Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Int J Cancer ; 142(9): 1842-1852, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29226331

RESUMEN

The pharmacological therapy of GH-secreting pituitary tumors is based on somatostatin (SS) analogs that reduce GH secretion and cell proliferation by binding mainly SS receptors type 2 (SST2). Antimigratory effects of SS have been demonstrated in different cell models, but no data on pituitary tumors are available. Aims of our study were to evaluate SST2 effects on migration and invasion of human and rat tumoral somatotrophs, and to elucidate the molecular mechanism involved focusing on the role of cofilin and filamin A (FLNA). Our data revealed that SST2 agonist BIM23120 significantly reduced GH3 cells migration (-22% ± 3.6%, p < 0.001) and invasion on collagen IV (-31.3% ± 12.2%, p < 0.01), both these effects being reproduced by octreotide and pasireotide. Similar results were obtained in primary cultured cells from human GH-secreting tumors. These inhibitory actions were accompanied by a marked increase in RhoA/ROCK-dependent cofilin phosphorylation (about 2.7-fold in GH3 and 2.1-fold in human primary cells). Accordingly, the anti-invasive effect of the SS analog was mimicked by the overexpression in GH3 cells of the S3D phosphomimetic cofilin mutant, and abolished by both phosphodeficient S3A cofilin and a specific ROCK inhibitor that prevented cofilin phosphorylation. Moreover, FLNA silencing and FLNA dominant-negative mutants FLNA19-20 and FLNA21-24 transfection demonstrated that FLNA plays a scaffold function for SST2-mediated cofilin phosphorylation. Accordingly, cofilin recruitment to agonist-activated SST2 was completely lost in FLNA silenced cells. In conclusion, we demonstrated that SST2 inhibits rat and human tumoral somatotrophs migration and invasion through a molecular mechanism that involves FLNA-dependent cofilin recruitment and phosphorylation.


Asunto(s)
Citoesqueleto/metabolismo , Neoplasias Hipofisarias/tratamiento farmacológico , Neoplasias Hipofisarias/metabolismo , Receptores de Somatostatina/agonistas , Receptores de Somatostatina/metabolismo , Somatostatina/análogos & derivados , Factores Despolimerizantes de la Actina/metabolismo , Animales , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Citoesqueleto/efectos de los fármacos , Filaminas/metabolismo , Humanos , Invasividad Neoplásica , Fosforilación , Neoplasias Hipofisarias/patología , Ratas , Transducción de Señal/efectos de los fármacos , Somatostatina/farmacología , Proteína de Unión al GTP rhoA/metabolismo
2.
Int J Cancer ; 140(8): 1870-1880, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28120505

RESUMEN

The role of progenitor/stem cells in pituitary tumorigenesis, resistance to pharmacological treatments and tumor recurrence is still unclear. This study investigated the presence of progenitor/stem cells in non-functioning pituitary tumors (NFPTs) and tested the efficacy of dopamine receptor type 2 (DRD2) and somatostatin receptor type 2 (SSTR2) agonists to inhibit in vitro proliferation. They found that 70% of 46 NFPTs formed spheres co-expressing stem cell markers, transcription factors (DAX1, SF1, ERG1) and gonadotropins. Analysis of tumor behavior showed that spheres formation was associated with tumor invasiveness (OR = 3,96; IC: 1.05-14.88, p = 0.036). The in vitro reduction of cell proliferation by DRD2 and SSTR2 agonists (31 ± 17% and 35 ± 13% inhibition, respectively, p < 0.01 vs. basal) occurring in about a half of NFPTs cells was conserved in the corresponding spheres. Accordingly, these drugs increased cyclin-dependent kinase inhibitor p27 and decreased cyclin D3 expression in spheres. In conclusion, they provided further evidence for the existence of cells with a progenitor/stem cells-like phenotype in the majority of NFPTs, particularly in those with invasive behavior, and demonstrated that the antiproliferative effects of dopaminergic and somatostatinergic drugs were maintained in progenitor/stem-like cells.


Asunto(s)
Carcinogénesis/genética , Recurrencia Local de Neoplasia/tratamiento farmacológico , Neoplasias Hipofisarias/tratamiento farmacológico , Receptores de Dopamina D2/genética , Receptores de Somatostatina/genética , Adulto , Carcinogénesis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Ciclina D3/biosíntesis , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/biosíntesis , Receptor Nuclear Huérfano DAX-1/biosíntesis , Dopaminérgicos/administración & dosificación , Resistencia a Antineoplásicos/genética , Canal de Potasio ERG1/biosíntesis , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Gonadotropinas/biosíntesis , Humanos , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/patología , Células Madre Neoplásicas/efectos de los fármacos , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/patología , Factores de Empalme de ARN/biosíntesis , Receptores de Dopamina D2/agonistas , Receptores de Somatostatina/agonistas , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/patología
3.
Exp Cell Res ; 339(2): 241-51, 2015 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-26589262

