolismo , Fenoles/farmacología , Unión Proteica , Estructura Terciaria de Proteína , Receptores de Estrógenos/química , Receptores de Estrógenos/genética , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Termodinámica , Activación Transcripcional/efectos de los fármacos
3.
Artículo en Inglés | MEDLINE | ID: mdl-26029163

RESUMEN

Endocrine-disrupting chemicals (EDCs) are exogenous substances interfering with hormone biosynthesis, metabolism, or action, and consequently causing disturbances in the endocrine system. Various pathways are activated by EDCs, including interactions with nuclear receptors (NRs), which are primary targets of numerous environmental contaminants. The main NRs targeted by environmental contaminants are the estrogen (ER α, ß) and the androgen (AR) receptors. ERs and AR have pleiotropic regulatory roles in a diverse range of tissues, notably in the mammary gland, the uterus, and the prostate. Thus, dysfunctional ERs and AR signaling due to inappropriate exposure to environmental pollutants may lead to hormonal cancers and infertility. The pregnane X receptor (PXR) is also recognized by many environmental molecules. PXR has a protective role of the body through its ability to regulate proteins involved in the metabolism, the conjugation, and the transport of many exogenous and endogenous compounds. However, the permanent activation of this receptor by xenobiotics may lead to premature drug metabolism, the formation, and accumulation of toxic metabolites and defects in hormones homeostasis. The activity of other NRs can also be affected by environmental molecules. Compounds capable of inhibiting or activating the estrogen related (ERRγ), the thyroid hormone (TRα, ß), the retinoid X receptors (RXRα, ß, γ), and peroxisome proliferator-activated (PPAR α, γ) receptors have been identified and are highly suspected to promote developmental, reproductive, neurological, or metabolic diseases in humans and wildlife. In this review, we provide an overview of reporter cell lines established to characterize the human NR activities of a large panel of EDCs including natural as well as industrial compounds such as pesticides, plasticizers, surfactants, flame retardants, and cosmetics.

4.
Front Neurosci ; 9: 212, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26106289

RESUMEN

Zebrafish is increasingly used as an animal model to study the effects of environmental nuclear receptors (NRs) ligands. As most of these compounds have only been tested on human NRs, it is necessary to measure their effects on zebrafish NRs. Estrogen receptors (ER) α and ß and peroxysome proliferator activated receptor (PPAR) γ are main targets of environmental disrupting compounds (EDCs). In humans there are two distinct nuclear ERs (hERα and hERß), whereas the zebrafish genome encodes three ERs, zfERα, zfERß1, and zfERß2. Only one isoform of PPARγ is expressed in both humans and zebrafish. In this review, we described reporter cell lines that we established to study the interaction of EDCs with human and zebrafish ERs and PPARγ. Using these cell lines, we observed that zfERs are thermo-sensitive while zfPPARγ is not. We also showed significant differences in the ability of environmental and synthetic ligands to modulate activation of zfERs and zfPPARγ in comparison to hERs and hPPARγ. Some environmental estrogens (bisphenol A, mycoestrogens) which are hER panagonists displayed greater potency for zfERα as compared to zfERßs. hERß selective agonists (8ßVE2, DPN, phytoestrogens) also displayed zfERα selectivity. Among hERα selective synthetic agonists, 16α-LE2 was the most zfERα selective compound. Almost all zfPPARγ environmental ligands (halogenated bisphenol A derivatives, phthalates, perfluorinated compounds) displayed similar affinity for human and zebrafish PPARγ while pharmaceutical hPPARγ agonists like thiazolidones are not recognized by zfPPARγ. Altogether, our studies show that all hERs and hPPARγ ligands do not control in a similar manner the transcriptional activity of zfERs and zfPPARγ and point out that care has to be taken in transposing the results obtained using the zebrafish as a model for human physiopathology.

