Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Eur J Endocrinol ; 184(3): P1-P16, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33444221

RESUMEN

BACKGROUND: Corticotroph tumor progression (CTP) leading to Nelson's syndrome (NS) is a severe and difficult-to-treat complication subsequent to bilateral adrenalectomy (BADX) for Cushing's disease. Its characteristics are not well described, and consensus recommendations for diagnosis and treatment are missing. METHODS: A systematic literature search was performed focusing on clinical studies and case series (≥5 patients). Definition, cumulative incidence, treatment and long-term outcomes of CTP/NS after BADX were analyzed using descriptive statistics. The results were presented and discussed at an interdisciplinary consensus workshop attended by international pituitary experts in Munich on October 28, 2018. RESULTS: Data covered definition and cumulative incidence (34 studies, 1275 patients), surgical outcome (12 studies, 187 patients), outcome of radiation therapy (21 studies, 273 patients), and medical therapy (15 studies, 72 patients). CONCLUSIONS: We endorse the definition of CTP-BADX/NS as radiological progression or new detection of a pituitary tumor on thin-section MRI. We recommend surveillance by MRI after 3 months and every 12 months for the first 3 years after BADX. Subsequently, we suggest clinical evaluation every 12 months and MRI at increasing intervals every 2-4 years (depending on ACTH and clinical parameters). We recommend pituitary surgery as first-line therapy in patients with CTP-BADX/NS. Surgery should be performed before extrasellar expansion of the tumor to obtain complete and long-term remission. Conventional radiotherapy or stereotactic radiosurgery should be utilized as second-line treatment for remnant tumor tissue showing extrasellar extension.


Asunto(s)
Adenoma Hipofisario Secretor de ACTH/cirugía , Adenoma/cirugía , Adrenalectomía/efectos adversos , Síndrome de Nelson/etiología , Adenoma Hipofisario Secretor de ACTH/patología , Adenoma/patología , Progresión de la Enfermedad , Humanos , Síndrome de Nelson/patología
2.
Neuroendocrinology ; 110(9-10): 728-739, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32126547

RESUMEN

The parasellar region, located around the sella turcica, is an anatomically complex area representing a crossroads for important adjacent structures. Several lesions, including tumoral, inflammatory vascular, and infectious diseases may affect this area. Although invasive pituitary tumors are the most common neoplasms encountered within the parasellar region, other tumoral (and cystic) lesions can also be detected. Craniopharyngiomas, meningiomas, as well as Rathke's cleft cysts, chordomas, and ectopic pituitary tumors can primarily originate from the parasellar region. Except for hormone-producing ectopic pituitary tumors, signs and symptoms of these lesions are usually nonspecific, due to a mass effect on the surrounding anatomical structures (i.e., headache, visual defects), while a clinically relevant impairment of endocrine function (mainly anterior hypopituitarism and/or diabetes insipidus) can be present if the pituitary gland is displaced or compressed. Differential diagnosis of parasellar lesions mainly relies on magnetic resonance imaging, which should be interpreted by neuroradiologists skilled in base skull imaging. Neurosurgery is the main treatment, alone or in combination with radiotherapy. Of note, recent studies have identified gene mutations or signaling pathway modulators that represent potential candidates for the development of targeted therapies, particularly for craniopharyngiomas and meningiomas. In summary, parasellar lesions still represent a diagnostic and therapeutic challenge. A deeper knowledge of this complex anatomical site, the improvement of imaging tools, as well as novel insights into the pathophysiology of presenting lesions are strongly needed to improve the management of parasellar lesions.


Asunto(s)
Neoplasias Encefálicas , Seno Cavernoso , Neoplasias Hipofisarias , Silla Turca , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/terapia , Seno Cavernoso/patología , Humanos , Neoplasias Hipofisarias/diagnóstico , Neoplasias Hipofisarias/patología , Neoplasias Hipofisarias/terapia
3.
Neuroendocrinology ; 109(1): 57-69, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30677777

RESUMEN

Prolactin-secreting tumors (prolactinomas) represent the most common pituitary tumor type, accounting for 47-66% of functional pituitary tumors. Prolactinomas are usually benign and controllable tumors as they express abundant levels of dopamine type 2 receptor (D2), and can be treated with dopaminergic drugs, effectively reducing prolactin levels and tumor volume. However, a proportion of prolactinomas exhibit aggressive features (including invasiveness, relevant growth despite adequate dopamine agonist treatment, and recurrence potential) and few may exhibit metastasizing potential (carcinomas). In this context, the clinical, pathological, and molecular definitions of malignant and aggressive prolactinomas remain to be clearly defined, as primary prolactin-secreting carcinomas are similar to aggressive adenomas until the presence of metastases is detected. Indeed, standard molecular and histological analyses do not reflect differences between carcinomas and adenomas at a first glance and have limitations in prediction of the aggressive progression of prolactinomas, wherein the causes underlying the aggressive behavior remain unknown. Herein we present a comprehensive, multidisciplinary review of the most relevant epidemiological, clinical, pathological, genetic, biochemical, and molecular aspects of aggressive and malignant prolactinomas.