RESUMEN

cAMP effects have been initially attributed to protein kinase A (PKA) activation. Subsequently, two exchange proteins directly activated by cAMP (Epac1/2) have been identified as cAMP targets. Aim of this study was to investigate cAMP effects in pancreatic-NET (P-NET) and bronchial carcinoids and in corresponding cell lines (QGP-1 and H727) on cell proliferation and adhesion and to determine PKA and Epac role in mediating these effects. We found that cAMP increased cyclin D1 expression in P-NET and QGP-1 cells, whereas it had opposite effects on bronchial carcinoids and H727 cells and it promoted cell adhesion in QGP-1 and H727 cells. These effects are mimicked by Epac and PKA specific analogs, activating the small GTPase Rap1. In conclusion, we demonstrated that cAMP exerted divergent effects on proliferation and promoted cell adhesion of different neuroendocrine cell types, these effects being mediated by both Epac and PKA and involving the same effector GTPase Rap1.


Asunto(s)
Proliferación Celular , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Tumores Neuroendocrinos/metabolismo , Adhesión Celular , Humanos , Tumores Neuroendocrinos/patología , Células Tumorales Cultivadas
4.
Horm Metab Res ; 46(12): 845-53, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25068602

RESUMEN

Molecular mechanisms underlying resistance of pituitary tumors to somatostatin (SS) and dopamine (DA) analogues treatment are not completely understood. Resistance has been associated with defective expression of functional somatostatin and dopamine receptors SSTR2, SSTR5, and DRD2, respectively. Recently, a role of cytoskeleton protein filamin A (FLNA) in DRD2 and SSTR receptors expression and signaling in PRL- and GH-secreting tumors, respectively, has been demonstrated, first revealing a link between FLNA expression and responsiveness of pituitary tumors to pharmacological therapy. No molecular events underlying the reduction of FLNA levels in resistant tumors have been so far identified. FLNA can be phosphorylated by PKA on Ser2152, with increased FLNA resistance to cleavage by calpain and conformational changes affecting FLNA regions involved in SSTR2 and DRD2 binding and signal transduction. In this respect, the effect of cAMP/PKA pathway in the regulation of FLNA stability and/or function by modulating its phosphorylation status could assume particular importance in pituitary, where cAMP cascade plays a crucial role in pituitary cell functions and tumorigenesis. This review will discuss the role of FLNA in the regulation of the main GPCRs target of pharmacological treatment of pituitary tumors, that is, SSTR2 and DRD2, focusing on the effects of cAMP/PKA-mediated FLNA phosphorylation on FLNA biological functions.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Filaminas/metabolismo , Hipófisis/metabolismo , Receptores Dopaminérgicos/metabolismo , Somatostatina/metabolismo , Animales , Humanos , Fosforilación
6.
Cancer Lett ; 497: 77-88, 2021 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-33075426

RESUMEN

Adrenocortical carcinomas (ACCs) overexpress insulin-like growth factor 2 (IGF2), that drives a proliferative autocrine loop by binding to IGF1R and IR, but IGF1R/IR-targeted therapies failed in ACC patients. The cytoskeleton actin-binding protein filamin A (FLNA) impairs IR signalling in melanoma cells. Aims of this study were to test FLNA involvement in regulating IGF1R and IR responsiveness to both IGF2 and inhibitors in ACC. In ACC cells H295R and SW13 and primary cultures (1ACC, 4 adenomas) we found that IGF1R and IR interacted with FLNA, and FLNA silencing increased IGF1R and reduced IR expression, with a downstream effect of increased cell proliferation and ERK phosphorylation. In addition, FLNA knockdown potentiated antiproliferative effects of IGF1R/IR inhibitor Linsitinib and IGF1R inhibitor NVP-ADW742 in H295R. Finally, Western blot showed lower FLNA expression in ACCs (n = 10) than in ACAs (n = 10) and an inverse correlation of FLNA/IGF1R ratio with ERK phosphorylation in ACCs only. In conclusion, we demonstrated that low FLNA levels enhance both IGF2 proliferative effects and IGF1R/IR inhibitors efficacy in ACC cells, suggesting FLNA as a new factor influencing tumor clinical behavior and the response to the therapy with IGF1R/IR-targeted drugs.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/patología , Carcinoma Corticosuprarrenal/patología , Biomarcadores de Tumor/metabolismo , Filaminas/metabolismo , Factor II del Crecimiento Similar a la Insulina/metabolismo , Receptor IGF Tipo 1/antagonistas & inhibidores , Receptor de Insulina/antagonistas & inhibidores , Citoesqueleto de Actina/metabolismo , Neoplasias de la Corteza Suprarrenal/tratamiento farmacológico , Neoplasias de la Corteza Suprarrenal/metabolismo , Carcinoma Corticosuprarrenal/tratamiento farmacológico , Carcinoma Corticosuprarrenal/metabolismo , Apoptosis , Biomarcadores de Tumor/genética , Proliferación Celular , Filaminas/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Imidazoles/farmacología , Factor II del Crecimiento Similar a la Insulina/genética , Mitógenos/farmacología , Pirazinas/farmacología , Pirimidinas/farmacología , Pirroles/farmacología , Transducción de Señal , Células Tumorales Cultivadas
7.
Mol Cell Endocrinol ; 520: 111092, 2021 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-33248230

