Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Nat Commun ; 13(1): 7858, 2022 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-36543805

RESUMEN

SUMOylation is a dynamic posttranslational modification, that provides fine-tuning of protein function involved in the cellular response to stress, differentiation, and tissue development. In the adrenal cortex, an emblematic endocrine organ that mediates adaptation to physiological demands, the SUMOylation gradient is inversely correlated with the gradient of cellular differentiation raising important questions about its role in functional zonation and the response to stress. Considering that SUMO-specific protease 2 (SENP2), a deSUMOylating enzyme, is upregulated by Adrenocorticotropic Hormone (ACTH)/cAMP-dependent Protein Kinase (PKA) signalling within the zona fasciculata, we generated mice with adrenal-specific Senp2 loss to address these questions. Disruption of SENP2 activity in steroidogenic cells leads to specific hypoplasia of the zona fasciculata, a blunted reponse to ACTH and isolated glucocorticoid deficiency. Mechanistically, overSUMOylation resulting from SENP2 loss shifts the balance between ACTH/PKA and WNT/ß-catenin signalling leading to repression of PKA activity and ectopic activation of ß-catenin. At the cellular level, this blocks transdifferentiation of ß-catenin-positive zona glomerulosa cells into fasciculata cells and sensitises them to premature apoptosis. Our findings indicate that the SUMO pathway is critical for adrenal homeostasis and stress responsiveness.


Asunto(s)
Transdiferenciación Celular , Cisteína Endopeptidasas , Glucocorticoides , Animales , Ratones , Corteza Suprarrenal/metabolismo , Corticoesteroides/metabolismo , Hormona Adrenocorticotrópica/metabolismo , beta Catenina/metabolismo , Transdiferenciación Celular/genética , Cisteína Endopeptidasas/genética , Cisteína Endopeptidasas/metabolismo , Glucocorticoides/metabolismo , Vía de Señalización Wnt
2.
Sci Adv ; 8(41): eadd0422, 2022 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-36240276

RESUMEN

Unlike most cancers, adrenocortical carcinomas (ACCs) are more frequent in women than in men, but the underlying mechanisms of this sexual dimorphism remain elusive. Here, we show that inactivation of Znrf3 in the mouse adrenal cortex, recapitulating the most frequent alteration in ACC patients, is associated with sexually dimorphic tumor progression. Although female knockouts develop metastatic carcinomas at 18 months, adrenal hyperplasia regresses in male knockouts. This male-specific phenotype is associated with androgen-dependent induction of senescence, recruitment, and differentiation of highly phagocytic macrophages that clear out senescent cells. In contrast, in females, macrophage recruitment is delayed and dampened, which allows for aggressive tumor progression. Consistently, analysis of TCGA-ACC data shows that phagocytic macrophages are more prominent in men and are associated with better prognosis. Together, these data show that phagocytic macrophages are key players in the sexual dimorphism of ACC that could be previously unidentified allies in the fight against this devastating cancer.


Asunto(s)
Neoplasias de la Corteza Suprarrenal , Carcinoma Corticosuprarrenal , Neoplasias de la Corteza Suprarrenal/genética , Neoplasias de la Corteza Suprarrenal/patología , Carcinoma Corticosuprarrenal/genética , Carcinoma Corticosuprarrenal/patología , Andrógenos , Animales , Femenino , Masculino , Ratones , Pronóstico
3.
J Clin Invest ; 131(23)2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34850745

RESUMEN

Large-cell calcifying Sertoli cell tumors (LCCSCTs) are among the most frequent lesions occurring in male Carney complex (CNC) patients. Although they constitute a key diagnostic criterion for this rare multiple neoplasia syndrome resulting from inactivating mutations of the tumor suppressor PRKAR1A, leading to unrepressed PKA activity, LCCSCT pathogenesis and origin remain elusive. Mouse models targeting Prkar1a inactivation in all somatic populations or separately in each cell type were generated to decipher the molecular and paracrine networks involved in the induction of CNC testis lesions. We demonstrate that the Prkar1a mutation was required in both stromal and Sertoli cells for the occurrence of LCCSCTs. Integrative analyses comparing transcriptomic, immunohistological data and phenotype of mutant mouse combinations led to the understanding of human LCCSCT pathogenesis and demonstrated PKA-induced paracrine molecular circuits in which the aberrant WNT4 signal production is a limiting step in shaping intratubular lesions and tumor expansion both in a mouse model and in human CNC testes.