5.
J Neurochem ; 134(3): 463-70, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25940785

RESUMEN

Brain peroxisome proliferator-activated receptor gamma (PPARγ), a member of the nuclear receptor superfamily of ligand-dependent transcription factors, is involved in neuroprotection. It is activated by the drug rosiglitazone, which then can increase the pro-survival protein B-cell lymphoma 2 (BCL-2), to mediate neuroprotection. However, the mechanism underlying this molecular cascade remains unknown. Here, we show that the neuroprotective protein neurotrophic factor-α1 (NF-α1), which also induces the expression of BCL-2, has a promoter that contains PPARγ-binding sites that are activated by rosiglitazone. Treatment of Neuro2a cells and primary hippocampal neurons with rosiglitazone increased endogenous NF-α1 expression and prevented H2 O2 -induced cytotoxicity. Concomitant with the increase in NF-α1, BCL-2 was also increased in these cells. When siRNA against NF-α1 was used, the induction of BCL-2 by rosiglitazone was prevented, and the neuroprotective effect of rosiglitazone was reduced. These results demonstrate that rosiglitazone-activated PPARγ directly induces the transcription of NF-α1, contributing to neuroprotection in neurons. We proposed the following cascade for neuroprotection against oxidative stress by rosiglitazone: Rosiglitazone enters the neuron and binds to peroxisome proliferator-activated receptor gamma (PPARγ) in the nucleus. The PPARγ-rosiglitazone complex binds to the neurotrophic factor-α1 (NF-α1) promoter and activates the transcription of NF-α1 mRNA which is then translated to the protein. NF-α1 is the secreted, binds to a cognate receptor and activates the extracellular signal-regulated kinases (ERK) pathway. This in turn enhances the expression of the pro-survival protein, B-cell lymphoma 2 (BCL-2) and inhibition of caspase 3 (Csp-3) to mediate neuroprotection under oxidative stress. Akt, protein kinase B (PKB).


Asunto(s)
Carboxipeptidasa H/metabolismo , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , PPAR gamma/metabolismo , Tiazolidinedionas/farmacología , Animales , Western Blotting , Regulación de la Expresión Génica , Ratones , Neuronas/efectos de los fármacos , Estrés Oxidativo , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa , Rosiglitazona , Transcripción Genética
6.
PLoS One ; 9(2): e88698, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24523932

RESUMEN

Granins and their derived peptides are valuable circulating biological markers of neuroendocrine tumors. The aim of the present study was to investigate the tumoral chromogranin A (CgA)-derived peptide WE-14 and the potential advantage to combine plasma WE-14 detection with the EM66 assay and the existing current CgA assay for the diagnosis of pheochromocytoma. Compared to healthy volunteers, plasma WE-14 levels were 5.4-fold higher in patients with pheochromocytoma, but returned to normal values after surgical resection of the tumor. Determination of plasma CgA and EM66 concentrations in the same group of patients revealed that the test assays for these markers had an overall 84% diagnostic sensitivity, which is identical to that determined for WE-14. However, we found that WE-14 measurement improved the diagnostic sensitivity when combined with the results of CgA or EM66 assays. By combining the results of the three assays, the sensitivity for the diagnosis of pheochromocytoma was increased to 95%. In fact, the combination of WE-14 with either CgA or EM66 test assays achieved 100% sensitivity for the diagnosis of paragangliomas and sporadic or malignant pheochromocytomas if taken separately to account for the heterogeneity of the tumor. These data indicate that WE-14 is produced in pheochromocytoma and secreted into the general circulation, and that elevated plasma WE-14 levels are correlated with the occurrence of this chromaffin cell tumor. In addition, in association with other biological markers, such as CgA and/or EM66, WE-14 measurement systematically improves the diagnostic sensitivity for pheochromocytoma. These findings support the notion that granin-processing products may represent complementary tools for the diagnosis of neuroendocrine tumors.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales/sangre , Cromogranina A/sangre , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/sangre , Tumores Neuroendocrinos/sangre , Péptidos/química , Feocromocitoma/sangre , Adulto , Anciano , Biomarcadores/sangre , Células Cromafines/citología , Femenino , Voluntarios Sanos , Humanos , Masculino , Persona de Mediana Edad , Mutación , Valor Predictivo de las Pruebas , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Adulto Joven
7.
Cell Mol Neurobiol ; 30(8): 1383-9, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21046451

