Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Endocrine ; 56(3): 621-632, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27585662

RESUMEN

Growth Hormone may influence neoplastic development of endometrial epithelium towards endometrial adenocarcinoma, which is one of the most occurring tumors in acromegalic patients. Since chemoresistance often develops in advanced endometrial adenocarcinoma, we investigated whether Growth Hormone might influence the development of chemoresistance to drugs routinely employed in endometrial adenocarcinoma treatment, such as Doxorubicin, Cisplatin, and Paclitaxel. Growth Hormone and Growth Hormone receptor expression was assessed by immunofluorescence in two endometrial adenocarcinoma cell lines, AN3 CA and HEC-1-A cells. Growth Hormone effects were assessed investigating cell viability, caspase3/7 activation, ERK1/2, and protein kinase C delta protein expression. AN3 CA and HEC-1-A cells display Growth Hormone and Growth Hormone receptor. Growth Hormone does not influence cell viability in both cells lines, but significantly reduces caspase 3/7 activation in AN3 CA cells, an effect blocked by a Growth Hormone receptor antagonist. Growth Hormone rescues AN3 CA cells from the inhibitory effects of Doxorubicin and Cisplatin on cell viability, while it has no effect on Paclitaxel. Growth Hormone does not influence the pro-apoptotic effects of Doxorubicin, but is capable of rescuing AN3 CA cells from the pro-apoptotic effects of Cisplatin. On the other hand, Growth Hormone did not influence the effects of Doxorubicin and Paclitaxel on HEC-1A cell viability. The protective action of Growth Hormone towards the effects of Doxorubicin may be mediated by ERK1/2 activation, while the pro-apoptotic effects of Cisplatin may be mediated by protein kinase C delta inhibition. All together our results indicate that Growth Hormone may differentially contribute to endometrial adenocarcinoma chemoresistance. This may provide new insights on novel therapies against endometrial adenocarcinoma chemoresistant aggressive tumors.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Cisplatino/farmacología , Doxorrubicina/farmacología , Neoplasias Endometriales/tratamiento farmacológico , Hormona de Crecimiento Humana/análogos & derivados , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Antineoplásicos/uso terapéutico , Antineoplásicos Hormonales/farmacología , Antineoplásicos Hormonales/uso terapéutico , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Línea Celular Tumoral , Cisplatino/uso terapéutico , Doxorrubicina/uso terapéutico , Resistencia a Antineoplásicos , Neoplasias Endometriales/metabolismo , Neoplasias Endometriales/patología , Femenino , Hormona de Crecimiento Humana/metabolismo , Hormona de Crecimiento Humana/farmacología , Humanos , Señales de Exportación Nuclear/efectos de los fármacos , Paclitaxel/farmacología , Paclitaxel/uso terapéutico , Receptores de Somatotropina/metabolismo
2.
Mol Cell Endocrinol ; 419: 252-8, 2016 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-26522132

RESUMEN

Protein Kinase C Delta (PRKCD) has been highlighted among disrupted pathways in corticotroph adenomas. PRKCD is expressed at low level in human corticotroph adenomas and controls cell cycle in vitro. Therefore, PRKCD may play an important role in the development/progression of corticotroph adenomas, warranting further studies to understand the role of PRKCD and related pathways in restraining pituitary cell growth. We evaluated PRKCD role in influencing cell behavior in terms of cell viability, hormone expression and protein expression profile, by silencing PRKCD in AtT-20/D16v-F2 cells. PRKCD silencing increases cell viability, enhances hormone expression and induces morphological changes associated with deregulation of adhesion molecules. PRKCD silencing is associated with an increase in Epithelial Growth Factor Receptor (EGFR) expression, a marker of tumor aggressive behavior, and sensitivity to anti-EGFR molecules. PRKCD might restrain corticotroph adenoma cells from acquiring an aggressive behavior, candidating PRKCD as a possible molecular target for the treatment of corticotroph adenomas.


Asunto(s)
Hormona Adrenocorticotrópica/metabolismo , Neoplasias Hipofisarias/metabolismo , Proteína Quinasa C-delta/metabolismo , Animales , Línea Celular Tumoral , Supervivencia Celular , Receptores ErbB/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Proopiomelanocortina/metabolismo
3.
Artículo en Inglés | MEDLINE | ID: mdl-25999915

RESUMEN

CONTEXT: Medullary thyroid carcinoma (MTC) is a rare tumor originating from thyroid parafollicular C cells. It has been previously demonstrated that insulin-like growth factor I (IGF-I) protects MTC from the effects of antiproliferative drugs. Everolimus, an mTOR inhibitor, has shown potent antiproliferative effects in a human MTC cell line, TT, and in two human MTC primary cultures. OBJECTIVE: To verify whether IGF-I may influence the effects of everolimus in a group of human MTC primary cultures. DESIGN: We collected 18 MTCs that were dispersed in primary cultures, treated without or with 10 nM-1 µM everolimus and/or 50 nM IGF-I. Cell viability was evaluated after 48 h, and calcitonin (CT) secretion was assessed after a 6 h incubation. IGF-I receptor downstream signaling protein expression profile was also investigated. RESULTS: Everolimus significantly reduced cell viability in eight MTC [by ~20%; P < 0.01 vs. control; everolimus-responders (E-R) MTCs], while cell viability did not change in 10 MTCs [everolimus-non-responders (E-NR) MTCs]. In E-R MTCs, IGF-I blocked the antiproliferative effects of everolimus that did not affect CT secretion, but blocked the stimulatory effects of IGF-I on this parameter. IGF-I receptor downstream signaling proteins were expressed at higher levels in E-NR MTC as compared to E-R MTCs. CONCLUSION: IGF-I protects a subset of MTC primary cultures from the antiproliferative effects of everolimus and stimulates CT secretion by an mTOR mediated pathway that, in turn, may represent a therapeutic target in the treatment of aggressive MTCs.