Asunto(s)
Complejo de Carney/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Células de Sertoli/citología , Neoplasias Testiculares/metabolismo , Proteína Wnt4/metabolismo , Animales , Apoptosis , Complejo de Carney/genética , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Genes Supresores de Tumor , Humanos , Masculino , Ratones , Ratones Noqueados , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Comunicación Paracrina , Fenotipo , Pigmentación , Túbulos Seminíferos/metabolismo , Testículo/metabolismo , Transcriptoma
4.
Br J Cancer ; 121(5): 384-394, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31363169

RESUMEN

BACKGROUND: EZH2 is overexpressed and associated with poor prognosis in adrenocortical carcinoma (ACC) and its inhibition reduces growth and aggressiveness of ACC cells in culture. Although EZH2 was identified as the methyltransferase that deposits the repressive H3K27me3 histone mark, it can cooperate with transcription factors to stimulate gene transcription. METHODS: We used bioinformatics approaches on gene expression data from three cohorts of patients and a mouse model of EZH2 ablation, to identify targets and mode of action of EZH2 in ACC. This was followed by ChIP and functional assays to evaluate contribution of identified targets to ACC pathogenesis. RESULTS: We show that EZH2 mostly works as a transcriptional inducer in ACC, through cooperation with the transcription factor E2F1 and identify three positive targets involved in cell cycle regulation and mitosis i.e., RRM2, PTTG1 and ASE1/PRC1. Overexpression of these genes is associated with poor prognosis, suggesting a potential role in acquisition of aggressive ACC features. Pharmacological and siRNA-mediated inhibition of RRM2 blocks cell proliferation, induces apoptosis and inhibits cell migration, suggesting that it may be an interesting target in ACC. CONCLUSIONS: Altogether, these data show an unexpected role of EZH2 and E2F1 in stimulating expression of genes associated with ACC aggressiveness.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/genética , Carcinoma Corticosuprarrenal/genética , Factor de Transcripción E2F1/genética , Proteína Potenciadora del Homólogo Zeste 2/genética , Regulación Neoplásica de la Expresión Génica , Adenosina/análogos & derivados , Adenosina/farmacología , Animales , Proteínas de Ciclo Celular/genética , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Inmunoprecipitación de Cromatina , Biología Computacional , Proteína Potenciadora del Homólogo Zeste 2/antagonistas & inhibidores , Humanos , Indoles/farmacología , Ratones Noqueados , Análisis Multivariante , Modelos de Riesgos Proporcionales , Ribonucleósido Difosfato Reductasa/antagonistas & inhibidores , Ribonucleósido Difosfato Reductasa/genética , Securina/genética
5.
FASEB J ; 33(9): 10218-10230, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31208233

RESUMEN

SUMOylation is a highly conserved and dynamic post-translational mechanism primarily affecting nuclear programs for adapting organisms to stressful challenges. Alteration of SUMOylation cycles leads to severe developmental and homeostatic defects and malignancy, but signals coordinating SUMOylation are still unidentified. The adrenal cortex is a zonated endocrine gland that controls body homeostasis and stress response. Here, we show that in human and in mouse adrenals, SUMOylation follows a decreasing centripetal gradient that mirrors cortical differentiation flow and delimits highly and weakly SUMOylated steroidogenic compartments, overlapping glomerulosa, and fasciculata zones. Activation of PKA signaling by acute hormonal treatment, mouse genetic engineering, or in Carney complex results in repression of small ubiquitin-like modifier (SUMO) conjugation in the inner cortex by coordinating expression of SUMO pathway inducers and repressors. Conversely, genetic activation of canonical wingless-related integration site signaling maintains high SUMOylation potential in the outer neoplastic cortex. Thus, SUMOylation is tightly regulated by signaling pathways that orchestrate adrenal zonation and diseases.-Dumontet, T., Sahut-Barnola, I., Dufour, D., Lefrançois-Martinez, A.-M., Berthon, A., Montanier, N., Ragazzon, B., Djari, C., Pointud, J.-C., Roucher-Boulez, F., Batisse-Lignier, M., Tauveron, I., Bertherat, J., Val, P., Martinez, A. Hormonal and spatial control of SUMOylation in the human and mouse adrenal cortex.


Asunto(s)
Corteza Suprarrenal/metabolismo , Hormona Adrenocorticotrópica/farmacología , Procesamiento Proteico-Postraduccional/fisiología , Sumoilación/fisiología , Corteza Suprarrenal/efectos de los fármacos , Corteza Suprarrenal/ultraestructura , Neoplasias de la Corteza Suprarrenal/patología , Hormona Adrenocorticotrópica/administración & dosificación , Animales , Complejo de Carney/metabolismo , Línea Celular Tumoral , Colforsina/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Cicloheximida/farmacología , Dactinomicina/farmacología , Preparaciones de Acción Retardada , Dexametasona/análogos & derivados , Dexametasona/farmacología , Femenino , Humanos , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas de Neoplasias/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Sumoilación/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos , Vía de Señalización Wnt/fisiología , Zona Fascicular/efectos de los fármacos , Zona Fascicular/metabolismo , Zona Glomerular/efectos de los fármacos , Zona Glomerular/metabolismo , beta Catenina/deficiencia , beta Catenina/genética
6.
Proc Natl Acad Sci U S A ; 115(52): E12265-E12274, 2018 12 26.
Artículo en Inglés | MEDLINE | ID: mdl-30541888