RESUMEN

Pheochromocytomas are catecholamine-producing tumors arising from chromaffin cells of the adrenal medulla or extra-adrenal location. Along with catecholamines, tumoral cells produce and secrete elevated quantities of trophic peptides which are normally released in a regulated manner by the normal adrenal medulla. Among these peptides, the amounts of pituitary adenylate cyclase-activating polypeptide (PACAP), adrenomedullin (AM), and neuropeptide Y (NPY) are particularly high. These peptides can exert endocrine, paracrine or autocrine effects in numerous cell types. In particular, they have been shown to be involved in cell proliferation and survival, catecholamine production and secretion, and angiogenesis. Some of these processes are exacerbated in pheochromocytomas, raising the possibility of the involvement of trophic peptides. Here, we review the expression levels of NPY, PACAP, and AM and theirs receptors in chromaffin cells and pheochromocytomas, and address their possible implication in the adrenal medulla tumorigenesis and malignant development of pheochromocytomas.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales/metabolismo , Células Cromafines/metabolismo , Neuropéptidos/metabolismo , Feocromocitoma/metabolismo , Receptores de Neuropéptido/metabolismo , Neoplasias de las Glándulas Suprarrenales/genética , Animales , Regulación Neoplásica de la Expresión Génica , Humanos , Neuropéptidos/genética , Feocromocitoma/genética , Receptores de Neuropéptido/genética
8.
Regul Pept ; 165(1): 21-9, 2010 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-20600356

RESUMEN

Pheochromocytomas are rare catecholamine-secreting tumors that arise from chromaffin tissue within the adrenal medulla and extra-adrenal sites. Typical clinical manifestations are sustained or paroxysmal hypertension, severe headaches, palpitations and sweating resulting from hormone excess. However, their presentation is highly variable and can mimic many other diseases. The diagnosis of pheochromocytomas depends mainly upon the demonstration of catecholamine excess by 24-h urinary catecholamines and metanephrines or plasma metanephrines. Occurrence of malignant pheochromocytomas can only be asserted by imaging of metastatic lesions, which are associated with a poor survival rate. The characterization of tissue, circulating or genetic markers is therefore crucial for the management of these tumors. Proteins of the granin family and their derived peptides are present in dense-core secretory vesicles and secreted into the bloodstream, making them useful markers for the identification of neuroendocrine cells and neoplasms. In this context, we will focus here on reviewing the distribution and characterization of granins and their processing products in normal and tumoral chromaffin cells, and their clinical usefulness for the diagnosis and prognosis of pheochromocytomas. It appears that, except SgIII, all members of the granin family i.e. CgA, CgB, SgII, SgIV-SgVII and proSAAS, and most of their derived peptides are present in adrenomedullary chromaffin cells and in pheochromocytes. Moreover, besides the routinely used CgA test assays, other assays have been developed to measure concentrations of tissue and/or circulating granins or their derived peptides in order to detect the occurrence of pheochromocytomas. In most cases, elevated levels of these entities were found, in correlation with tumor occurrence, while rarely discriminating between benign and malignant neoplasms. Nevertheless, measurement of the levels of granins and derived peptides improves the diagnostic sensitivity and may therefore provide a complementary tool for the management of pheochromocytomas. However, the existing data need to be substantiated in larger groups of patients, particularly in the case of malignant disease.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales/diagnóstico , Neoplasias de las Glándulas Suprarrenales/patología , Células Cromafines/metabolismo , Células Cromafines/patología , Cromograninas/metabolismo , Feocromocitoma/diagnóstico , Feocromocitoma/patología , Neoplasias de las Glándulas Suprarrenales/metabolismo , Humanos , Feocromocitoma/metabolismo
9.
Endocr Relat Cancer ; 17(3): 637-51, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20483910

RESUMEN

Pheochromocytomas are catecholamine-producing tumors which are generally benign, but which can also present as or develop into malignancy. Molecular pathways of malignant transformation remain poorly understood. Pheochromocytomas express various trophic peptides which may influence tumoral cell behavior. Here, we investigated the expression of trophic amidated peptides, including pituitary adenylate cyclase-activating polypeptide (PACAP), neuropeptide Y (NPY), and adrenomedullin (AM), and their receptors in benign and malignant pheochromocytomas in order to assess their potential role in chromaffin cell tumorigenesis and malignant transformation. PACAP, NPY, and AM are expressed in the majority of pheochromocytomas studied; NPY exhibiting the highest mRNA levels relative to reference genes. Although median gene expression or peptide levels were systematically lower in malignant compared to benign tumors, no statistically significant difference was found. Among all the receptors of these peptides that were analyzed, only the AM receptor RDC1 displayed a differential expression between benign and malignant pheochromocytomas. This receptor exhibited a fourfold higher expression in malignant than in benign tumors. AM and stromal cell-derived factor 1, which has also been described as a ligand for RDC1, increased the number of human pheochromocytoma cells in primary culture and exerted anti-apoptotic activity on rat pheochromocytoma PC12 cells. In addition, RDC1 gene silencing decreased the number of viable PC12 cells. This study shows the expression of several trophic peptides and their receptors in benign and malignant pheochromocytomas, and suggests that AM and its RDC1 receptor could be involved in chromaffin cell tumorigenesis through pro-survival effects. Therefore, AM and RDC1 may represent valuable targets for the treatment of malignant pheochromocytomas.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales/metabolismo , Feocromocitoma/metabolismo , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/metabolismo , Receptores de Péptidos/biosíntesis , Neoplasias de las Glándulas Suprarrenales/genética , Adrenomedulina/biosíntesis , Adrenomedulina/genética , Animales , Western Blotting , Supervivencia Celular/fisiología , Formazáns/química , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Neuropéptido Y/biosíntesis , Neuropéptido Y/genética , Células PC12 , Feocromocitoma/genética , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Interferente Pequeño/farmacología , Ratas , Receptores de Adrenomedulina , Receptores de Neuropéptido Y/biosíntesis , Receptores de Neuropéptido Y/genética , Receptores de Péptidos/genética , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/biosíntesis , Receptores del Polipéptido Activador de la Adenilato-Ciclasa Hipofisaria/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sales de Tetrazolio/química
10.
Endocr Relat Cancer ; 16(1): 281-90, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18948374