RESUMEN

Medullary thyroid carcinoma (MTC) is a rare neuroendocrine neoplasm of the parafollicular thyroid C cells. Although somatostatin receptors are expressed by MTCs, treatment with octreotide has shown poor efficacy, whereas recently pasireotide has demonstrated antiproliferative effects in persistent postoperative MTCs. Aim of this study was to test the effects of octreotide and pasireotide on MTC cells proliferation, cell cycle proteins expression, MAPK activation, apoptosis, calcitonin secretion, migration and invasion in TT cell line as well as in primary MTC cultured cells. Our results showed that both octreotide and pasireotide reduced TT cell proliferation (-35.2 ± 12.1%, p < 0.001, and -25.3 ± 24.8%, p < 0.05, at 10-8 M, respectively), with concomitant inhibition of ERK phosphorylation and cyclin D1 expression. This cytostatic effect was accompanied by a proapoptotic action, with an increase of caspase3/7 activity of 1.5-fold. Moreover, both octreotide and pasireotide inhibited cell migration (-50.9 ± 11.3%, p < 0.01, and -40.5 ± 17%, p < 0.05, respectively) and invasion (-61.3 ± 35.1%, p < 0.05, and -49.7 ± 18%, p < 0.01, respectively). No effect was observed on calcitonin secretion. We then tried to extend these observations to primary cultures (n = 5). Octreotide and/or pasireotide were effective in reducing cells proliferation in 3 out of 5 tumors, and to induce cell apoptosis in 1 out of 3 MTCs. Both octreotide and pasireotide were able to reduce cell migration in all MTC tested. SST2, SST3 and SST5 were expressed in all MTC, with a tendency to increased expression of SST2 in RET mutated vs wild type MTCs. In agreement, inhibition of mutated RET in TT cells reduced SST2 expression. In conclusion, we demonstrated that octreotide and pasireotide inhibited cell proliferation and invasiveness in a subset of MTC, supporting their potential use in the control of tumor growth.


Asunto(s)
Carcinoma Neuroendocrino/patología , Octreótido/farmacología , Somatostatina/análogos & derivados , Neoplasias de la Tiroides/patología , Apoptosis/efectos de los fármacos , Calcitonina/metabolismo , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Mutación/genética , Invasividad Neoplásica , Proteínas Proto-Oncogénicas c-ret/genética , Somatostatina/metabolismo , Somatostatina/farmacología , Células Tumorales Cultivadas
8.
J Endocrinol Invest ; 32(6): 501-4, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19494712

RESUMEN

Cardiac myxomas are rare tumors that usually occur as sporadic lesions or,more rarely, in the familial form,mostly in the context of Carney complex (CNC). The molecular basis for the development of cardiac myxomas is unclear. However, somatic activating mutations in the GNAS1 gene (the gsp oncogene) are detected in the myocardium ofMcCune-Albright syndrome patients while germ-line mutations in the PRKAR1A gene are associated with CNC and familial myxomas. We investigated the presence of activating missense mutations in the GNAS1 gene as well as of inactivating mutations in PRKAR1A in 29 sporadically occurring cardiac myxomas. No gsp and no PRKAR1A mutations were found by direct sequencing of PCR products amplified from tumoral DNA. This is the first study including a large series of sporadic, isolated cardiac myxomas and showing that these cardiac neoplasms do not share the same mutations found in familial forms.


Asunto(s)
Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico/genética , Subunidades alfa de la Proteína de Unión al GTP Gs/genética , Neoplasias Cardíacas/genética , Mutación Missense , Mixoma/genética , Adulto , Anciano , Western Blotting , Cromograninas , ADN de Neoplasias/química , ADN de Neoplasias/genética , Femenino , Variación Genética , Neoplasias Cardíacas/enzimología , Neoplasias Cardíacas/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Mixoma/enzimología , Mixoma/metabolismo , Reacción en Cadena de la Polimerasa , Análisis de Secuencia de ADN
9.
Mol Cell Endocrinol ; 495: 110519, 2019 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-31352037