RESUMEN

Adrenal cortex steroids are essential for body homeostasis, and adrenal insufficiency is a life-threatening condition. Adrenal endocrine activity is maintained through recruitment of subcapsular progenitor cells that follow a unidirectional differentiation path from zona glomerulosa to zona fasciculata (zF). Here, we show that this unidirectionality is ensured by the histone methyltransferase EZH2. Indeed, we demonstrate that EZH2 maintains adrenal steroidogenic cell differentiation by preventing expression of GATA4 and WT1 that cause abnormal dedifferentiation to a progenitor-like state in Ezh2 KO adrenals. EZH2 further ensures normal cortical differentiation by programming cells for optimal response to adrenocorticotrophic hormone (ACTH)/PKA signaling. This is achieved by repression of phosphodiesterases PDE1B, 3A, and 7A and of PRKAR1B. Consequently, EZH2 ablation results in blunted zF differentiation and primary glucocorticoid insufficiency. These data demonstrate an all-encompassing role for EZH2 in programming steroidogenic cells for optimal response to differentiation signals and in maintaining their differentiated state.


Asunto(s)
Corteza Suprarrenal/enzimología , Subunidad RIbeta de la Proteína Quinasa Dependiente de AMP Cíclico/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Transducción de Señal , Corteza Suprarrenal/metabolismo , Animales , Diferenciación Celular , Subunidad RIbeta de la Proteína Quinasa Dependiente de AMP Cíclico/genética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 1/genética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 1/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/genética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/metabolismo , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 7/genética , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 7/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/genética , Femenino , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Esteroides/metabolismo , Zona Fascicular/citología , Zona Fascicular/enzimología , Zona Fascicular/metabolismo , Zona Glomerular/citología , Zona Glomerular/enzimología , Zona Glomerular/metabolismo
7.
Ann Endocrinol (Paris) ; 79(3): 95-97, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29673697

RESUMEN

The adrenocortical gland undergoes structural and functional remodelling in the fetal and postnatal periods. After birth, the fetal zone of the gland undergoes rapid involution in favor of the definitive cortex, which reaches maturity with the emergence of the zona reticularis(zR) at the adrenarche. The mechanisms underlying the adrenarche, the process leading to pre-puberty elevation of plasma androgens in higher primates, remain unknown, largely due to lack of any experimental model. By following up fetal and definitive cortex cell lines in mice, we showed that activation of protein kinase A (PKA) signaling mainly impacts the adult cortex by stimulating centripetal regeneration, with differentiation and then conversion of the zona fasciculata into a functional zR. Animals developed Cushing syndrome associated with primary hyperaldosteronism, suggesting possible coexistence of these hypersecretions in certain patients. Remarkably, all of these traits were sex-dependent: testicular androgens promoted WNT signaling antagonism on PKA, slowing cortical renewal and delaying onset of Cushing syndrome and the establishment of the zR in male mice, this being corrected by orchidectomy. In conclusion, zR derives from centripetal conversion of the zona fasciculata under cellular renewal induced by PKA signaling, determining the size of the adult cortex. Finally, we demonstrated that this PKA-dependent mobilization of cortical progenitors is sexually dimorphic and could, if confirmed in humans, account for female preponderance in adrenocortical pathologies.


Asunto(s)
Corteza Suprarrenal/embriología , Corteza Suprarrenal/crecimiento & desarrollo , Ratones , Modelos Animales , Glándulas Suprarrenales/embriología , Glándulas Suprarrenales/crecimiento & desarrollo , Animales , Diferenciación Celular , Femenino , Humanos , Masculino , Ratones Noqueados , Maduración Sexual/fisiología
8.
JCI Insight ; 3(2)2018 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-29367455

RESUMEN

The adrenal cortex undergoes remodeling during fetal and postnatal life. How zona reticularis emerges in the postnatal gland to support adrenarche, a process whereby higher primates increase prepubertal androgen secretion, is unknown. Using cell-fate mapping and gene deletion studies in mice, we show that activation of PKA has no effect on the fetal cortex, while it accelerates regeneration of the adult cortex, triggers zona fasciculata differentiation that is subsequently converted into a functional reticularis-like zone, and drives hypersecretion syndromes. Remarkably, PKA effects are influenced by sex. Indeed, testicular androgens increase WNT signaling that antagonizes PKA, leading to slower adrenocortical cell turnover and delayed phenotype whereas gonadectomy sensitizes males to hypercorticism and reticularis-like formation. Thus, reticularis results from ultimate centripetal conversion of adult cortex under the combined effects of PKA and cell turnover that dictate organ size. We show that PKA-induced progenitor recruitment is sexually dimorphic and may provide a paradigm for overrepresentation of women in adrenal diseases.