4.
Reprod Toxicol ; 45: 71-6, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24486453

RESUMEN

Mitotane represents the mainstay medical treatment for metastatic, inoperable or recurrent adrenocortical carcinoma. Besides the well-known adverse events, mitotane therapy is associated also with endocrinological effects, including sexual and reproductive dysfunction. The majority of male patients undergoing adjuvant mitotane therapy show a picture of hypogonadism, characterized by low free testosterone and high sex hormone binding globulin levels and unmodified LH concentrations. Since mitotane has been shown to have direct pituitary effects, we investigated whether mitotane may influence both cell viability and function of gonadotroph cells in the settings of two pituitary cell lines. We found that mitotane reduces cell viability, induces apoptosis, modifies cell cycle phase distribution and secretion of gonadotroph cells. The present data strengthen previous evidence showing a direct mitotane effect at pituitary level and represent a possible explanation of the lack of LH increase following decrease in free testosterone in patients undergoing adjuvant mitotane therapy.


Asunto(s)
Antineoplásicos Hormonales/toxicidad , Gonadotrofos/efectos de los fármacos , Mitotano/toxicidad , Animales , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular , Supervivencia Celular/efectos de los fármacos , Hormona Folículo Estimulante/metabolismo , Gonadotrofos/citología , Gonadotrofos/metabolismo , Hormona Luteinizante/metabolismo , Ratones
5.
J Endocrinol ; 218(3): 275-85, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23814013

RESUMEN

Medical therapy for Cushing's disease (CD) is currently based on agents mainly targeting adrenocortical function. Lately, pituitary-directed drugs have been developed, with limited efficacy. Mitotane, a potent adrenolytic drug, has been recently investigated for the treatment of CD, but the direct pituitary effects have not been clarified so far. The aim of our study was to investigate whether mitotane may affect corticotroph function and cell survival in the mouse pituitary cell line AtT20/D16v-F2 and in the primary cultures of human ACTH-secreting pituitary adenomas, as an in vitro model of pituitary corticotrophs. We found that in the AtT20/D16v-F2 cell line and in primary cultures, mitotane reduces cell viability by inducing caspase-mediated apoptosis and reduces ACTH secretion. In the AtT20/D16v-F2 cell line, mitotane reduces Pomc expression and blocks the stimulatory effects of corticotropin-releasing hormone on cell viability, ACTH secretion, and Pomc expression. These effects were apparent at mitotane doses greater than those usually necessary for reducing cortisol secretion in Cushing's syndrome, but still in the therapeutic window for adrenocortical carcinoma treatment. In conclusion, our results demonstrate that mitotane affects cell viability and function of human and mouse ACTH-secreting pituitary adenoma cells. These data indicate that mitotane could have direct pituitary effects on corticotroph cells.


Asunto(s)
Adenoma Hipofisario Secretor de ACTH/metabolismo , Corticotrofos/metabolismo , Mitotano/farmacología , Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT)/metabolismo , Adenoma Hipofisario Secretor de ACTH/tratamiento farmacológico , Adenoma Hipofisario Secretor de ACTH/genética , Hormona Adrenocorticotrópica/metabolismo , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Corticotrofos/citología , Corticotrofos/efectos de los fármacos , Humanos , Ratones , Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT)/tratamiento farmacológico , Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT)/genética , Hipófisis/citología , Hipófisis/efectos de los fármacos , Hipófisis/metabolismo , Proopiomelanocortina/genética , Proopiomelanocortina/metabolismo
6.
Bone ; 56(2): 426-31, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23871850

RESUMEN

Bisphosphonates (BPs) are the first-line therapy for osteoporosis. In recent years, atypical femoral fractures (AFF) have been described in patients on BPs therapy. However, the relationship between BPs and AFF remains to be clarified. We evaluated clinical and hormonal characteristics of AFF patients, in order to determine AFF risk factors. We studied 11 females with AFF and 58 females with typical femoral fractures (TFF), admitted to our Department for surgical repair between January 2008 and December 2011. All AFF patients received BPs therapy for 6 to 13 yrs, whereas 36.2% (p<0.0001) of TFF patients received BPs for shorter period (TFF, 6.1±1.8 yr vs. AFF, 8.6±1.9 yr, p<0.0001). A higher prevalence of hypocalcemia was observed in AFF patients compared with TFF (p<0.02), with significantly (p<0.05) lower corrected calcium levels in AFF patients. By contrast a reduced prevalence of elevated PTH levels (p<0.05) was found in AFF patients. No significant difference in prevalence of vitamin D defect was observed between the two groups. Younger age (p<0.004), higher BMI (>30 kg/m2, p<0.03) and early menopausal age (p<0.05) were observed in AFF patients. At time of fracture, prevalence of osteopenia/osteoporosis and levels of bone turnover markers were significantly (p<0.01) lower in AFF compared with TFF patients. By multivariate analysis hypocalcemia, obesity, and younger age (<70 yr) were confirmed to be independent predictors of AFF; elevated PTH level was the predominant independent protective factor (p<0.004). In conclusion, our data indicate that clinical characteristics and metabolic factors may favor the development of AFF in BP treated patients. We identified hypocalcemia due to latent hypoparathyroidism as primary risk factor for AFF; age, obesity, early menopause, and BMD may also influence the development of AFF. An adequate clinical and metabolic assessment is suggested to prevent the development of AFF in BP treated patients.