RESUMEN

Medullary thyroid carcinoma (MTC) is a rare neuroendocrine tumor that originates from parafollicular thyroid C cells and accounts for 5% of thyroid cancers. In inherited cases of MTC, and in about 40% of sporadic cases, activating mutations of the receptor tyrosine kinase proto-oncogene RET are found. Constitutively active RET triggers signaling pathways involved in cell proliferation, survival and motility, but the mechanisms underlying malignant transformation of C-cells have been only partially elucidated. Cofilin is a key regulator of actin cytoskeleton dynamics. A crucial role of cofilin in tumor development, progression, invasion and metastasis has been demonstrated in different human cancers, but no data are available in MTC. Interestingly, RET activation upregulates cofilin gene expression. The aim of this study was to investigate cofilin contribution in invasiveness and growth of MTC cells, and its relevance in the context of mutant RET signaling. We found that cofilin transfection in human MTC cell line TT significantly increased migration (178 ±â€¯44%, p < 0.001), invasion (165 ±â€¯28%, p < 0.01) and proliferation (146 ±â€¯18%, p < 0.001), accompanied by an increase of ERK1/2 phosphorylation (2.23-fold) and cyclin D1 levels (1.43-fold). Accordingly, all these responses were significantly reduced after genetic silencing of cofilin (-55 ±â€¯10% migration, p < 0.001, -41 ±â€¯8% invasion, p < 0.001, -17 ±â€¯3% proliferation, p < 0.001). These results have been confirmed in primary cells cultures obtained from human MTCs. The inhibition of constitutively active RET in TT cells by both the RET pharmacological inhibitor RPI-1 and the transfection of dominant negative RET mutant (RETΔTK) resulted in a reduction of cofilin expression (-37 ±â€¯8%, p < 0.001 and -31 ±â€¯16%, p < 0.01, respectively). Furthermore, RPI-1 inhibitory effects on TT cell migration (-57 ±â€¯13%, p < 0.01), but not on cell proliferation, were completely abolished in cells transfected with cofilin. In conclusion, these data indicate that an unbalanced cofilin expression, induced by oncogenic RET, contributes to promote MTC invasiveness and growth, suggesting the possibility of targeting cofilin pathway for more effective treatment of MTC.


Asunto(s)
Factores Despolimerizantes de la Actina/metabolismo , Carcinoma Neuroendocrino/metabolismo , Carcinoma Neuroendocrino/patología , Movimiento Celular , Proteínas Proto-Oncogénicas c-ret/metabolismo , Neoplasias de la Tiroides/metabolismo , Neoplasias de la Tiroides/patología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Silenciador del Gen/efectos de los fármacos , Humanos , Mutación/genética , Invasividad Neoplásica , Inhibidores de Proteínas Quinasas/farmacología , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-ret/antagonistas & inhibidores
10.
Endocr Relat Cancer ; 26(2): R95-R108, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30589642

RESUMEN

Although generally benign, pituitary tumors are frequently locally invasive, with reduced success of neurosurgery and unresponsive to pharmacological treatment with somatostatin or dopamine analogues. The molecular basis of the different biological behavior of pituitary tumors are still poorly identified, but a body of work now suggests that the activity of specific cytoskeleton proteins is a key factor regulating both the invasiveness and drug resistance of these tumors. This review recapitulates the experimental evidence supporting a role for the actin-binding protein filamin A (FLNA) in the regulation of somatostatin and dopamine receptors expression and signaling in pituitary tumors, thus in determining the responsiveness to currently used drugs, somatostatin analogues and dopamine receptor type 2 agonists. Regarding the regulation of invasive behavior of pituitary tumoral cells, we bring evidence to the role of the actin-severing protein cofilin, whose activation status may be modulated by dopaminergic and somatostatinergic drugs, through FLNA involvement. Molecular mechanisms involved in the regulation of FLNA expression and function in pituitary tumors will also be discussed.


Asunto(s)
Filaminas/metabolismo , Neoplasias Hipofisarias/metabolismo , Animales , Citoesqueleto/metabolismo , Humanos , Neoplasias Hipofisarias/tratamiento farmacológico , Neoplasias Hipofisarias/patología
11.
Cancer Lett ; 435: 101-109, 2018 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-30098401

RESUMEN

An efficient intracellular response to somatostatin analogs (SSA) in pituitary tumors requires filamin A (FLNA). Since cAMP pathway plays an important role in GH-secreting pituitary tumors pathogenesis and FLNA is phosphorylated by PKA on S2152, aim of this study was to investigate in tumoral somatotrophs the impact of cAMP pathway activation and SSA stimulation on FLNA phosphorylation and the consequences on SST2 function. We found a PKA-mediated increase (2-fold) and SST2 agonist-induced decrease (-50%) of FLNA phosphorylation in GH3, GH4C1 and primary somatotroph tumor cells. This modification regulates FLNA function. Indeed, phosphomimetic S2152D FLNA mutant, but not phosphodeficient S2152A, abolished the known SSA antitumoral effects, namely: 1) inhibition of cell proliferation, reduction of cyclin D3 and increase of p27; 2) increase of cell apoptosis; 3) inhibition of cell migration via RhoA activation and cofilin phosphorylation. Coimmunoprecipitation and immunofluorescence assays showed that S2152A FLNA was recruited to activated SST2, whereas S2152D FLNA constitutively bound SST2 on the plasma membrane, but prevented Gαi proteins recruitment to SST2. In conclusion, we demonstrated that FLNA phosphorylation, promoted by cAMP pathway activation and inhibited by SSA, prevented SST2 signaling in GH-secreting tumoral pituitary cells.