Asunto(s)
Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/metabolismo , Caracteres Sexuales , Transducción de Señal/fisiología , Zona Fascicular/metabolismo , Zona Reticular/metabolismo , Adrenarquia/metabolismo , Factores de Edad , Andrógenos/metabolismo , Animales , Diferenciación Celular/fisiología , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/genética , Embrión de Mamíferos , Femenino , Masculino , Ratones , Ratones Noqueados , Modelos Animales
9.
Nat Commun ; 7: 12751, 2016 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-27624192

RESUMEN

Adrenal cortex physiology relies on functional zonation, essential for production of aldosterone by outer zona glomerulosa (ZG) and glucocorticoids by inner zona fasciculata (ZF). The cortex undergoes constant cell renewal, involving recruitment of subcapsular progenitors to ZG fate and subsequent lineage conversion to ZF identity. Here we show that WNT4 is an important driver of WNT pathway activation and subsequent ZG differentiation and demonstrate that PKA activation prevents ZG differentiation through WNT4 repression and WNT pathway inhibition. This suggests that PKA activation in ZF is a key driver of WNT inhibition and lineage conversion. Furthermore, we provide evidence that constitutive PKA activation inhibits, whereas partial inactivation of PKA catalytic activity stimulates ß-catenin-induced tumorigenesis. Together, both lower PKA activity and higher WNT pathway activity lead to poorer prognosis in adrenocortical carcinoma (ACC) patients. These observations suggest that PKA acts as a tumour suppressor in the adrenal cortex, through repression of WNT signalling.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales/etiología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Vía de Señalización Wnt , Zona Fascicular/metabolismo , Zona Glomerular/metabolismo , Animales , Carcinogénesis , Diferenciación Celular , Línea Celular Tumoral , Femenino , Humanos , Ratones , Fosforilación , Zona Fascicular/citología , Zona Glomerular/citología , beta Catenina/metabolismo
10.
Artículo en Inglés | MEDLINE | ID: mdl-27499746

RESUMEN

Aldose reductase (AKR1B) proteins are monomeric enzymes, belonging to the aldo-keto reductase (AKR) superfamily. They perform oxidoreduction of carbonyl groups from a wide variety of substrates, such as aliphatic and aromatic aldehydes or ketones. Due to the involvement of human aldose reductases in pathologies, such as diabetic complications and cancer, AKR1B subgroup enzymatic properties have been extensively characterized. However, the issue of AKR1B function in non-pathologic conditions remains poorly resolved. Adrenal activities generated large amount of harmful aldehydes from lipid peroxidation and steroidogenesis, including 4-hydroxynonenal (4-HNE) and isocaproaldehyde (4-methylpentanal), which can both be reduced by AKR1B proteins. More recently, some AKR1B isoforms have been shown to be endowed with prostaglandin F synthase (PGFS) activity, suggesting that, in addition to possible scavenger function, they could instigate paracrine signals. Interestingly, the adrenal gland is one of the major sites for human and murine AKR1B expression, suggesting that their detoxifying/signaling activity could be specifically required for the correct handling of adrenal function. Moreover, chronic effects of ACTH result in a coordinated regulation of genes encoding the steroidogenic enzymes and some AKR1B isoforms. This review presents the molecular mechanisms accounting for the adrenal-specific expression of some AKR1B genes. Using data from recent mouse genetic models, we will try to connect their enzymatic properties and regulation with adrenal functions.

11.
Hum Mol Genet ; 25(13): 2789-2800, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27149985

RESUMEN

Adrenal Cortex Carcinoma (ACC) is an aggressive tumour with poor prognosis. Common alterations in patients include constitutive WNT/ß-catenin signalling and overexpression of the growth factor IGF2. However, the combination of both alterations in transgenic mice is not sufficient to trigger malignant tumour progression, suggesting that other alterations are required to allow development of carcinomas. Here, we have conducted a study of publicly available gene expression data from three cohorts of ACC patients to identify relevant alterations. Our data show that the histone methyltransferase EZH2 is overexpressed in ACC in the three cohorts. This overexpression is the result of deregulated P53/RB/E2F pathway activity and is associated with increased proliferation and poorer prognosis in patients. Inhibition of EZH2 by RNA interference or pharmacological treatment with DZNep inhibits cellular growth, wound healing and clonogenic growth and induces apoptosis of H295R cells in culture. Further growth inhibition is obtained when DZNep is combined with mitotane, the gold-standard treatment for ACC. Altogether, these observations suggest that overexpression of EZH2 is associated with aggressive progression and may constitute an interesting therapeutic target in the context of ACC.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/genética , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Neoplasias de la Corteza Suprarrenal/metabolismo , Animales , Línea Celular Tumoral , Proliferación Celular , Bases de Datos de Ácidos Nucleicos , Progresión de la Enfermedad , Expresión Génica , Predisposición Genética a la Enfermedad/genética , Humanos , Factor II del Crecimiento Similar a la Insulina/genética , Factor II del Crecimiento Similar a la Insulina/metabolismo , Ratones , Ratones Transgénicos , Interferencia de ARN , Factores de Riesgo , Vía de Señalización Wnt , beta Catenina/genética
12.
Endocrinology ; 156(5): 1671-84, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25730106