Asunto(s)
Neoplasias Hipofisarias/patología , Prolactinoma/patología , Femenino , Humanos , Masculino
4.
Endocr Pract ; 24(10): 907-914, 2018 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-30084690

RESUMEN

OBJECTIVE: Cushing disease is a rare severe condition caused by pituitary tumors that secrete adrenocorticotropic hormone (ACTH), leading to excessive endogenous glucocorticoid production. Tumors causing Cushing disease, also called corticotropinomas, are typically monoclonal neoplasms that mainly occur sporadically. METHODS: Literature review. RESULTS: Cushing disease is very rarely encountered in genetic familial syndromes. Oncogenes and tumor suppressor genes commonly associated with other tumor types are only rarely mutated in this tumor type. The advent of next-generation sequencing led to the identification of a single mutational hotspot in the ubiquitin-specific protease 8 ( USP8) gene in almost half of Cushing disease tumors. CONCLUSION: The new discoveries showcase a novel mechanism responsible for corticotroph tumorigenesis and ACTH hypersecretion and highlight USP8 and its downstream signaling pathways as potential promising pharmacologic targets for the management of Cushing disease. ABBREVIATIONS: ACTH = adrenocorticotropic hormone; BRG1 = Brahma-related gene 1; CABLES1 = CDK5 and ABL1 enzyme substrate 1; CD = Cushing disease; CNC = Carney complex; DICER1 = cytoplasmic endoribonuclease III; EGFR = epidermal growth factor receptor; GR = glucocorticoid receptor; IL = interleukin; MEN = multiple endocrine neoplasia; miRNA = microRNA; POMC = proopiomelanocortin; SSTR = somatostatin receptor; USP8 = ubiquitin-specific protease 8.


Asunto(s)
Adenoma Hipofisario Secretor de ACTH/genética , Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT)/genética , Neoplasias Hipofisarias/genética , Adenoma Hipofisario Secretor de ACTH/complicaciones , Adenoma Hipofisario Secretor de ACTH/metabolismo , Hormona Adrenocorticotrópica/metabolismo , Humanos , Mutación , Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT)/metabolismo , Neoplasias Hipofisarias/complicaciones , Neoplasias Hipofisarias/metabolismo , Transducción de Señal/genética
5.
Artículo en Inglés | MEDLINE | ID: mdl-29957855

RESUMEN

OBJECTIVE: Almost half of the cases of Cushing's disease (CD) tumours carry recurrent activating somatic mutations in the ubiquitin-specific protease eight gene (USP8). The USP8 mutational status could predict remission in patients with CD, so our objective was to correlate the presence of somatic USP8 mutations with the rate of recurrence after transsphenoidal surgery (TSS) retrospectively. DESIGN: Biochemical, radiological and clinical data were retrospectively assessed in 48 patients. USP8 mutational status was determined from corticotroph tumour samples. Association between USP8 mutational status, remission and recurrence was investigated. PATIENTS: Patients with Cushing's disease from a single-centre cohort who underwent TSS between 1991 and 2012. MEASUREMENTS: Long-term remission and recurrence rate after TSS with at least 6 months follow-up. Biochemical, radiological and clinical data, including sex, age at diagnosis, tumour size and pre-operative hormonal levels. USP8 mutational status. RESULTS: Patients with USP8 mutant corticotroph tumours (18 of 48; 37%) were diagnosed significantly earlier (mean ± SD 46 ± 10 years vs 53 ± 11 years; P = 0.028) and presented with higher pre-operative 24-hour urinary-free cortisol levels (median IQR µg/24 hours 1174.0, 1184.5 vs 480.0, 405.3; P = 0.045). The incidence of recurrence in a 10-year follow-up was significantly higher in patients with USP8 mutant tumours after the initial remission (58% vs 18% P = 0.026). Recurrence appeared significantly earlier in these patients (months 70, 44-97 95% CI vs 102, 86-119 95% CI; P = 0.019). CONCLUSION: Recurrence appears to be more frequent and earlier after TSS in patients with USP8 mutant corticotroph tumours.

6.
Eur J Endocrinol ; 178(1): 57-63, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28982703

RESUMEN

OBJECTIVE: Somatic mutations in the ubiquitin-specific protease 8 (USP8) gene are frequent in corticotroph tumors causing Cushing's disease (CD). Corticotroph tumor progression, the so-called Nelson's syndrome (NS), is a potentially life-threatening complication of bilateral adrenalectomy in patients with refractory CD that is caused by the development of an ACTH-secreting tumor of the pituitary gland. Whether USP8 alterations are also present in progressive Nelson's tumors has not been studied in detail so far. DESIGN AND METHODS: Retrospective, multicenter study involving tumors from 33 patients with progressive corticotroph tumors (29 females) and screening for somatic mutations on the mutational hotspot of the USP8 gene in the exon 14 with Sanger sequencing. RESULTS: Fifteen out of 33 tumors (45%) presented with a mutation in the exon 14 of USP8, with c.2159C>A (p.Pro720Gln) being the most frequent (9/33), followed by c.2155_2157delTCC (p.Ser718del, 4/33) and c.2152T>C (p.Ser718Pro, 2/33). This prevalence is similar to that previously reported for CD. Mutations were found exclusively in females. Other variables, such as age at diagnosis with NS, body mass index, hyperpigmentation, visual field defects, adenoma size or mortality, did not significantly differ between patients with wild-type and mutant tumors. Patients with USP8 mutant tumors exhibited higher levels of plasma ACTH after surgery (median: 640 vs 112 pg/mL, P = 0.03). No differences were observed in ACTH normalization (<50 pg/mL) and tumor control after surgery for Nelson's tumor. CONCLUSION: Somatic mutations in USP8 are common in Nelson's tumors, indicating that they do not drive the corticotroph tumor progression that leads to NS, and may be associated with a less favorable biochemical outcome after surgery for Nelson's tumor.


Asunto(s)
Carcinogénesis/genética , Progresión de la Enfermedad , Endopeptidasas/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Mutación/genética , Síndrome de Nelson/genética , Ubiquitina Tiolesterasa/genética , Hormona Adrenocorticotrópica/sangre , Adulto , Carcinogénesis/metabolismo , Estudios de Cohortes , Corticotrofos/fisiología , Femenino , Humanos , Masculino , Síndrome de Nelson/sangre , Síndrome de Nelson/cirugía , Estudios Retrospectivos , Adulto Joven
8.
J Clin Endocrinol Metab ; 101(8): 3010-7, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27270477

RESUMEN

CONTEXT: Somatic mutations have been found causative for endocrine autonomy in aldosterone-producing adenomas (APAs). Whereas mutations of PRKACA (catalytic subunit of protein kinase A) have been identified in cortisol-producing adenomas, the presence of PRKACA variants in APAs is unknown, especially in those that display cosecretion of cortisol. OBJECTIVE: The objective of the study was to investigate PRKACA somatic variants identified in APA cases. DESIGN: Identification of PRKACA somatic variants in APAs by whole-exome sequencing followed by in vitro analysis of the enzymatic activity of PRKACA variants and functional characterization by double immunofluorescence of CYP11B2 and CYP11B1 expression in the corresponding tumor tissues. SETTING AND PATIENTS: APA tissues were collected from 122 patients who underwent unilateral adrenalectomy for primary aldosteronism between 2005 and 2015 at a single institution. RESULTS: PRKACA somatic mutations were identified in two APA cases (1.6%). One APA carried a newly identified p.His88Asp variant, whereas in a second case, a p.Leu206Arg mutation was found, previously described only in cortisol-producing adenomas with overt Cushing's syndrome. Functional analysis showed that the p.His88Asp variant was not associated with gain of function. Although CYP11B2 was strongly expressed in the p.His88Asp-mutated APA, the p.Leu206Arg carrying APA predominantly expressed CYP11B1. Accordingly, biochemical Cushing's syndrome was present only in the patient with the p.Leu206Arg mutation. After adrenalectomy, both patients improved with a reduced number of antihypertensive medications and normalized serum potassium levels. CONCLUSIONS: We describe for the first time PRKACA mutations as rare findings associated with unilateral primary aldosteronism. As cortisol cosecretion occurs in a subgroup of APAs, other molecular mechanisms are likely to exist.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/genética , Neoplasias de la Corteza Suprarrenal/metabolismo , Adenoma Corticosuprarrenal/genética , Adenoma Corticosuprarrenal/metabolismo , Aldosterona/metabolismo , Subunidades Catalíticas de Proteína Quinasa Dependientes de AMP Cíclico/genética , Mutación Missense , Neoplasias de la Corteza Suprarrenal/sangre , Adenoma Corticosuprarrenal/sangre , Adulto , Sustitución de Aminoácidos , Células Cultivadas , Análisis Mutacional de ADN , Femenino , Frecuencia de los Genes , Células HEK293 , Humanos , Hiperaldosteronismo/genética , Hiperaldosteronismo/metabolismo , Metaboloma , Persona de Mediana Edad
9.
J Clin Endocrinol Metab ; 100(7): E997-1004, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25942478

RESUMEN

CONTEXT: We have recently reported somatic mutations in the ubiquitin-specific protease USP8 gene in a small series of adenomas of patients with Cushing's disease. OBJECTIVE: To determine the prevalence of USP8 mutations and the genotype-phenotype correlation in a large series of patients diagnosed with Cushing's disease. DESIGN: We performed a retrospective, multicentric, genetic analysis of 134 functioning and 11 silent corticotroph adenomas using Sanger sequencing. Biochemical and clinical features were collected and examined within the context of the mutational status of USP8, and new mutations were characterized by functional studies. PATIENTS: A total of 145 patients who underwent surgery for an ACTH-producing pituitary adenoma. MAIN OUTCOMES MEASURES: Mutational status of USP8. Biochemical and clinical features included sex, age at diagnosis, tumor size, preoperative and postoperative hormonal levels, and comorbidities. RESULTS: We found somatic mutations in USP8 in 48 (36%) pituitary adenomas from patients with Cushing's disease but in none of 11 silent corticotropinomas. The prevalence was higher in adults than in pediatric cases (41 vs 17%) and in females than in males (43 vs 17%). Adults having USP8-mutated adenomas were diagnosed at an earlier age than those with wild-type lesions (36 vs 44 y). Mutations were primarily found in adenomas of 10 ± 7 mm and were inversely associated with the development of postoperative adrenal insufficiency. All the mutations affected the residues Ser718 or Pro720, including five new identified alterations. Mutations reduced the interaction between USP8 and 14-3-3 and enhanced USP8 activity. USP8 mutants diminished epidermal growth factor receptor ubiquitination and induced Pomc promoter activity in immortalized AtT-20 corticotropinoma cells. CONCLUSIONS: USP8 is frequently mutated in adenomas causing Cushing's disease, especially in those from female adult patients diagnosed at a younger age.


Asunto(s)
Adenoma Hipofisario Secretor de ACTH/genética , Adenoma/genética , Endopeptidasas/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Mutación , Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT)/genética , Ubiquitina Tiolesterasa/genética , Adenoma Hipofisario Secretor de ACTH/epidemiología , Adenoma/epidemiología , Adolescente , Adulto , Animales , Células COS , Niño , Chlorocebus aethiops , Análisis Mutacional de ADN , Femenino , Frecuencia de los Genes , Estudios de Asociación Genética , Células HeLa , Humanos , Masculino , Ratones , Persona de Mediana Edad , Hipersecreción de la Hormona Adrenocorticotrópica Pituitaria (HACT)/epidemiología , Estudios Retrospectivos , Células Tumorales Cultivadas , Adulto Joven
10.
PLoS One ; 9(3): e91884, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24632820

RESUMEN

Recurrent breast cancer occurring after the initial treatment is associated with poor outcome. A bimodal relapse pattern after surgery for primary tumor has been described with peaks of early and late recurrence occurring at about 2 and 5 years, respectively. Although several clinical and pathological features have been used to discriminate between low- and high-risk patients, the identification of molecular biomarkers with prognostic value remains an unmet need in the current management of breast cancer. Using microarray-based technology, we have performed a microRNA expression analysis in 71 primary breast tumors from patients that either remained disease-free at 5 years post-surgery (group A) or developed early (group B) or late (group C) recurrence. Unsupervised hierarchical clustering of microRNA expression data segregated tumors in two groups, mainly corresponding to patients with early recurrence and those with no recurrence. Microarray data analysis and RT-qPCR validation led to the identification of a set of 5 microRNAs (the 5-miRNA signature) differentially expressed between these two groups: miR-149, miR-10a, miR-20b, miR-30a-3p and miR-342-5p. All five microRNAs were down-regulated in tumors from patients with early recurrence. We show here that the 5-miRNA signature defines a high-risk group of patients with shorter relapse-free survival and has predictive value to discriminate non-relapsing versus early-relapsing patients (AUC = 0.993, p-value<0.05). Network analysis based on miRNA-target interactions curated by public databases suggests that down-regulation of the 5-miRNA signature in the subset of early-relapsing tumors would result in an overall increased proliferative and angiogenic capacity. In summary, we have identified a set of recurrence-related microRNAs with potential prognostic value to identify patients who will likely develop metastasis early after primary breast surgery.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , MicroARNs/genética , Recurrencia Local de Neoplasia/genética , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/cirugía , Análisis por Conglomerados , Supervivencia sin Enfermedad , Femenino , Perfilación de la Expresión Génica , Ontología de Genes , Humanos , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Curva ROC , Factores de Tiempo
11.
Int J Oncol ; 41(6): 2200-6, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23076779

RESUMEN

Surgery is the primary treatment for non-metastatic breast cancer. However, the risk of early recurrence remains after surgical removal of the primary tumor. Recurrence is suggested to result from hidden micrometastatic foci, which are triggered to escape from dormancy by surgical resection of the primary tumor. In this study, we focused on the differential impact of breast surgery on the serum profiles of early breast cancer patients and healthy women. Serum samples from invasive breast cancer patients, in situ carcinoma breast cancer patients and healthy women were analyzed using reverse phase protein array technology. Samples were collected prior to breast surgery and 24 h following breast surgery. Both the expression level and the velocity of 42 serum proteins were quantified and compared among groups. We found that surgery increased the concentration of several proteins (CSF1, THSB2, IL6, IL7, IL16, FasL and VEGF-B) in the overall population. Compared with healthy women and patients with non-invasive tumors, invasive tumor patients exhibited higher preoperative levels of several serum proteins, such as αFP, IFNß1, VEGF-A, IL18, E-cadherin or CD31, and lower postoperative levels of TNFα and IL5. Similarly, we detected significant surgery-induced changes in the velocity of VEGF-A and IL16 accumulation in samples derived from invasive breast cancer patients. In conclusion, breast surgery induced distinct changes in the concentrations and dynamics of serum proteins in invasive breast cancer patients compared with healthy women and non-invasive tumor patients.


Asunto(s)
Proteínas Sanguíneas , Neoplasias de la Mama/sangre , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/sangre , Neoplasias de la Mama/patología , Neoplasias de la Mama/cirugía , Análisis por Conglomerados , Femenino , Humanos , Persona de Mediana Edad , Clasificación del Tumor , Estadificación de Neoplasias , Periodo Posoperatorio , Análisis por Matrices de Proteínas , Proteómica , Adulto Joven
12.
Cell Mol Life Sci ; 69(22): 3881-93, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22752157

RESUMEN

The spatial and temporal regulation of intracellular signaling is determined by the spatial and temporal organization of complexes assembled on scaffold proteins, which can be modulated by their interactions with additional proteins as well as subcellular localization. The scaffold KSR1 protein interacts with MAPK forming a complex that conveys a differential signaling in response to growth factors. The aim of this work is to determine the unknown mechanism by which VRK2A downregulates MAPK signaling. We have characterized the multiprotein complex formed by KSR1 and the Ser-Thr kinase VRK2A. VRK2A is a protein bound to the endoplasmic reticulum (ER) and retains a fraction of KSR1 complexes on the surface of this organelle. Both proteins, VRK2A and KSR1, directly interact by their respective C-terminal regions. In addition, MEK1 is also incorporated in the basal complex. MEK1 independently interacts with the CA5 region of KSR1 and with the N-terminus of VRK2A. Thus, VRK2A can form a high molecular size (600-1,000 kDa) stable complex with both MEK1 and KSR1. Knockdown of VRK2A resulted in disassembly of these high molecular size complexes. Overexpression of VRK2A increased the amount of KSR1 in the particulate fraction and prevented the incorporation of ERK1/2 into the complex after stimulation with EGF. Neither VRK2A nor KSR1 interact with the VHR, MKP1, MKP2, or MKP3 phosphatases. The KSR1 complex assembled and retained by VRK2A in the ER can have a modulatory effect on the signal mediated by MAPK, thus locally affecting the magnitude of its responses, and can explain differential responses depending on cell type.


Asunto(s)
Retículo Endoplásmico/metabolismo , MAP Quinasa Quinasa 1/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Sitios de Unión , Línea Celular Tumoral , Regulación hacia Abajo , Células HEK293 , Células HeLa , Humanos , Células MCF-7 , Complejos Multiproteicos/metabolismo , Unión Proteica , Proteínas Quinasas/genética , Proteínas Serina-Treonina Quinasas/genética , Estructura Cuaternaria de Proteína , Interferencia de ARN , ARN Interferente Pequeño
13.
BMC Cancer ; 10: 136, 2010 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-20385028

RESUMEN

BACKGROUND: Mutational analysis of the KRAS gene has recently been established as a complementary in vitro diagnostic tool for the identification of patients with colorectal cancer who will not benefit from anti-epidermal growth factor receptor (EGFR) therapies. Assessment of the mutation status of KRAS might also be of potential relevance in other EGFR-overexpressing tumors, such as those occurring in breast cancer. Although KRAS is mutated in only a minor fraction of breast tumors (5%), about 60% of the basal-like subtype express EGFR and, therefore could be targeted by EGFR inhibitors. We aimed to study the mutation frequency of KRAS in that subtype of breast tumors to provide a molecular basis for the evaluation of anti-EGFR therapies. METHODS: Total, genomic DNA was obtained from a group of 35 formalin-fixed paraffin-embedded, triple-negative breast tumor samples. Among these, 77.1% (27/35) were defined as basal-like by immunostaining specific for the established surrogate markers cytokeratin (CK) 5/6 and/or EGFR. KRAS mutational status was determined in the purified DNA samples by Real Time (RT)-PCR using primers specific for the detection of wild-type KRAS or the following seven oncogenic somatic mutations: Gly12Ala, Gly12Asp, Gly12Arg, Gly12Cys, Gly12Ser, Gly12Val and Gly13Asp. RESULTS: We found no evidence of KRAS oncogenic mutations in all analyzed tumors. CONCLUSIONS: This study indicates that KRAS mutations are very infrequent in triple-negative breast tumors and that EGFR inhibitors may be of potential benefit in the treatment of basal-like breast tumors, which overexpress EGFR in about 60% of all cases.


Asunto(s)
Biomarcadores de Tumor/análisis , Neoplasias de la Mama/genética , Mutación , Neoplasias Basocelulares/genética , Proteínas Proto-Oncogénicas/genética , Proteínas ras/genética , Neoplasias de la Mama/química , Neoplasias de la Mama/tratamiento farmacológico , Análisis Mutacional de ADN , Receptores ErbB/análisis , Receptores ErbB/antagonistas & inhibidores , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunohistoquímica , Queratina-5/análisis , Queratina-6/análisis , Neoplasias Basocelulares/química , Neoplasias Basocelulares/tratamiento farmacológico , Selección de Paciente , Reacción en Cadena de la Polimerasa , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas p21(ras) , Receptor ErbB-2/análisis , Receptores de Estrógenos/análisis , Receptores de Progesterona/análisis
14.
J Biol Chem ; 283(25): 17450-62, 2008 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-18426801

RESUMEN

Identifying 14-3-3 isoform-specific substrates and functions may be of broad relevance to cell signaling research because of the key role played by this family of proteins in many vital processes. A multitude of ligands have been identified, but the extent to which they are isoform-specific is a matter of debate. Herein we demonstrate, both in vitro and in vivo, a specific, functionally relevant interaction of human 14-3-3gamma with the molecular scaffold KSR1, which is mediated by the C-terminal stretch of 14-3-3gamma. Specific binding to 14-3-3gamma protected KSR1 from epidermal growth factor-induced dephosphorylation and impaired its ability to activate ERK2 and facilitate Ras signaling in Xenopus oocytes. Furthermore, RNA interference-mediated inhibition of 14-3-3gamma resulted in the accumulation of KSR1 in the plasma membrane, all in accordance with 14-3-3gamma being the cytosolic anchor that keeps KSR1 inactive. We also provide evidence that KSR1-bound 14-3-3gamma heterodimerized preferentially with selected isoforms and that KSR1 bound monomeric 14-3-3gamma. In sum, we have demonstrated ligand discrimination among 14-3-3 isoforms and shed light on molecular mechanisms of 14-3-3 functional specificity and KSR1 regulation.


Asunto(s)
Proteínas 14-3-3/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Quinasas/metabolismo , Animales , Células COS , Chlorocebus aethiops , Dimerización , Humanos , Ligandos , Modelos Moleculares , Fosforilación , Unión Proteica , Isoformas de Proteínas , Estructura Terciaria de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...