Asunto(s)
AMP Cíclico/metabolismo , Filaminas/metabolismo , Adenoma Hipofisario Secretor de Hormona del Crecimiento/metabolismo , Neoplasias Hipofisarias/metabolismo , Proteínas Quinasas/metabolismo , Receptores de Somatostatina/metabolismo , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Adenoma Hipofisario Secretor de Hormona del Crecimiento/genética , Adenoma Hipofisario Secretor de Hormona del Crecimiento/patología , Hormonas/farmacología , Humanos , Fosforilación/efectos de los fármacos , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/patología , Ratas , Transducción de Señal/efectos de los fármacos , Somatostatina/farmacología , Somatotrofos/efectos de los fármacos , Somatotrofos/metabolismo , Células Tumorales Cultivadas
12.
J Endocrinol Invest ; 30(10): 828-32, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18075284

RESUMEN

ACTH-dependent Cushing's syndrome is due to ACTH overproduction originating from a pituitary corticotroph adenoma (Cushing's disease) or from ectopic tumors (ectopic ACTH syndrome). Due to difficulties in the differential diagnosis between these two forms of hypercortisolism it would be important to have molecular tools able to discriminate the two conditions. It is known that proopiomelanocortin (POMC) gene transcription can originate messengers of different length. ACTHomas show the normal 1072 nucleotides (nt) transcript, whereas ectopic tumors seem to be associated with a longer mRNA form (1450 nt). In order to analyse the presence of different POMC transcripts, we extracted total RNA from peripheral lymphocytes of 10 patients with Cushing's disease, 10 with ectopic Cushing syndrome, and 20 controls as well as from pituitary tissues (2 ACTH-omas and a normal pituitary polyA+ sample). Northern blot analysis correctly revealed a 1072 nt mRNA molecule in pituitary ACTH-oma and in the normal pituitary polyA+ RNA samples, whereas neither this molecule nor other alternative transcripts were detected in blood samples from patients and controls. These data were confirmed by the more sensitive RT-PCR technique. This study further underlines the need for alternative approaches in the diagnosis of ACTH-dependent Cushing's syndrome.


Asunto(s)
Síndrome de ACTH Ectópico/diagnóstico , Adenoma Hipofisario Secretor de ACTH/diagnóstico , Adenoma/diagnóstico , Biomarcadores de Tumor/genética , Síndrome de Cushing/diagnóstico , Proopiomelanocortina/genética , Síndrome de ACTH Ectópico/complicaciones , Síndrome de ACTH Ectópico/fisiopatología , Adenoma Hipofisario Secretor de ACTH/complicaciones , Adenoma Hipofisario Secretor de ACTH/fisiopatología , Adenoma/complicaciones , Adenoma/fisiopatología , Biomarcadores de Tumor/sangre , Northern Blotting , Síndrome de Cushing/etiología , Síndrome de Cushing/fisiopatología , Diagnóstico Diferencial , Humanos , ARN Mensajero/sangre , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
Cancer Lett ; 406: 54-63, 2017 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-28826686

RESUMEN

cAMP pathway plays a major role in the pathogenesis of cortisol-producing adrenocortical adenomas (CPA). cAMP-induced steroidogenesis is preceded by actin cytoskeleton reorganization, a process regulated by cofilin activity. In this study we investigated cofilin role in mediating cAMP effects on cell morphology and steroidogenesis in adrenocortical tumor cells. We demonstrated that forskolin induced cell rounding and strongly reduced phosphorylated (P)-cofilin/total cofilin ratio in Y1 (-52 ± 16%, p < 0.001) and human CPA cells (-53 ± 18%, p < 0.05). Cofilin silencing significantly reduced both forskolin-induced morphological changes and progesterone production (1.3-fold vs 1.8-fold in controls, p < 0.05), whereas transfection of wild-type or S3A (active), but not S3D (inactive) cofilin, potentiated forskolin effects on cell rounding and increased 3-fold progesterone synthesis with respect to control (p < 0.05). Furthermore, cofilin dephosphorylation by a ROCK inhibitor potentiated forskolin-induced cell rounding and steroidogenesis (2-fold increase vs forskolin alone). Finally, we found a reduced P-cofilin/total cofilin ratio and increased cofilin expression in CPA vs endocrine inactive adenomas by western blot and immunohistochemistry. Overall, these results identified cofilin as a mediator of cAMP effects on both morphological changes and steroidogenesis in mouse and human adrenocortical tumor cells.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Factores Despolimerizantes de la Actina/metabolismo , Neoplasias de la Corteza Suprarrenal/metabolismo , Adenoma Corticosuprarrenal/metabolismo , AMP Cíclico/farmacología , Esteroides/biosíntesis , Factores Despolimerizantes de la Actina/antagonistas & inhibidores , Factores Despolimerizantes de la Actina/genética , Neoplasias de la Corteza Suprarrenal/tratamiento farmacológico , Neoplasias de la Corteza Suprarrenal/patología , Adenoma Corticosuprarrenal/tratamiento farmacológico , Adenoma Corticosuprarrenal/patología , Animales , Colforsina/farmacología , Humanos , Hidrocortisona/metabolismo , Ratones , Fosforilación/efectos de los fármacos , ARN Interferente Pequeño/genética , Células Tumorales Cultivadas , Vasodilatadores/farmacología
14.
Endocr Relat Cancer ; 13(3): 955-62, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16954443