RESUMEN

Aldose reductases (AKR1B) are widely expressed oxidoreductases whose physiological function remains elusive. Some isoforms are genuine prostaglandin F2α (PGF2α) synthases, suggesting they might influence adipose homeostasis because PGF2α inhibits adipogenesis. This was shown by Akr1b7 gene ablation in the mouse, which resulted in increased adiposity related to a lower PGF2α content in fat. Yet humans have no ortholog gene for Akr1b7, so the role of aldose reductases in human adipose homeostasis remains to be explored. We analyzed expression of genes encoding human and mouse aldose reductase isoforms in adipose tissues and differentiating adipocytes to assess conserved mechanisms regulating PGF2α synthesis and adipogenesis. The Akr1b3 gene encoded the most abundant isoform in mouse adipose tissue, whereas Akr1b7 encoded the only isoform enriched in the stromal vascular fraction. Most mouse aldose reductase gene expression peaked in early adipogenesis of 3T3-L1 cells and diminished with differentiation. In contrast with its mouse ortholog Akr1b3, AKR1B1 expression increased throughout differentiation of human multipotent adipose-derived stem cells, paralleling PGF2α release, whereas PGF2α receptor (FP) levels collapsed in early differentiation. Pharmacological inhibition of aldose reductase using Statil altered PGF2α production and enhanced human multipotent adipose-derived stem adipocyte differentiation. As expected, the adipogenic effects of Statil were counteracted by an FP agonist (cloprostenol). Thus, in both species aldose reductase-dependent PGF2α production could be important in early differentiation to restrict adipogenesis. PGF2α antiadipogenic signaling could then be toned down through the FP receptor or aldose reductases down-regulation in human and mouse cells, respectively. Our data suggest that aldose reductase inhibitors could have obesogenic potential.


Asunto(s)
Adipocitos/metabolismo , Aldehído Reductasa/genética , Diferenciación Celular/genética , Dinoprost/biosíntesis , Hidroxiprostaglandina Deshidrogenasas/genética , ARN Mensajero/metabolismo , Células 3T3-L1 , Adipogénesis/genética , Tejido Adiposo/metabolismo , Adulto , Aldehído Reductasa/antagonistas & inhibidores , Aldehído Reductasa/metabolismo , Animales , Cloprostenol/farmacología , Inhibidores Enzimáticos/farmacología , Perfilación de la Expresión Génica , Humanos , Hidroxiprostaglandina Deshidrogenasas/antagonistas & inhibidores , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Luteolíticos/farmacología , Masculino , Ratones , Persona de Mediana Edad , Células Madre Multipotentes , Obesidad/metabolismo , Ftalazinas/farmacología , Receptores de Prostaglandina/agonistas , Receptores de Prostaglandina/genética , Receptores de Prostaglandina/metabolismo , Grasa Subcutánea Abdominal/metabolismo
13.
Hum Mol Genet ; 23(20): 5418-28, 2014 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-24865460

RESUMEN

Primary pigmented nodular adrenocortical disease (PPNAD) is associated with inactivating mutations of the PRKAR1A tumor suppressor gene that encodes the regulatory subunit R1α of the cAMP-dependent protein kinase (PKA). In human and mouse adrenocortical cells, these mutations lead to increased PKA activity, which results in increased resistance to apoptosis that contributes to the tumorigenic process. We used in vitro and in vivo models to investigate the possibility of a crosstalk between PKA and mammalian target of rapamycin (mTOR) pathways in adrenocortical cells and its possible involvement in apoptosis resistance. Impact of PKA signaling on activation of the mTOR pathway and apoptosis was measured in a mouse model of PPNAD (AdKO mice), in human and mouse adrenocortical cell lines in response to pharmacological inhibitors and in PPNAD tissues by immunohistochemistry. AdKO mice showed increased mTOR complex 1 (mTORC1) pathway activity. Inhibition of mTORC1 by rapamycin restored sensitivity of adrenocortical cells to apoptosis in AdKO but not in wild-type mice. In both cell lines and mouse adrenals, rapid phosphorylation of mTORC1 targets including BAD proapoptotic protein was observed in response to PKA activation. Accordingly, BAD hyperphosphorylation, which inhibits its proapoptotic activity, was increased in both AdKO mouse adrenals and human PPNAD tissues. In conclusion, mTORC1 pathway is activated by PKA signaling in human and mouse adrenocortical cells, leading to increased cell survival, which is correlated with BAD hyperphosphorylation. These alterations could be causative of tumor formation.