Asunto(s)
Conservadores de la Densidad Ósea/efectos adversos , Difosfonatos/efectos adversos , Fracturas del Fémur/inducido químicamente , Anciano , Anciano de 80 o más Años , Conservadores de la Densidad Ósea/uso terapéutico , Difosfonatos/uso terapéutico , Femenino , Humanos , Masculino , Persona de Mediana Edad , Osteoporosis/tratamiento farmacológico
7.
Endocrinology ; 154(5): 1690-700, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23525216

RESUMEN

The functional aftermath of microRNA (miRNA) dysregulation in ACTH-secreting pituitary adenomas has not been demonstrated. miRNAs represent diagnostic and prognostic biomarkers as well as putative therapeutic targets; their investigation may shed light on the mechanisms that underpin pituitary adenoma development and progression. Drugs interacting with such pathways may help in achieving disease control also in the settings of ACTH-secreting pituitary adenomas. We investigated the expression of 10 miRNAs among those that were found as most dysregulated in human pituitary adenoma tissues in the settings of a murine ACTH-secreting pituitary adenoma cell line, AtT20/D16v-F2. The selected miRNAs to be submitted to further investigation in AtT20/D16v-F2 cells represent an expression panel including 5 up-regulated and 5 down-regulated miRNAs. Among these, we selected the most dysregulated mouse miRNA and searched for miRNA targets and their biological function. We found that AtT20/D16v-F2 cells have a specific miRNA expression profile and that miR-26a is the most dysregulated miRNA. The latter is overexpressed in human pituitary adenomas and can control viable cell number in the in vitro model without involving caspase 3/7-mediated apoptosis. We demonstrated that protein kinase Cδ (PRKCD) is a direct target of miR-26a and that miR26a inhibition delays the cell cycle in G1 phase. This effect involves down-regulation of cyclin E and cyclin A expression via PRKCD modulation. miR-26a and related pathways, such as PRKCD, play an important role in cell cycle control of ACTH pituitary cells, opening new therapeutic possibilities for the treatment of persistent/recurrent Cushing's disease.


Asunto(s)
Adenoma Hipofisario Secretor de ACTH/genética , Adenoma/genética , Ciclo Celular/genética , MicroARNs/fisiología , Proteína Quinasa C-delta/metabolismo , Adenoma Hipofisario Secretor de ACTH/metabolismo , Adenoma Hipofisario Secretor de ACTH/patología , Adenoma/metabolismo , Adenoma/patología , Animales , Línea Celular Tumoral , Supervivencia Celular/genética , Activación Enzimática , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Proteína Quinasa C-delta/genética
8.
PLoS Pathog ; 8(10): e1002951, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23055929

RESUMEN

Hashimoto's thyroiditis (HT) is the most common of all thyroid diseases and is characterized by abundant lymphocyte infiltrate and thyroid impairment, caused by various cell- and antibody-mediated immune processes. Viral infections have been suggested as possible environmental triggers, but conclusive data are not available. We analyzed the presence and transcriptional state of human herpesvirus 6 (HHV-6) in thyroid fine needle aspirates (FNA) and peripheral blood mononuclear cells (PBMCs) from 34 HT patients and 28 controls, showing that HHV-6 DNA prevalence (82% vs. 10%, p≤0.001) and viral load were significantly increased in FNA from HT patients, and thyrocytes from HT FNA displayed a 100-fold higher HHV-6 DNA load compared to infiltrating lymphocytes. In addition, while HHV-6 was strictly latent in positive samples from controls, a low grade acute infection was detected in HT samples. HHV-6 variant characterization was carried out in 10 HT FNA samples, determining that all specimens harbored HHV-6 Variant A.The tropism of HHV-6 for thyroid cells was verified by infection of Nthy-ori3-1, a thyroid follicular epithelial cell line, showing that thyrocytes are permissive to HHV-6 replication, which induces de novo expression of HLA class II antigens. Furthermore, HHV-6-infected Nthy-ori3-1 cells become targets for NK-mediated killing, NK cells from HT patients show a significantly more efficient killing of HHV-6 infected thyroid cells than healthy controls, and HT patients have increased T-cell responses to HHV-6 U94 protein, associated to viral latency. These observations suggest a potential role for HHV-6 (possibly variant A) in the development or triggering of HT.