RESUMEN

Somatostatin analogs currently used in the treatment of acromegaly and other neuroendocrine tumors inhibit hormone secretion and cell proliferation by binding to somatostatin receptor type (SST) 2 and 5. The antiproliferative pathways coupled to these receptors have been only partially characterized. The aim of this study was to evaluate the effect of octreotide and super selective SST2 (BIM23120) and SST5 (BIM23206) analogs on apoptotic activity and apoptotic gene expression in human somatotroph tumor cells. Eight somatotroph tumors expressing similar levels of SST2 and SST5 evaluated by real-time PCR and western blot analyses were included in the study. In cultured cells obtained from these tumors, octreotide induced a dose-dependent increase of caspase-3 activity (160+/-20% vs basal at 10 nM) and cleaved cytokeratin 18 levels (172+/-25% vs basal) at concentrations higher than 0.1 nM. This effect was due to SST2 activation since BIM23120 elicited comparable responses, while BIM23206 was ineffective. BIM23120-stimulated apoptosis was dependent on phosphatases, since it was abrogated by the inhibitor orthovanadate, and independent from the induction of apoptosis-related genes, such as p53, p63, p73, Bcl-2, Bax, BID, BIK, TNFSF8, and FADD. In somatotroph tumors, both BIM23120 and BIM2306 caused growth arrest as indicated by the increase in p27 and decrease in cyclin D1 expression. In conclusion, the present study showed that octreotide-induced apoptosis in human somatotroph tumor cells by activating SST2. This effect, together with the cytostatic action exerted by both SST2 and SST5 analogs, might account for the tumor shrinkage observed in acromegalic patients treated with long-acting somatostatin analogs.


Asunto(s)
Apoptosis/efectos de los fármacos , Octreótido/farmacología , Neoplasias Hipofisarias/patología , Receptores de Somatostatina/fisiología , Acromegalia , Caspasa 3/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Neoplasias Hipofisarias/genética , ARN Mensajero/genética , Receptores de Somatostatina/efectos de los fármacos , Receptores de Somatostatina/genética , Células Tumorales Cultivadas
15.
Mol Cell Endocrinol ; 247(1-2): 135-9, 2006 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-16414177

RESUMEN

Hesx1 is a paired-like homeobox gene first expressed during mouse embryogenesis in the anterior midline visceral endoderm. As gastrulation proceeds, Hesx1 is expressed in the ventral prosencephalon and, subsequently, at E9.0 appears in the ventral diencephalon and in the thickened layer of oral ectoderm that give rise to Rathke's pouch, the primordium of the anterior pituitary gland. Hesx1 continues to be expressed in the developing anterior pituitary until E11.5 when its transcripts disappear in a spatiotemporal sequence corresponding to progressive pituitary cell differentiation, becoming undetectable by E15.5. In the present study, we investigated whether HESX1 is expressed during adult life in human normal pituitaries and in different types of human pituitary adenomas. We analysed, using quantitative RT-PCR method, three normal pituitaries, seven GH-, two TSH-, two PRL-, one ACTH-secreting adenomas, and seven nonfunctioning pituitary tumors. HESX1 mRNA was found to be expressed in normal pituitaries and in all the pituitary tumors that we have analysed. These results suggest that in humans HESX1 is not turned-off during the adult life as it occurs in mice. Thus, HESX1 in humans might play a role in the maintenance of the anterior pituitary cell types and function, as well as in the differentiation of pituitary adenomas, whose pathogenetic mechanisms remain to be further investigated. This is the first study on HESX1 expression in humans during adult life.


Asunto(s)
Adenoma/metabolismo , Proteínas de Homeodominio/biosíntesis , Hipófisis/metabolismo , Neoplasias Hipofisarias/metabolismo , Hormona Adrenocorticotrópica/metabolismo , Adulto , Anciano , Femenino , Hormona de Crecimiento Humana/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Hipófisis/patología , Prolactina/metabolismo , ARN Mensajero/biosíntesis , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tirotropina/metabolismo
16.
Cancer Lett ; 381(2): 279-86, 2016 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-27519461