Asunto(s)
Enfermedades de la Corteza Suprarrenal/metabolismo , Enfermedades de la Corteza Suprarrenal/patología , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/genética , Proteína Letal Asociada a bcl/metabolismo , Enfermedades de la Corteza Suprarrenal/genética , Hormona Adrenocorticotrópica/administración & dosificación , Hormona Adrenocorticotrópica/farmacología , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/metabolismo , Modelos Animales de Enfermedad , Técnicas de Inactivación de Genes , Humanos , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Complejos Multiproteicos/metabolismo , Fosforilación , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/metabolismo
14.
Diabetes ; 61(11): 2796-806, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22851578

RESUMEN

Negative regulators of white adipose tissue (WAT) expansion are poorly documented in vivo. Prostaglandin F(2α) (PGF(2α)) is a potent antiadipogenic factor in cultured preadipocytes, but evidence for its involvement in physiological context is lacking. We previously reported that Akr1b7, an aldo-keto reductase enriched in adipose stromal vascular fraction but absent from mature adipocytes, has antiadipogenic properties possibly supported by PGF(2α) synthase activity. To test whether lack of Akr1b7 could influence WAT homeostasis in vivo, we generated Akr1b7(-/-) mice in 129/Sv background. Akr1b7(-/-) mice displayed excessive basal adiposity resulting from adipocyte hyperplasia/hypertrophy and exhibited greater sensitivity to diet-induced obesity. Following adipose enlargement and irrespective of the diet, they developed liver steatosis and progressive insulin resistance. Akr1b7 loss was associated with decreased PGF(2α) WAT contents. Cloprostenol (PGF(2α) agonist) administration to Akr1b7(-/-) mice normalized WAT expansion by affecting both de novo adipocyte differentiation and size. Treatment of 3T3-L1 adipocytes and Akr1b7(-/-) mice with cloprostenol suggested that decreased adipocyte size resulted from inhibition of lipogenic gene expression. Hence, Akr1b7 is a major regulator of WAT development through at least two PGF(2α)-dependent mechanisms: inhibition of adipogenesis and lipogenesis. These findings provide molecular rationale to explore the status of aldo-keto reductases in dysregulations of adipose tissue homeostasis.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Adiposidad , Aldehído Reductasa/metabolismo , Dieta Alta en Grasa/efectos adversos , Dinoprost/metabolismo , Regulación hacia Abajo , Obesidad/metabolismo , Células 3T3-L1 , Adipogénesis/efectos de los fármacos , Tejido Adiposo Blanco/efectos de los fármacos , Tejido Adiposo Blanco/patología , Adiposidad/efectos de los fármacos , Aldehído Reductasa/genética , Animales , Fármacos Antiobesidad/farmacología , Fármacos Antiobesidad/uso terapéutico , Tamaño de la Célula/efectos de los fármacos , Cloprostenol/farmacología , Cloprostenol/uso terapéutico , Cruzamientos Genéticos , Dinoprost/agonistas , Susceptibilidad a Enfermedades , Regulación hacia Abajo/efectos de los fármacos , Resistencia a la Insulina , Lipogénesis/efectos de los fármacos , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Terapia Molecular Dirigida , Obesidad/tratamiento farmacológico , Obesidad/etiología , Obesidad/patología
15.
Front Pharmacol ; 3: 148, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22876234

RESUMEN

The aldose reductase (AR; human AKR1B1/mouse Akr1b3) has been the focus of many research because of its role in diabetic complications. The starting point of these alterations is the massive entry of glucose in polyol pathway where it is converted into sorbitol by this enzyme. However, the issue of AR function in non-diabetic condition remains unresolved. AR-like enzymes (AKR1B10, Akr1b7, and Akr1b8) are highly related isoforms often co-expressed with bona fide AR, making functional analysis of one or the other isoform a challenging task. AKR1B/Akr1b members share at least 65% protein identity and the general ability to reduce many redundant substrates such as aldehydes provided from lipid peroxidation, steroids and their by-products, and xenobiotics in vitro. Based on these properties, AKR1B/Akr1b are generally considered as detoxifying enzymes. Considering that divergences should be more informative than similarities to help understanding their physiological functions, we chose to review specific hallmarks of each human/mouse isoforms by focusing on tissue distribution and specific mechanisms of gene regulation. Indeed, although the AR shows ubiquitous expression, AR-like proteins exhibit tissue-specific patterns of expression. We focused on three organs where certain isoforms are enriched, the adrenal gland, enterohepatic, and adipose tissues and tried to connect recent enzymatic and regulation data with endocrine and metabolic functions of these organs. We presented recent mouse models showing unsuspected physiological functions in the regulation of glucido-lipidic metabolism and adipose tissue homeostasis. Beyond the widely accepted idea that AKR1B/Akr1b are detoxification enzymes, these recent reports provide growing evidences that they are able to modify or generate signal molecules. This conceptually shifts this class of enzymes from unenviable status of scavenger to upper class of messengers.

16.
J Biol Chem ; 286(38): 32976-85, 2011 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-21808064

RESUMEN

In the adrenal gland, adrenocorticotropin (ACTH) acting through the cAMP protein kinase (PKA) transduction pathway is the main regulator of genes involved in glucocorticoid synthesis. The prolactin (PRL) receptor is expressed in the adrenal cortex of most mammals, but experimental proof that PRL ensures direct control on glucocorticoid synthesis in rodents remains elusive. To unravel the physiological importance of PRL in adrenocortical functions, we measured steroidogenic capacity of Prlr-deficient mice (Prlr(-/-)) and explored the influence of JAK/STAT signaling, the major PRL transduction pathway, on the steroidogenic activity of adrenocortical cell cultures. We demonstrate that lack of Prlr does not affect basal (nor stress-induced) corticosterone levels in mice. PRL triggers JAK2/STAT5-dependent transcription in adrenal cells, but this does not influence corticosterone release. In contrast, pharmacological or siRNA-mediated inhibition of JAK2 reveals its essential role in both basal and ACTH/cAMP-induced steroidogenesis. We demonstrate that nuclear JAK2 regulates the amount of active transcription factor CREB (cAMP response element-binding protein) through tyrosine phosphorylation and prevention of proteasomal degradation, which in turn leads to transcriptional activation of the rate-limiting steroidogenic Star gene. Hence, we describe a novel link between PKA and JAK2 by which nuclear JAK2 signaling controls adrenal steroidogenesis by increasing the stability of CREB.


Asunto(s)
Glándulas Suprarrenales/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Janus Quinasa 2/metabolismo , Esteroides/biosíntesis , Transcripción Genética , Glándulas Suprarrenales/efectos de los fármacos , Animales , Núcleo Celular/efectos de los fármacos , Núcleo Celular/enzimología , Corticosterona/biosíntesis , AMP Cíclico/farmacología , Humanos , Masculino , Ratones , Complejos Multiproteicos/metabolismo , Fosforilación/efectos de los fármacos , Prolactina/farmacología , Unión Proteica/efectos de los fármacos , Estabilidad Proteica/efectos de los fármacos , Ratas , Ovinos , Transcripción Genética/efectos de los fármacos
17.
PLoS Genet ; 6(6): e1000980, 2010 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-20548949

RESUMEN

Carney complex (CNC) is an inherited neoplasia syndrome with endocrine overactivity. Its most frequent endocrine manifestation is primary pigmented nodular adrenocortical disease (PPNAD), a bilateral adrenocortical hyperplasia causing pituitary-independent Cushing's syndrome. Inactivating mutations in PRKAR1A, a gene encoding the type 1 alpha-regulatory subunit (R1alpha) of the cAMP-dependent protein kinase (PKA) have been found in 80% of CNC patients with Cushing's syndrome. To demonstrate the implication of R1alpha loss in the initiation and development of PPNAD, we generated mice lacking Prkar1a specifically in the adrenal cortex (AdKO). AdKO mice develop pituitary-independent Cushing's syndrome with increased PKA activity. This leads to autonomous steroidogenic genes expression and deregulated adreno-cortical cells differentiation, increased proliferation and resistance to apoptosis. Unexpectedly, R1alpha loss results in improper maintenance and centrifugal expansion of cortisol-producing fetal adrenocortical cells with concomitant regression of adult cortex. Our data provide the first in vivo evidence that loss of R1alpha is sufficient to induce autonomous adrenal hyper-activity and bilateral hyperplasia, both observed in human PPNAD. Furthermore, this model demonstrates that deregulated PKA activity favors the emergence of a new cell population potentially arising from the fetal adrenal, giving new insight into the mechanisms leading to PPNAD.


Asunto(s)
Corteza Suprarrenal/metabolismo , Síndrome de Cushing/metabolismo , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/metabolismo , Hormona Adrenocorticotrópica/metabolismo , Animales , Proliferación Celular , Síndrome de Cushing/embriología , Síndrome de Cushing/genética , Síndrome de Cushing/patología , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/deficiencia , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Noqueados , Transducción de Señal , Esteroide 17-alfa-Hidroxilasa/metabolismo
18.
PLoS One ; 4(10): e7309, 2009 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-19809495

RESUMEN

Prostaglandin F(2alpha) (PGF(2alpha)), represses ovarian steroidogenesis and initiates parturition in mammals but its impact on adrenal gland is unknown. Prostaglandins biosynthesis depends on the sequential action of upstream cyclooxygenases (COX) and terminal synthases but no PGF(2alpha) synthases (PGFS) were functionally identified in mammalian cells. In vitro, the most efficient mammalian PGFS belong to aldo-keto reductase 1B (AKR1B) family. The adrenal gland is a major site of AKR1B expression in both human (AKR1B1) and mouse (AKR1B3, AKR1B7). Thus, we examined the PGF(2alpha) biosynthetic pathway and its functional impact on both cortical and medullary zones. Both compartments produced PGF(2alpha) but expressed different biosynthetic isozymes. In chromaffin cells, PGF(2alpha) secretion appeared constitutive and correlated to continuous expression of COX1 and AKR1B3. In steroidogenic cells, PGF(2alpha) secretion was stimulated by adrenocorticotropic hormone (ACTH) and correlated to ACTH-responsiveness of both COX2 and AKR1B7/B1. The pivotal role of AKR1B7 in ACTH-induced PGF(2alpha) release and functional coupling with COX2 was demonstrated using over- and down-expression in cell lines. PGF(2alpha) receptor was only detected in chromaffin cells, making medulla the primary target of PGF(2alpha) action. By comparing PGF(2alpha)-responsiveness of isolated cells and whole adrenal cultures, we demonstrated that PGF(2alpha) repressed glucocorticoid secretion by an indirect mechanism involving a decrease in catecholamine release which in turn decreased adrenal steroidogenesis. PGF(2alpha) may be regarded as a negative autocrine/paracrine regulator within a novel intra-adrenal feedback loop. The coordinated cell-specific regulation of COX2 and AKR1B7 ensures the generation of this stress-induced corticostatic signal.