Asunto(s)
Enfermedad de Hashimoto/etiología , Enfermedad de Hashimoto/virología , Herpesvirus Humano 6/patogenicidad , Infecciones por Roseolovirus/virología , Glándula Tiroides/patología , Biopsia con Aguja Fina , Línea Celular , ADN Viral , Células Epiteliales/virología , Enfermedad de Hashimoto/inmunología , Herpesvirus Humano 6/genética , Herpesvirus Humano 6/inmunología , Herpesvirus Humano 6/aislamiento & purificación , Antígenos de Histocompatibilidad Clase II/biosíntesis , Humanos , Células Asesinas Naturales/inmunología , Leucocitos Mononucleares/virología , Glándula Tiroides/virología , Carga Viral
9.
J Clin Endocrinol Metab ; 97(7): 2354-61, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22535974

RESUMEN

CONTEXT: Ultrasound (US)-guided fine-needle aspiration biopsy (FNAB) is the most reliable nonsurgical test for distinguishing benign from malignant thyroid nodules. However, there is no consensus on which nodules should undergo FNAB. AIMS: The aims of this study were to evaluate the utility of US-guided FNAB in the diagnostic assessment of nodules with or without clinical/US features suggestive for malignancy and to investigate the additional contribution of BRAF V600E mutation analysis in the detection of differentiated thyroid cancer. DESIGN AND METHODS: Thyroid cytoaspirates from 2421 nodules at least 4 mm in diameter were performed in 1856 patients who underwent cytological evaluation and biomolecular analysis. RESULTS: Cytology showed high positive predictive value and specificity for the diagnosis of malignant lesions. BRAF V600E mutation was found in 115 samples, 80 of which were also cytologically diagnosed as papillary thyroid cancer. BRAF mutation analysis significantly enhanced the diagnostic value of cytology, increasing FNAB diagnostic sensitivity for malignant nodules by approximately 28%. Micro PTC (63% of diagnosed papillary thyroid carcinoma) showed a high prevalence of multifocality, extrathyroidal extension, and lymph node metastases, underlining the malignant potential of thyroid microcarcinomas. Each investigated US/clinical characteristic of suspected malignancy correlated with the presence of a thyroid cancer in thyroid nodules with diameter of at least 4 mm. CONCLUSIONS: These data indicate that nodules of at least 4 mm may underlie a thyroid cancer independently of US/clinical characteristics of suspected malignancy, suggesting the need to perform FNAB. The diagnostic sensitivity for thyroid cancer is significantly increased by BRAF V600E mutation analysis, indicating that the screening for BRAF mutation in FNAB samples has a relevant diagnostic potential.


Asunto(s)
Análisis Mutacional de ADN , Detección Precoz del Cáncer/métodos , Proteínas Proto-Oncogénicas B-raf/genética , Neoplasias de la Tiroides/diagnóstico , Nódulo Tiroideo/patología , Ultrasonografía Intervencional , Adulto , Sustitución de Aminoácidos/genética , Biopsia con Aguja Fina/métodos , Carcinoma , Carcinoma Papilar , Técnicas Citológicas , Femenino , Ácido Glutámico/genética , Humanos , Masculino , Persona de Mediana Edad , Técnicas de Diagnóstico Molecular , Mutación Missense , Valor Predictivo de las Pruebas , Estudios Prospectivos , Proteínas Proto-Oncogénicas B-raf/análisis , Estudios Retrospectivos , Sensibilidad y Especificidad , Cáncer Papilar Tiroideo , Neoplasias de la Tiroides/diagnóstico por imagen , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/patología , Nódulo Tiroideo/diagnóstico por imagen , Nódulo Tiroideo/genética , Ultrasonografía Intervencional/métodos , Ultrasonografía Intervencional/estadística & datos numéricos , Valina/genética
10.
Endocrinology ; 153(5): 2088-98, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22374978

RESUMEN

We investigate the role of protein kinase C (PKC) in the control of medullary thyroid carcinoma (MTC) cell proliferation by a PKC inhibitor, Enzastaurin, in human MTC primary cultures and in the TT cell line. We found that PKC inhibition reduces cell proliferation by inducing caspase-mediated apoptosis and blocks the stimulatory effect of IGF-I on calcitonin secretion. Enzastaurin reduces PKCßII (Thr500) phosphorylation, indicating a direct involvement of this isoform as well as the phosphorylated levels of Akt (Ser 473) and glycogen synthase kinase (Ser9), PKC pathway downstream targets and pharmacodynamic markers for PKC inhibition. PKCßII and PKCδ enzyme isoforms expression and localization were investigated. These data indicate that in vitro PKC is involved in the control of human MTC proliferation and survival by modulating apoptosis, with a mechanism that implicates PKCßII inhibition and translocation in different subcellular compartments. Targeting PKC may represent a useful therapeutic approach for controlling MTC proliferation.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Indoles/farmacología , Proteína Quinasa C/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Glándula Tiroides/efectos de los fármacos , Adulto , Anciano , Apoptosis/efectos de los fármacos , Carcinoma Medular/metabolismo , Línea Celular Tumoral , Femenino , Humanos , Masculino , Persona de Mediana Edad , Fosforilación/efectos de los fármacos , Proteína Quinasa C/metabolismo , Glándula Tiroides/metabolismo , Neoplasias de la Tiroides/metabolismo
11.
Endocr Relat Cancer ; 18(4): 439-50, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21606156

RESUMEN

Dysregulation of the protein kinase C (PKC) signaling pathway has been implicated in tumor progression. In this study, we investigate the effects of a PKC inhibitor, Enzastaurin, in human pancreatic neuroendocrine neoplasms (PNN) primary cultures and in the human pancreatic endocrine cancer cell line, BON1. To this aim six human PNN dispersed in primary cultures and BON1 cells were treated without or with 1-10 µM Enzastaurin and/or 100 nM IGF1 in the presence or absence of serum. Cell viability and apoptosis were evaluated after 48-72 h; Chromogranin A (CgA) and/or insulin secretion was assessed after 6 h of incubation. PKC expression was investigated by immunofluorescence and western blot. We found that Enzastaurin significantly reduced human PNN primary culture cell viability, as well as CgA and insulin secretion. Moreover, in the BON1 cell line Enzastaurin inhibited cell proliferation at 5 and 10 µM by inducing caspase-mediated apoptosis, and reduced phosphorylation of glycogen synthetase kinase 3ß (GSK3ß) and of Akt, both downstream targets of PKC pathway and pharmacodynamic markers for Enzastaurin. In addition, Enzastaurin blocked the stimulatory effect of IGF1 on cell proliferation, and reduced CgA expression and secretion in BON1 cells. Two different PKC isoforms are expressed at different levels and have partially different subcellular localization in BON1 cells. In conclusion, Enzastaurin reduces cell proliferation by inducing apoptosis, with a mechanism likely involving GSK3ß signaling, and inhibits secretory activity in PNN in vitro models, suggesting that Enzastaurin might represent a possible medical treatment of human PNN.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Cromogranina A/metabolismo , Indoles/farmacología , Insulina/metabolismo , Tumores Neuroendocrinos/tratamiento farmacológico , Neoplasias Pancreáticas/tratamiento farmacológico , Proteína Quinasa C/antagonistas & inhibidores , Apoptosis/efectos de los fármacos , Western Blotting , Ensayo de Inmunoadsorción Enzimática , Técnica del Anticuerpo Fluorescente , Humanos , Secreción de Insulina , Tumores Neuroendocrinos/enzimología , Tumores Neuroendocrinos/patología , Neoplasias Pancreáticas/enzimología , Neoplasias Pancreáticas/patología , Proteína Quinasa C/metabolismo , Células Tumorales Cultivadas
12.
Eur J Endocrinol ; 164(3): 355-62, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21169415

RESUMEN

CONTEXT: KISS1 was originally identified as a metastasis-suppressor gene able to inhibit tumor progression. KISS1 gene products, the kisspeptins, bind to a G-protein-coupled receptor (KISS1R, formerly GPR54), which is highly expressed in placenta, pituitary, and pancreas, whereas KISS1 mRNA is mainly expressed in placenta, hypothalamus, striatum, and pituitary. OBJECTIVE AND DESIGN: KISS1/KISS1R pituitary expression profile, coupled to their anti-tumoral capacities, led us to hypothesize that this system may be involved in the biology of pituitary tumors. To explore this notion, expression levels of KISS1R and KISS1 were evaluated in normal and adenomatous pituitaries. Additionally, functionality of this system was assessed by treating dispersed pituitary adenoma cells in primary culture with kisspeptin-10 and evaluating intracellular calcium kinetics and apoptotic rate. RESULTS: Both KISS1 and KISS1R were expressed in normal pituitary, whereas this simultaneous expression was frequently lost in pituitary tumors, where diverse patterns of KISS1/KISS1R expression were observed that differed among distinct types of pituitary adenomas. Measurement of calcium kinetics revealed that kisspeptin-10 elicits a remarkable increase in [Ca(2+)](i) in individual cells from four out of the five GH-producing adenomas studied, whereas cells derived from non-functioning pituitary adenomas (NFPA, n=45) did not respond. In contrast, kisspeptin-10 treatment increased the apoptotic rate in cells derived from both GH-producing and NFPA. CONCLUSIONS: These results provide primary evidence that KISS1 and KISS1R expression can be differentially lost in pituitary tumor subtypes, where this system can exert functional, proapoptotic actions, and thereby offer novel insights to investigate the biology and therapeutic options to treat these tumors.


Asunto(s)
Apoptosis , Regulación Neoplásica de la Expresión Génica , Neoplasias Hipofisarias/metabolismo , Neoplasias Hipofisarias/fisiopatología , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Apoptosis/genética , Apoptosis/fisiología , Calcio/metabolismo , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Humanos , Técnicas In Vitro , Kisspeptinas , Hipófisis/metabolismo , Hipófisis/patología , Neoplasias Hipofisarias/patología , Receptores Acoplados a Proteínas G/genética , Receptores de Kisspeptina-1 , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Temperatura , Proteínas Supresoras de Tumor/genética
13.
Anal Cell Pathol (Amst) ; 33(5): 207-16, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20978326

RESUMEN

BACKGROUND: pituitary tumour transforming gene 1 (PTTG1) is over-expressed in a variety of endocrine-related tumours. We aimed at evaluating PTTG1 expression and function in human neoplastic parafollicular C-cells, represented by medullary thyroid carcinoma (MTC) and C-cell hyperplasia (CCH) samples and by the TT cell line. METHODS: TT cells and tissues derived from human CCH (8 samples) and MTC (12 samples) were analyzed by northern blot, furthermore TT cells were subjected to PTTG gene silencing and cells were analyzed for DNA synthesis. RESULTS: PTTG1 expression was significantly higher (p<0.01) in CCH (3-fold), in papillary thyroid cancer and in MTC (5-fold) than in normal thyroid, and in MTC lymph-node metastases as compared to primary lesions (approximately 2-fold; p<0.05). PTTG1 mRNA and protein correlated with tumour diameter and TNM status (p<0.05). In TT cells, PTTG1 silencing did not completely block DNA synthesis, but significantly reduced [3H]Thymidine incorporation (~50%; p<0.01) for up to 3 days. CONCLUSIONS: PTTG1 levels correlate with tumour aggressiveness. PTTG1 silencing causes reduced MTC cell proliferation, supporting the hypothesis that PTTG1 might have an important role in C-cell neoplastic proliferation.


Asunto(s)
Biomarcadores de Tumor/análisis , Proteínas de Neoplasias/biosíntesis , Adolescente , Adulto , Anciano , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Northern Blotting , Western Blotting , Carcinoma Neuroendocrino , Proliferación Celular , Niño , Femenino , Silenciador del Gen , Humanos , Hiperplasia , Metástasis Linfática , Masculino , Microscopía Fluorescente , Persona de Mediana Edad , Proteínas de Neoplasias/genética , Estadificación de Neoplasias , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Securina , Neoplasias de la Tiroides/genética , Neoplasias de la Tiroides/metabolismo , Neoplasias de la Tiroides/patología
14.
Hum Mutat ; 31(11): E1825-35, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20824794

RESUMEN

Multiple endocrine neoplasia (MEN) syndromes are characterized by tumors involving two or more endocrine glands. Two MEN syndromes have long been known: MEN1 and MEN2,caused by germline mutations in MEN1 or RET, respectively. Recently, mutations in CDKN1B,encoding the cyclin-dependent kinase (Cdk) inhibitor p27, were identified in patients having a MEN1-like phenotype but no MEN1 gene mutations. Currently, the molecular mechanisms mediating the role of p27 in tumor predisposition are ill defined. We here report a novel germline missense variant in CDKN1B (c.678C>T, p.P69L) found in a patient with multiple endocrine tumors. We previously reported a nonsense p27 mutation (c.692G>A, p.W76X) in two patients with MEN1-like phenotype. Functional assays were used to characterize p27P69L and p27W76X in vitro. We show that p27P69L is expressed at reduced level and is impaired in both binding toCdk2 and inhibiting cell growth. p27W76X, which is mislocalized to the cytoplasm, can no longer efficiently bind Cyclins-Cdks, nor inhibit cell growth or induce apoptosis. In the patient's tumor tissues, p27P69L associates with reduced/absent p27 expression and in one tumor with loss-of heterozygosity.Our results extend previous findings of CDKN1B mutations in patients with MEN1-related states and support the hypothesis of a tumor suppressor role for p27 in neuroendocrine cells.


Asunto(s)
Mutación de Línea Germinal , Péptidos y Proteínas de Señalización Intracelular/genética , Neoplasia Endocrina Múltiple/genética , Anciano , Línea Celular , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Análisis Mutacional de ADN , ADN de Neoplasias/genética , Femenino , Técnicas de Silenciamiento del Gen , Células HeLa , Humanos , Técnicas In Vitro , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Neoplasia Endocrina Múltiple/clasificación , Neoplasia Endocrina Múltiple Tipo 1/genética , Mutagénesis Sitio-Dirigida , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Fenotipo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
15.
J Clin Endocrinol Metab ; 95(10): 4660-8, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20660027

RESUMEN

BACKGROUND: Stroke is a leading cause of death in industrialized countries, representing the main cause of long-term disability. Recent studies indicate that hypopituitarism may be observed after an acute stroke. OBJECTIVE: The aim was to prospectively investigate incidence and pattern of pituitary dysfunction in patients suffering ischemic stroke and to assess the predictive value of different clinical and radiological parameters for hypopituitarism. PATIENTS AND METHODS: We assessed endocrine, clinical, radiological, and functional parameters in 56 patients (34 males; mean age, 64.8 ± 1.3 yr; mean body mass index, 25.8 ± 0.45 kg/m(2)) at 1-3 months (visit 1) and 12-15 months (visit 2) after an ischemic stroke. RESULTS: At visit 1, hypopituitarism was detected in 20 (35.7%) of 56 stroke patients, with multiple deficits in three and isolated deficits in 17. At visit 2, hypopituitarism was detected in 18 (37.5%) of 48 stroke patients, with multiple deficits in two. Four patients with previously diagnosed isolated GH or LH/FSH deficit exhibited normal pituitary function, whereas GH deficiency was newly diagnosed in three cases. Hypopituitarism was associated with worse outcome. We identified both clinical (preexisting diabetes mellitus, medical complications during hospitalization) and radiological (Alberta Stroke Programme Early CT Score ≤ 7) parameters as major risk factors for developing hypopituitarism after ischemic stroke. CONCLUSIONS: Hypopituitarism may associate with ischemic stroke in one third of cases and persist in a long-term period, aggravating the functional outcome. We identified specific risk factors for hypopituitarism after stroke, which may help to select patients needing an accurate endocrine evaluation to improve stroke outcome.


Asunto(s)
Isquemia Encefálica/complicaciones , Enfermedades de la Hipófisis/diagnóstico , Accidente Cerebrovascular/complicaciones , Sobrevivientes/estadística & datos numéricos , Anciano , Isquemia Encefálica/diagnóstico por imagen , Isquemia Encefálica/epidemiología , Isquemia Encefálica/rehabilitación , Femenino , Humanos , Hipopituitarismo/epidemiología , Hipopituitarismo/etiología , Incidencia , Masculino , Persona de Mediana Edad , Enfermedades de la Hipófisis/diagnóstico por imagen , Enfermedades de la Hipófisis/epidemiología , Enfermedades de la Hipófisis/etiología , Pruebas de Función Hipofisaria , Valor Predictivo de las Pruebas , Pronóstico , Radiografía , Factores de Riesgo , Accidente Cerebrovascular/diagnóstico por imagen , Accidente Cerebrovascular/epidemiología , Rehabilitación de Accidente Cerebrovascular
16.
Endocr Relat Cancer ; 17(3): 719-29, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20554786

RESUMEN

Bronchial carcinoids (BCs) are rare tumors originating from endocrine cells dispersed in the respiratory epithelium. It has been previously demonstrated that everolimus, or RAD001, an mTOR inhibitor, has potent antiproliferative effects in human endocrine tumors. Our aim was to evaluate the possible antiproliferative effects of everolimus in human BCs in primary culture. We collected 24 BCs that were dispersed in primary cultures, treated without or with 1 nM-1 muM everolimus, 10 nM SOM230 (pasireotide, a somatostatin receptor multiligand), and/or 50 nM IGF1. Cell viability was evaluated after 48 h, and chromogranin A (CgA) as well as vascular endothelial growth factor (VEGF) secretion was assessed after 8 h incubation. Somatostatin receptors, mTOR, and AKT expression were investigated by quantitative PCR. We found that in 15 cultures (67.5%), everolimus significantly reduced cell viability (by approximately 30%; P<0.05 versus control), inhibited p70S6K activity (-30%), and blocked IGF1 proliferative effects. Everolimus also significantly reduced CgA (by approximately 20%) and VEGF (by approximately 15%) secretion. Cotreatment with SOM230 did not exert additive effects on cell viability and secretory activity. AKT expression was similar in responder and nonresponder tissues, while mTOR expression was significantly higher in the responder group, which was characterized by higher CgA plasma levels and bigger tumors with higher mitotic index and angiogenesis. Our data demonstrate that everolimus reduces VEGF secretion and cell viability in BCs with a mechanism likely involving IGF1 signaling, suggesting that it might represent a possible medical treatment for BCs.


Asunto(s)
Neoplasias de los Bronquios/tratamiento farmacológico , Tumor Carcinoide/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Inmunosupresores/farmacología , Sirolimus/análogos & derivados , Adulto , Anciano , Western Blotting , Neoplasias de los Bronquios/metabolismo , Neoplasias de los Bronquios/patología , Tumor Carcinoide/metabolismo , Tumor Carcinoide/patología , Línea Celular Tumoral , Everolimus , Femenino , Humanos , Técnicas para Inmunoenzimas , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Persona de Mediana Edad , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , Receptores de Somatostatina/genética , Receptores de Somatostatina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Quinasas S6 Ribosómicas 70-kDa/genética , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal , Sirolimus/farmacología , Serina-Treonina Quinasas TOR , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo
17.
Endocrinology ; 151(6): 2453-61, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20392828

RESUMEN

Mitotane therapy is associated with many side effects, including thyroid function perturbations mimicking central hypothyroidism, possibly due to laboratory test interference or pituitary direct effects of mitotane. We investigated whether increasing concentrations of mitotane in the therapeutic range might interfere with thyroid hormone assays and evaluated the effects of mitotane on a mouse TSH-producing pituitary cell line. TSH, free T(4), and free T(3) levels do not significantly change in sera from hypo-, hyper-, or euthyroid patients after addition of mitotane at concentrations in the therapeutic window. In the mouse TalphaT1 cell line, mitotane inhibits both TSH expression and secretion, blocks TSH response to TRH, and reduces cell viability, inducing apoptosis at concentrations in the therapeutic window. TRH is not capable of rescuing TalphaT1 cells from the inhibitory effects of mitotane on TSH expression and secretion, which appear after short time treatment and persist over time. Our results demonstrate that mitotane does not interfere with thyroid hormone laboratory tests but directly reduces both secretory activity and cell viability on pituitary TSH-secreting mouse cells. These data represent a possible explanation of the biochemical picture consistent with central hypothyroidism in patients undergoing mitotane therapy and open new perspectives on the direct pituitary effects of this drug.


Asunto(s)
Antineoplásicos Hormonales/farmacología , Antineoplásicos Hormonales/uso terapéutico , Mitotano/farmacología , Mitotano/uso terapéutico , Tirotrofos/efectos de los fármacos , Tirotrofos/metabolismo , Tirotropina/metabolismo , Animales , Antineoplásicos Hormonales/efectos adversos , Supervivencia Celular/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Humanos , Ratones , Mitotano/efectos adversos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Tirotrofos/citología , Tirotropina/genética , Hormona Liberadora de Tirotropina/farmacología , Tiroxina/sangre , Triyodotironina/sangre
18.
J Clin Endocrinol Metab ; 95(2): 968-76, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19965918

RESUMEN

CONTEXT: Pituitary adenomas can cause specific syndromes due to hormone excess and/or determine sellar mass symptoms. Pituitary cell growth can sometimes be influenced by medical therapy, such as for somatotroph adenomas treated with somatostatin analogs or prolactinomas treated with dopaminergic drugs. However, nonfunctioning pituitary adenomas (NFAs) are still orphans of medical therapy. Everolimus (RAD001), a derivative of rapamycin, is a well-known immunosuppressant drug, which has been recently shown to have antineoplastic activity in several human cancers. OBJECTIVE: The objective of the study was to investigate the possible antiproliferative effects of RAD001 in human NFAs. DESIGN: We collected 40 NFAs that were dispersed in primary cultures, treated without or with 1 nm to 1 microm RAD001, 10 nm cabergoline, 10 nm SOM230 (a somatostatin receptor multiligand), and/or 50 nm IGF-I. Cell viability and apoptosis were evaluated after 48 h, and vascular endothelial growth factor (VEGF) secretion was assessed after an 8-h incubation. Somatostatin and dopamine subtype 2 receptor expression was investigated by quantitative PCR. RESULTS: In 28 cultures (70%), Everolimus significantly reduced cell viability (by approximately 40%; P < 0.05 vs. control), promoted apoptosis (+30%; P < 0.05 vs. control), inhibited p70S6K activity (-20%), and blocked IGF-I proliferative and antiapoptotic effects. In selected tissues cotreatment with SOM230, but not cabergoline, exerted an additive effect. Everolimus did not affect VEGF secretion but blocked the stimulatory effects of IGF-I on this parameter. CONCLUSIONS: Everolimus reduced NFA cell viability by inducing apoptosis, with a mechanism likely involving IGF-I signaling but not VEGF secretion, suggesting that it might represent a possible medical treatment of invasive/recurrent NFAs.


Asunto(s)
Adenoma/tratamiento farmacológico , Inmunosupresores/farmacología , Neoplasias Hipofisarias/tratamiento farmacológico , Sirolimus/análogos & derivados , Adenoma/patología , Anciano , Apoptosis/efectos de los fármacos , Cabergolina , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ergolinas/farmacología , Everolimus , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neoplasias Hipofisarias/patología , Receptores de Somatostatina/fisiología , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Sirolimus/farmacología , Somatostatina/análogos & derivados , Somatostatina/farmacología , Factor A de Crecimiento Endotelial Vascular/metabolismo
19.
Eur J Endocrinol ; 161(3): 467-73, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19574281

RESUMEN

OBJECTIVE: Papillary thyroid carcinoma (PTC) represents the majority of differentiated thyroid cancers, presenting the V600E activating BRAF mutation in 29-83% of cases. The aim of our study is to analyze the influence of BRAF mutation analysis on the diagnostic accuracy of fine-needle aspiration biopsy (FNAB) in patients with suspected PTC. DESIGN AND METHODS: Thyroid cytoaspirates from 469 nodules (size: 1.1+/-0.8 cm) with ultrasonographic features suspicious of malignant lesion, performed in 374 patients, were submitted to cytological evaluation and to biomolecular analysis, carried out after somatic DNA isolation, specific PCR amplification, and subsequent automated direct sequencing. All PCR fragments were also processed by specific enzyme restriction analysis. RESULTS: BRAF V600E mutation was found in 48 samples, 41 of which were also cytologically diagnosed as PTC, with histologic confirmation after thyroidectomy. Total thyroidectomy was perfomed also in seven patients with negative cytology but positive BRAF mutation, with histological confirmation of PTC in all. Among the 429 BRAF-negative samples, 407 had negative cytology for PTC, while 22 were diagnosed as suspected PTC and underwent total thyroidectomy with histological diagnosis of PTC in 17 and benign lesion in five. The prevalence of BRAF V600E mutation among histologically diagnosed PTC patients was 64%. Biomolecular analysis significantly increased cytology sensitivity for PTC from 77.3 to 86.7% (P<0.01). CONCLUSIONS: These data indicate that BRAF V600E mutation analysis can significantly improve FNAB diagnostic accuracy. However, biomolecular analysis is complementary to cytology, which should always be performed.


Asunto(s)
Carcinoma Papilar/diagnóstico , Carcinoma Papilar/genética , Análisis Mutacional de ADN , Proteínas Proto-Oncogénicas B-raf/genética , Neoplasias de la Tiroides/diagnóstico , Neoplasias de la Tiroides/genética , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Algoritmos , Biopsia con Aguja Fina , Carcinoma Papilar/patología , Análisis Mutacional de ADN/métodos , Femenino , Ácido Glutámico/genética , Humanos , Masculino , Persona de Mediana Edad , Polimorfismo de Longitud del Fragmento de Restricción , Sensibilidad y Especificidad , Neoplasias de la Tiroides/patología , Valina/genética , Adulto Joven
20.
Semin Reprod Med ; 26(6): 453-60, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18951327

RESUMEN

This review reports the current knowledge of microRNA (miRNA) expression in pituitary adenomas, focusing on recent microarray data. Moreover, a discussion is provided concerning the possible role of validated and putative targets of the most dysregulated miRNA in pituitary adenoma pathogenesis.


Asunto(s)
Adenoma/genética , MicroARNs/fisiología , Neoplasias Hipofisarias/genética , Análisis por Conglomerados , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , MicroARNs/genética , Análisis de Secuencia por Matrices de Oligonucleótidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...