RESUMEN

Non-functioning pituitary tumors (NFPTs) frequently present local invasiveness. Dopamine receptor 2 (DRD2) agonists are the only medical therapy that induces tumor shrinkage in some patients. Invasion requires cytoskeleton rearrangements that are tightly regulated by cofilin pathway, whose alterations correlate with invasion in different tumors. We investigated the effect of DR2D agonist on NFPT cells migration/invasion and the molecular mechanisms involved. We demonstrated that DRD2 agonist reduced migration (-44 ± 25%, p < 0.01) and invasion (-34 ± 6%, p < 0.001) and increased about 4-fold Ser3-phosphorylated inactive cofilin (P-cofilin) in NFPT cells. These effects were abolished by inhibiting ROCK, a kinase that phosphorylates cofilin. The overexpression of wild-type or phosphodeficient S3A-cofilin increased HP75 cells migration (+49 ± 6% and +57 ± 9% vs empty vector, respectively, p < 0.05), while phosphomimetic mutant had no effect. Interestingly, P-cofilin levels were lower in invasive vs non-invasive tumors by both western blot (mean P-cofilin/total cofilin ratio 0.77 and 1.93, respectively, p < 0.05) and immunohistochemistry (mean percentage of P-cofilin positive cells 17.6 and 45.7, respectively, p < 0.05). In conclusion, we showed that the invasiveness of pituitary tumors is promoted by the activation of cofilin, which can be regulated by DRD2 and might represent a novel biomarker for pituitary tumors' invasive behavior.


Asunto(s)
Factores Despolimerizantes de la Actina/metabolismo , Movimiento Celular , Neoplasias Hipofisarias/enzimología , Receptores de Dopamina D2/metabolismo , Quinasas Asociadas a rho/metabolismo , Factores Despolimerizantes de la Actina/genética , Adulto , Anciano , Antineoplásicos/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Agonistas de Dopamina/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Masculino , Persona de Mediana Edad , Invasividad Neoplásica , Fosforilación , Neoplasias Hipofisarias/tratamiento farmacológico , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/patología , Mutación Puntual , Inhibidores de Proteínas Quinasas/farmacología , Receptores de Dopamina D2/agonistas , Transducción de Señal , Transfección , Células Tumorales Cultivadas , Quinasas Asociadas a rho/antagonistas & inhibidores
17.
J Clin Endocrinol Metab ; 90(12): 6721-4, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16204369

RESUMEN

CONTEXT: Alterations in cAMP signaling have been identified as a cause of endocrine neoplasia. In particular, activating mutations of the G(s)alpha gene and protein kinase A (PKA) overactivity due to low expression of PKA regulatory subunit 1A (R1A) have been implicated in somatotroph proliferation. OBJECTIVE: The objective of this study was to evaluate the effects of cAMP-PKA cascade activation in nonfunctioning pituitary adenomas (NFPA). DESIGN AND METHODS: By immunohistochemistry, R1A, R2A, and R2B expression was evaluated in cells obtained from eight surgically removed NFPA positive for gonadotropins. Cyclin D1 expression and ERK1/2 activity were analyzed under basal conditions and after cAMP-PKA cascade activation. RESULTS: Immunohistochemistry studies demonstrated a low R1/R2 ratio in all NFPA. Additional unbalance of R1/R2 ratio by 8-chloroadenosine cAMP (8-Cl-cAMP) and direct adenylyl cyclase stimulation by forskolin did not increase cyclin D1 expression or ERK1/2 activity in five NFPA (group 1), but even caused 74 +/- 15% and 85 +/- 13% inhibitions of cyclin D1 and ERK1/2 activity, respectively, in the remaining NFPA (group 2). Moreover, in group 2, PKA blockade by the specific inhibitor PKI increased cyclin D1 expression (96 +/- 25% over basal) and ERK1/2 activity (116 +/- 28% over basal). CONCLUSIONS: These data show that in contrast with what was previously observed in transformed somatotrophs, activation of the cAMP-PKA pathway did not generate proliferative signals in tumoral cells of the gonadotroph lineage, and in a subset of tumors even exerted a tonic inhibitory effect, thus confirming a different role for the cAMP-mediated pathway in promoting proliferation in the pituitary.


Asunto(s)
Adenoma/metabolismo , Adenoma/patología , Biomarcadores de Tumor/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Neoplasias Hipofisarias/metabolismo , Neoplasias Hipofisarias/patología , Adenoma/fisiopatología , Secuencia de Bases , Proliferación Celular , Subunidad RIalfa de la Proteína Quinasa Dependiente de AMP Cíclico , Ciclina D1/metabolismo , Activación Enzimática , Humanos , Técnicas In Vitro , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Neoplasias Hipofisarias/fisiopatología , Proteínas/genética , Proteínas/metabolismo , Receptores de Superficie Celular/metabolismo
18.
Allerg Immunol (Paris) ; 21(3): 115-8, 1989 Mar.
Artículo en Francés | MEDLINE | ID: mdl-2713044

RESUMEN

From 01/01/1981 to 31/12/1986 inclusive, the S.A.M.U. of Lyon investigated 143 anaphylactic accidents in an extra-hospital environment. The allergological study has shown that 36.3% of the patients are already the victims of a similar incident (in 9.8% of cases, with the same product). The main causative products are the analgesics and non-steroid anti-inflammatory compounds, hymenoptera venoms and antibiotics. In about 1 in 4 cases the enquiry gave negative results. In 1 case in 2 there was a cardiovascular collapse; however it was not possible to establish the relationship between the severity of the clinical picture and the different classes of products. There were no deaths. In comparison with preceding studies there were no developments in the place occupied by the causative products.