Asunto(s)
Glándulas Suprarrenales/metabolismo , Oxidorreductasas de Alcohol/metabolismo , Aldehído Reductasa/metabolismo , Dinoprost/metabolismo , Regulación Enzimológica de la Expresión Génica , Aldo-Ceto Reductasas , Animales , Sitios de Unión , Células Cromafines/metabolismo , Ciclooxigenasa 2/metabolismo , Sistema Endocrino , Glucocorticoides/metabolismo , Humanos , Masculino , Ratones , Ratas , Ratas Wistar
19.
J Biochem ; 145(2): 161-8, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19010934

RESUMEN

Here, we show that three enzymes belonging to the 1B group of the aldo-keto reductase (AKR) superfamily, i.e., human placental aldose reductase (AKR1B1), mouse kidney aldose reductase (AKR1B3) and mouse vas deferens protein (AKR1B7), catalyse the reduction of prostaglandin (PG) H(2), a common intermediate of various prostanoids, to form PGF(2alpha) in the presence of NADPH. AKR1B1, AKR1B3 and AKR1B7 displayed higher affinities for PGH(2) (K(m) = 1.9, 9.3 and 3.8 microM, respectively) and V(max) values (26, 53 and 44 nmol/min/mg protein, respectively) than did the human lung PGF(2alpha) synthase (AKR1C3; 18 microM and 4 nmol/min/mg protein, respectively). The PGF(2alpha) synthase activity of AKR1B1 and AKR1B3 was efficiently inhibited by two AKR inhibitors, tolrestat (K(i) = 3.6 and 0.26 microM, respectively) and sorbinil (K(i) = 21.7 and 0.89 microM, respectively), in a non-competitive or mixed-type manner, whereas that of AKR1B7 was not sensitive to these inhibitors (K(i) = 9.2 and 18 mM, respectively). These data provide a molecular basis for investigating novel functional roles for AKR1B members and PGF(2alpha) as mediators of physiological and pathological processes in mammalian organisms.


Asunto(s)
Aldehído Reductasa/metabolismo , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Aldehído Reductasa/genética , Animales , Catálisis , Humanos , Hidroxiprostaglandina Deshidrogenasas/genética , Cinética , Ratones , Prostaglandina H2/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
20.
Development ; 134(16): 2947-58, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17626060

RESUMEN

The TAF4 subunit of transcription factor TFIID was inactivated in the basal keratinocytes of foetal and adult mouse epidermis. Loss of TAF4 in the foetal epidermis results in reduced expression of the genes required for skin barrier function, leading to early neonatal death. By contrast, TAF4 inactivation in adult epidermis leads to extensive fur loss and an aberrant hair cycle characterised by a defective anagen phase. Although the mutant epidermis contains few normal anagen-phase hair follicles, many genes expressed at this stage are strongly upregulated indicating desynchronized and inappropriate gene expression. The TAF4 mutant adult epidermis also displays interfollicular hyperplasia associated with a potent upregulation of several members of the EGF family of mitogens. Moreover, loss of TAF4 leads to malignant transformation of chemically induced papillomas and the appearance of invasive melanocytic tumours. Together, our results show that TAF4 is an important regulator of keratinocyte proliferation and has cell-autonomous and non-cell-autonomous tumour suppressor activity.


Asunto(s)
Proliferación Celular , Epidermis/metabolismo , Queratinocitos/citología , Factores Asociados con la Proteína de Unión a TATA/metabolismo , Factores Asociados con la Proteína de Unión a TATA/fisiología , Factor de Transcripción TFIID/metabolismo , Factor de Transcripción TFIID/fisiología , Proteínas Supresoras de Tumor/metabolismo , Animales , Diferenciación Celular/genética , Epidermis/embriología , Epidermis/patología , Femenino , Predisposición Genética a la Enfermedad , Cabello/citología , Cabello/embriología , Hiperplasia/inducido químicamente , Masculino , Ratones , Ratones Noqueados , Nevo Pigmentado/inducido químicamente , Nevo Pigmentado/genética , Subunidades de Proteína/fisiología , Neoplasias Cutáneas/inducido químicamente , Neoplasias Cutáneas/patología , Factores Asociados con la Proteína de Unión a TATA/genética , Factor de Transcripción TFIID/genética , Tretinoina/efectos adversos , Proteínas Supresoras de Tumor/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...