RESUMEN

UNLABELLED: The gastroprokinetic agent metoclopramide is known to stimulate catecholamine secretion from pheochromocytomas. The aim of the study was to investigate the mechanism of action of metoclopramide and expression of serotonin type 4 (5-HT(4)) receptors in pheochromocytoma tissues. Tissue explants, obtained from 18 pheochromocytomas including the tumor removed from a 46-year-old female patient who experienced life-threatening hypertension crisis after metoclopramide administration and 17 additional pheochromocytomas (9 benign and 8 malignant) were studied. Cultured pheochromocytoma cells derived from the patient who previously received metoclopramide were incubated with metoclopramide and various 5-HT(4) receptor ligands. In addition, total mRNAs were extracted from all the 18 tumors. Catecholamine- and granin-derived peptide concentrations were measured in pheochromocytoma cell incubation medium by HPLC and radioimmunological assays. In addition, expression of 5-HT(4) receptor mRNAs in the 18 pheochromocytomas was investigated by the use of reverse transcriptase-PCR. RESULTS: Metoclopramide and the 5-HT(4) receptor agonist cisapride were found to activate catecholamine- and granin-derived peptide secretions by cultured tumor cells. Metoclopramide- and cisapride-evoked catecholamine- and granin-derived peptide productions were inhibited by the 5-HT(4) receptor antagonist GR 113808. 5-HT(4) receptor mRNAs were detected in the patient's tumor and the series of 17 additional pheochromocytomas. This study shows that pheochromocytomas express functional 5-HT(4) receptors that are responsible for the stimulatory action of metoclopramide on catecholamine- and granin-derived peptide secretion. All 5-HT(4) receptor agonists must therefore be contraindicated in patients with proven or suspected pheochromocytoma.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales/tratamiento farmacológico , Antagonistas de Dopamina/farmacología , Metoclopramida/farmacología , Feocromocitoma/tratamiento farmacológico , Receptores de Serotonina 5-HT4/genética , Neoplasias de las Glándulas Suprarrenales/metabolismo , Médula Suprarrenal/citología , Médula Suprarrenal/efectos de los fármacos , Catecolaminas/metabolismo , Cromograninas/metabolismo , Cisaprida/farmacología , Contraindicaciones , Domperidona/farmacología , Femenino , Humanos , Persona de Mediana Edad , Feocromocitoma/metabolismo , ARN Mensajero/metabolismo , Receptores de Serotonina 5-HT4/metabolismo , Estudios Retrospectivos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Agonistas de Receptores de Serotonina/farmacología , Células Tumorales Cultivadas
11.
Expert Rev Endocrinol Metab ; 4(1): 45-52, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30934373

RESUMEN

The last 5 years have witnessed important advances in understanding the mechanisms of tumorigenesis of chromaffin cells. Large-scale microarray analyses of pheochromocytomas have identified two distinct gene-expression profiles encompassing all hereditary and sporadic tumors. Gene-expression profiling of benign and malignant pheochromocytomas is providing a better understanding of the mechanisms of metastasis. Such studies hold promise for the development of new prognostic markers for early detection of malignant pheochromocytoma and for the identification of novel targets for therapeutic intervention.

12.
J Neurosci ; 28(26): 6616-26, 2008 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-18579734

RESUMEN

An increase in circulating catecholamine levels represents one of the mechanisms whereby organisms cope with stress. In the periphery, catecholamines mainly originate from the sympathoadrenal system. As we reported, in addition to the central control through cholinergic innervation, a local gap junction-delineated route between adrenal chromaffin cells contributes to catecholamine exocytosis. Here, we investigated whether this intercellular communication is modified when the hormonal demand is increased as observed during cold stress. Our results show that in cold exposed rats, gap-junctional communication undergoes a functional plasticity, as evidenced by an increased number of dye-coupled cells. Of a physiological interest is that this upregulation of gap-junctional coupling results in the appearance of a robust electrical coupling between chromaffin cells that allows the transmission of action potentials between coupled cells. This enhancement of gap-junctional communication parallels an increase in expression levels of connexin36 (Cx36) and connexin43 (Cx43) proteins. Both transcriptional and posttranslational mechanisms are involved because Cx36 transcripts are increased in stressed rats and the expression of the scaffolding protein zonula occludens-1, known to interact with both Cx36 and Cx43, is also upregulated. Consistent with an upregulated coupling extent in stressed rats, the cytosolic Ca(2+) concentration rises triggered in a single cell by an iontophoretic application of nicotine occur simultaneously in several neighboring cells. These results describe for the first time a functional plasticity of junctional coupling between adult chromaffin cells that should be crucial for adaptation to stress or sensitization to subsequent stressors.


Asunto(s)
Médula Suprarrenal/metabolismo , Catecolaminas/metabolismo , Comunicación Celular/fisiología , Células Cromafines/metabolismo , Uniones Comunicantes/metabolismo , Estrés Psicológico/metabolismo , Potenciales de Acción/fisiología , Médula Suprarrenal/ultraestructura , Animales , Calcio/metabolismo , Señalización del Calcio/fisiología , Células Cromafines/ultraestructura , Frío/efectos adversos , Conexina 43/genética , Conexina 43/metabolismo , Conexinas/genética , Conexinas/metabolismo , Uniones Comunicantes/ultraestructura , Masculino , Potenciales de la Membrana/fisiología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Plasticidad Neuronal/fisiología , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Ratas , Ratas Wistar , Estrés Psicológico/fisiopatología , Regulación hacia Arriba/fisiología , Proteína de la Zonula Occludens-1 , Proteína delta-6 de Union Comunicante
13.
Endocrinology ; 149(6): 2840-52, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18292192

RESUMEN

Chromaffin cells of the adrenal medulla elaborate and secrete catecholamines and neuropeptides for hormonal and paracrine signaling in stress and during inflammation. We have recently documented the action of the cytokine TNF-alpha on neuropeptide secretion and biosynthesis in isolated bovine chromaffin cells. Here, we demonstrate that the type 2 TNF-alpha receptor (TNF-R2) mediates TNF-alpha signaling in chromaffin cells via activation of nuclear factor (NF)-kappaB. Microarray and suppression subtractive hybridization have been used to identify TNF-alpha target genes in addition to those encoding the neuropeptides galanin, vasoactive intestinal polypeptide, and secretogranin II in chromaffin cells. TNF-alpha, acting through the TNF-R2, causes an early up-regulation of NF-kappaB, long-lasting induction of the NF-kappaB target gene inhibitor kappaB (IkappaB), and persistent stimulation of other NF-kappaB-associated genes including mitogen-inducible gene-6 (MIG-6), which acts as an IkappaB signaling antagonist, and butyrate-induced transcript 1. Consistent with long-term activation of the NF-kappaB signaling pathway, delayed induction of neuropeptide gene transcription by TNF-alpha in chromaffin cells is blocked by an antagonist of NF-kappaB signaling. TNF-alpha-dependent signaling in neuroendocrine cells thus leads to a unique, persistent mode of NF-kappaB activation that features long-lasting transcription of both IkappaB and MIG-6, which may play a role in the long-lasting effects of TNF-alpha in regulating neuropeptide output from the adrenal, a potentially important feedback station for modulating long-term cytokine effects in inflammation.


Asunto(s)
Células Cromafines/fisiología , Inflamación/fisiopatología , FN-kappa B/fisiología , Neuropéptidos/genética , Transducción de Señal/fisiología , Factor 2 Asociado a Receptor de TNF/fisiología , Factor de Necrosis Tumoral alfa/farmacología , Glándulas Suprarrenales/citología , Glándulas Suprarrenales/fisiología , Animales , Bovinos , Células Cromafines/efectos de los fármacos , Regulación de la Expresión Génica , Humanos , Hibridación in Situ , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN/genética , Proteínas Recombinantes/farmacología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos
14.
Front Neuroendocrinol ; 29(1): 128-41, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18048093

RESUMEN

Sympathetic neurons and chromaffin cells derive from common sympathoadrenal precursors which arise from the neural crest. Cells from this lineage migrate to their final destination and differentiate by acquiring a catecholaminergic phenotype in response to different environmental factors. It has been shown that the neuropeptide pituitary adenylate cyclase-activating polypeptide (PACAP) and its PAC1 receptor are expressed at early stages of sympathetic development, and participate to the control of neuroblast proliferation and differentiation. PACAP also acts as a neurotransmitter to stimulate catecholamine and neuropeptide biosynthesis and release from sympathetic neurons and chromaffin cells, during development and in adulthood. In addition, PACAP and its receptors have been described in neuroblastoma and pheochromocytoma, and the neuropeptide regulates the differentiation and activity of sympathoadrenal-derived tumoral cell lines, suggestive of an important role in the pathophysiology of the sympathoadrenal lineage. Transcriptome studies uncovered genes and pathways of known and unknown roles that underlie the effects of PACAP in the sympathoadrenal system.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales/patología , Glándulas Suprarrenales/fisiología , Neuroblastoma/patología , Feocromocitoma/patología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/fisiología , Sistema Nervioso Simpático/fisiología , Neoplasias de las Glándulas Suprarrenales/genética , Glándulas Suprarrenales/metabolismo , Animales , Diferenciación Celular , Células Cromafines/metabolismo , Células Cromafines/fisiología , Regulación Neoplásica de la Expresión Génica , Humanos , Neuroblastoma/genética , Feocromocitoma/genética , Sistema Nervioso Simpático/metabolismo
15.
J Clin Endocrinol Metab ; 92(12): 4865-72, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17878247

RESUMEN

CONTEXT: Pheochromocytomas are catecholamine-producing tumors that are generally benign but that can also present as or develop into malignancy. Occurrence of malignant pheochromocytomas can only be asserted by imaging of metastatic lesions. OBJECTIVES: We conducted a gene expression profiling of benign and malignant tumors to identify a gene signature that would allow us to discriminate benign from malignant pheochromocytomas and to gain a better understanding of tumorigenic pathways associated with malignancy. DESIGN: A total of 36 patients with pheochromocytoma was studied retrospectively. There were 18 (nine benign and nine malignant) tumors used for gene expression profiling on pangenomic oligonucleotide microarrays. RESULTS: We identified and validated a set of predictor genes that could accurately distinguish the two tumor subtypes through unsupervised clustering. Most of the differentially expressed genes were down-regulated in malignant tumors, and several of these genes encoded neuroendocrine factors involved in prominent characteristics of chromaffin cell biology. In particular, the expression of two key processing enzymes of trophic peptides, peptidylglycine alpha-amidating monooxygenase and glutaminyl-peptide cyclotransferase, was reduced in malignant pheochromocytomas. CONCLUSION: The gene expression profiling of benign and malignant pheochromocytomas clearly identified a set of genes that could be used as a prognostic multi-marker and revealed that the expression of several genes encoding neuroendocrine proteins was reduced in malignant compared with benign tumors.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales/genética , Neoplasias de las Glándulas Suprarrenales/patología , Feocromocitoma/genética , Feocromocitoma/patología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Niño , Preescolar , Femenino , Marcadores Genéticos , Humanos , Lactante , Masculino , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Neoplásico/biosíntesis , ARN Neoplásico/genética , Reproducibilidad de los Resultados , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
16.
Ann N Y Acad Sci ; 1073: 527-32, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17102121

RESUMEN

The aim of the present study was to compare the expression levels of secretogranin II (SgII), prohormone convertases (PC)1 and PC2, and the proteolytic processing of SgII in benign versus malignant pheochromocytomas. Quantitative (Q)-PCR experiments indicated that SgII, PC1, and PC2 mRNAs were overexpressed in pheochromocytoma compared to non-tumoral chromaffin cells (P<0.001) and in benign compared to malignant tumors (P<0.01). Western blot analysis using a human SgII antiserum revealed the occurrence of a 97-kDa band corresponding to the expected size of SgII, with significantly higher quantities in benign than in malignant tumors (P<0.05). Using antisera directed against sequential regions of SgII (N-terminal, secretoneurin [SN], EM66, internal, and C-terminal sequences), we observed distinct processing profiles between benign and malignant pheochromocytomas. In contrast, using PC1 and PC2 antisera no differences between the two types of tumors were found. RIA measurement showed that EM66 median values between benign and malignant chromaffin cell tumors were significantly different (128.5 vs. 6.3 ng/mg protein, respectively; P<0.001). Taken together, these results indicate that, in pheochromocytoma, malignancy is associated with reduced PC1, PC2, and SgII mRNA expression and decreased levels of processing products of SgII, in line with the low concentrations of EM66 that occur in malignant tumors. These data support the notion that SgII-processing products, such as EM66, could represent prognostic markers of pheochromocytomas.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales/diagnóstico , Perfilación de la Expresión Génica , Feocromocitoma/diagnóstico , Neoplasias de las Glándulas Suprarrenales/patología , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Feocromocitoma/patología , Pronóstico , Radioinmunoensayo
17.
Ann N Y Acad Sci ; 1073: 533-40, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17102122

RESUMEN

Pheochromocytomas (PHEOs) are rare catecholamine-producing neoplasias that arise from chromaffin cells of the adrenal medulla or from extra-adrenal locations. These neuroendocrine tumors are usually benign, but may also present as or develop into a malignancy. There are currently no means to predict or to cure malignant tumors which have a poor prognosis. We have recently validated several assays for the measurement of peptides derived from chromogranin A (CgA) and secretogranin II (SgII) in order to determine whether these secreted neuroendocrine products could provide useful, complementary markers for the diagnosis and prognosis of PHEOs. Both the CgA-derived peptide WE14 and the SgII-derived peptide EM66 proved to be sensitive circulating markers for the diagnosis of PHEO. In addition, much higher EM66 levels were measured in benign than in malignant tumoral tissues, suggesting that this peptide could represent a valuable tool for the prognosis of PHEO. We have also initiated a comparative microarray study of benign and malignant PHEOs, which allowed the identification of a set of about 100 genes that were differentially expressed and best discriminated the two types of tumors. A large majority of these genes were expressed at lower levels in the malignant disease and were associated with various characteristics of chromaffin cells, such as hormone secretion signaling and machinery, peptide maturation, and cellular morphology. Altogether, these studies provide novel tools for the management of PHEO, and new insights for the understanding of tumorigenesis in chromaffin cells, which may offer potential therapeutic strategies.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales/genética , Perfilación de la Expresión Génica , Feocromocitoma/genética , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos
18.
Regul Pept ; 137(1-2): 89-94, 2006 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-16928405

RESUMEN

PACAP inhibits cell proliferation and promotes cell survival and neurite outgrowth of pheochromocytoma PC12 cells. Transcriptome analysis of PACAP-treated PC12 cells allowed to identify potential genes implicated in this differentiation process. Among the genes whose expression is up-regulated by PACAP, we identified the Inhibitor of DNA binding 3 (Id3). Id3 is a member of the helix-loop-helix (HLH) family of transcription factors which acts as a negative dominant inhibitor of basic HLH factors. Time-course studies revealed that Id3 is an early PACAP response gene (8-fold after 1 h of stimulation), and that the up-regulation of its expression persists over 12 h after the onset of PACAP treatment. The stimulatory effect of PACAP on Id3 mRNA levels was mimicked by adenylate cyclase/PKA activators like forskolin and dibutyryl cyclic AMP. Moreover, PACAP-induced Id3 gene expression was inhibited by phosphatidylinositol 3'-OH-kinase and p38 MAP kinase blockers. Northern blot analysis of Id3 distribution in rat tissues showed a strong expression of this gene in the adrenal medulla. Overexpression of Id3 increased the number of living PC12 cells, in basal condition and after exposure to oxidative stress. These results indicate that Id3 is a cAMP-responsive gene whose up-regulation could be involved in PACAP-induced pro-survival signaling during sympathoadrenal cell differentiation.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Proteínas Inhibidoras de la Diferenciación/fisiología , Polipéptido Hipofisario Activador de la Adenilato-Ciclasa/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Secuencia de Bases , Northern Blotting , Cartilla de ADN , Células PC12 , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...