Asunto(s)
Anafilaxia/etiología , Adolescente , Adulto , Anciano , Analgésicos/efectos adversos , Anestesia General/efectos adversos , Antibacterianos/efectos adversos , Antiinflamatorios no Esteroideos/efectos adversos , Venenos de Abeja/efectos adversos , Niño , Preescolar , Medios de Contraste/efectos adversos , Desensibilización Inmunológica/efectos adversos , Hipersensibilidad a las Drogas/etiología , Femenino , Hipersensibilidad a los Alimentos/complicaciones , Humanos , Hipersensibilidad/complicaciones , Lactante , Masculino , Persona de Mediana Edad
19.
Endocrinology ; 155(8): 2932-41, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24828612

RESUMEN

Somatostatin receptor type 2 (SST2) is the main pharmacological target of medical therapy for GH-secreting pituitary tumors, but molecular mechanisms regulating its expression and signaling are largely unknown. The aim of this study was to investigate the role of cytoskeleton protein filamin A (FLNA) in SST2 expression and signaling in somatotroph tumor cells. We found a highly variable expression of FLNA in human GH-secreting tumors, without a correlation with SST2 levels. FLNA silencing in human tumoral cells did not affect SST2 expression and localization but abolished the SST2-induced reduction of cyclin D1 (-37% ± 15% in control cells, P < .05 vs basal) and caspase-3/7 activation (+63% ± 31% in control cells, P < .05 vs basal). Overexpression of a FLNA dominant-negative mutant that specifically prevents SST2-FLNA binding reduced SST2 expression after prolonged agonist exposure (-55% ± 5%, P < .01 vs untreated cells) in GH3 cells. Moreover, SST2-induced apoptotic effect (77% ± 54% increase of caspase activity, P < .05 vs basal) and SST2-mediated ERK1/2 inhibition (48% ± 17% reduction of ERK1/2 phosphorylation, P < .01 vs basal) were abrogated in cells overexpressing another FLNA mutant that prevents FLNA interaction with partner proteins but not with SST2, suggesting a scaffold function of FLNA in somatotrophs. In conclusion, these data demonstrate that FLNA is involved in SST2 stabilization and signaling in tumoral somatotrophs, playing both a structural and functional role.


Asunto(s)
Filaminas/fisiología , Receptores de Somatostatina/fisiología , Transducción de Señal/fisiología , Somatotrofos/metabolismo , Animales , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Humanos , Estabilidad Proteica , Ratas , Receptores de Somatostatina/agonistas
20.
Mol Cell Endocrinol ; 383(1-2): 193-202, 2014 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-24373949

RESUMEN

In the pituitary the activation of cyclic adenosine 3'-5'-monophosphate (cAMP) dependent pathways generates proliferative signals in somatotrophs, whereas in pituitary cells of other lineages its effect remains uncertain. Moreover, the specific role of the two main cAMP effectors, protein kinase A (PKA) and exchange proteins directly activated by cAMP (Epac), has not been defined. Aim of this study was to investigate the effect of cAMP on pituitary adenomatous cells proliferation and to identify PKA and Epac differential involvement. We found that cAMP increased DNA synthesis and cyclin D1 expression in somatotropinomas, whereas it reduced both parameters in prolactinomas and nonfunctioning adenomas, these effects being replicated in corresponding cell lines. Moreover, the divergent cAMP effects were mimicked by Epac and PKA analogs, which activated Rap1 and CREB, respectively. In conclusion, we demonstrated that cAMP exerted opposite effects on different pituitary cell types proliferation, these effects being mediated by both Epac and PKA.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/genética , AMP Cíclico/metabolismo , Regulación Neoplásica de la Expresión Génica , Factores de Intercambio de Guanina Nucleótido/genética , Hipófisis/metabolismo , Subunidades de Proteína/genética , Adenoma/genética , Adenoma/metabolismo , Adenoma/patología , Animales , Proliferación Celular , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Ciclina D1/genética , Ciclina D1/metabolismo , Gonadotrofos/metabolismo , Gonadotrofos/patología , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Lactotrofos/metabolismo , Lactotrofos/patología , Hipófisis/patología , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/metabolismo , Neoplasias Hipofisarias/patología , Prolactinoma/genética , Prolactinoma/metabolismo , Prolactinoma/patología , Subunidades de Proteína/metabolismo , Ratas , Transducción de Señal , Somatotrofos/metabolismo , Somatotrofos/patología , Proteínas de Unión al GTP rap1/genética , Proteínas de Unión al GTP rap1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA