Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

País/Región como asunto
País de afiliación
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(19)2023 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-37834295

RESUMEN

Prostate cancer (PCa) has a high prevalence and represents an important health problem, with an increased risk of metastasis. With the advance of CRISPR-Cas9 genome editing, new possibilities have been created for investigating PCa. The technique is effective in knockout oncogenes, reducing tumor resistance. MMP9 and miR-21 target genes are associated with PCa progression; therefore, we evaluated the MMP-9 and miR-21 targets in PCa using the CRISPR-Cas9 system. Single guide RNAs (sgRNAs) of MMP9 and miR-21 sequences were inserted into a PX-330 plasmid, and transfected in DU145 and PC-3 PCa cell lines. MMP9 and RECK expression was assessed by qPCR, WB, and IF. The miR-21 targets, integrins, BAX and mTOR, were evaluated by qPCR. Flow cytometry was performed with Annexin5, 7-AAD and Ki67 markers. Invasion assays were performed with Matrigel. The miR-21 CRISPR-Cas9-edited cells upregulated RECK, MARCKS, BTG2, and PDCD4. CDH1, ITGB3 and ITGB1 were increased in MMP9 and miR-21 CRISPR-Cas9-edited cells. Increased BAX and decreased mTOR were observed in MMP9 and miR-21 CRISPR-Cas9-edited cells. Reduced cell proliferation, increased apoptosis and low invasion in MMP9 and miR-21 edited cells was observed, compared to Scramble. CRISPR-Cas9-edited cells of miR-21 and MMP9 attenuate cell proliferation, invasion and stimulate apoptosis, impeding PCa evolution.


Asunto(s)
Proteínas Inmediatas-Precoces , MicroARNs , Neoplasias de la Próstata , Masculino , Humanos , Edición Génica , Sistemas CRISPR-Cas/genética , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , ARN Guía de Sistemas CRISPR-Cas , Proteína X Asociada a bcl-2/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , MicroARNs/genética , MicroARNs/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Proteínas Inmediatas-Precoces/genética , Proteínas Supresoras de Tumor/genética , Proteínas de Unión al ARN/metabolismo
2.
Pituitary ; 24(2): 252-261, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33156432

RESUMEN

PURPOSE: Non-syndromic pituitary gigantism (PG) is a very rare disease. Aryl hydrocarbon receptor-interacting protein (AIP) and G protein-coupled receptor 101 (GPR101) genetic abnormalities represent important etiologic causes of PG and may account for up to 40% of these cases. Here, we aimed to characterize the clinical and molecular findings and long-term outcomes in 18 patients (15 males, three females) with PG followed at a single tertiary center in Sao Paulo, Brazil. METHODS: Genetic testing for AIP and GPR101 were performed by DNA sequencing, droplet digital PCR and array comparative genomic hybridization (aCGH). RESULTS: Pathogenic variants in the AIP gene were detected in 25% of patients, including a novel variant in splicing regulatory sequences which was present in a sporadic male case. X-LAG due to GPR101 microduplication was diagnosed in two female patients (12.5%). Of interest, these patients had symptoms onset by age 5 and 9 years old and diagnosis at 5 and 15 years, respectively. X-LAG, but not AIP, patients had a significantly lower age of symptoms onset and diagnosis and a higher height Z-score when compared to non-X-LAG. No other differences in clinical features and/or treatment outcomes were observed among PG based on their genetic background. CONCLUSION: We characterize the clinical and molecular findings and long-term outcome of the largest single-center PG cohort described so far.


Asunto(s)
Gigantismo/genética , Gigantismo/patología , Adolescente , Adulto , Brasil , Niño , Hibridación Genómica Comparativa , Femenino , Pruebas Genéticas , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Imagen por Resonancia Magnética , Masculino , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/patología , Receptores Acoplados a Proteínas G/genética , Adulto Joven
3.
Pituitary ; 21(4): 355-361, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29564694

RESUMEN

Sirtuins 1-7 (SIRT) are a highly conserved family of histone deacetylases involved in the regulation of longevity that have a considerable impact in transcription, DNA repair regulation, telomeric stability, cell senescence and apoptosis. In the present study, SIRT1-7 mRNA levels were evaluated in 37 somatotropinomas and 31 nonfunctioning pituitary adenomas (NFPAs) using qPCR and relation to tumor size, invasiveness and Ki-67 proliferative index was made. Overexpression of SIRT1 was observed in 86.5% of somatotropinomas versus 41.9% of NFPAs (P < 0.01). SIRT3 was more underexpressed in NFPAs than somatotropinomas (77.4 and 40.5%, respectively, P < 0.01) as well as SIRT4 and SIRT7. Despite the lack of association between sirtuins and invasiveness or Ki-67 index, SIRT1 and SIRT3 expressions were related to tumor size. Mean of the largest diameter was smaller in adenomas with SIRT1 overexpression than with normal expression (P < 0.01) and SIRT3 underexpression was associated with larger tumors (P < 0.01). In conclusion, a pronounced difference in sirtuins expression was identified between pituitary adenomas, suggesting that these genes are potential markers of pituitary adenomas and could be employed in the characterization of somatotropinomas and NFPAs. The role of sirtuins in pathogenesis of pituitary tumors merits further investigation and possibly will provide new molecular insight about their progression.


Asunto(s)
Adenoma/metabolismo , Neoplasias Hipofisarias/metabolismo , Sirtuinas/metabolismo , Adenoma/genética , Adenoma/patología , Adulto , Femenino , Adenoma Hipofisario Secretor de Hormona del Crecimiento/genética , Adenoma Hipofisario Secretor de Hormona del Crecimiento/metabolismo , Adenoma Hipofisario Secretor de Hormona del Crecimiento/patología , Histona Desacetilasas/genética , Histona Desacetilasas/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/patología , Sirtuina 1/genética , Sirtuina 1/metabolismo , Sirtuina 2/genética , Sirtuina 2/metabolismo , Sirtuina 3/genética , Sirtuina 3/metabolismo , Sirtuinas/genética
4.
Pituitary ; 20(3): 319-324, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27900634

RESUMEN

PURPOSE: Suppressor of cytokine signaling 2 (SOCS2) is a STAT5b-regulated gene and one of its functions is to influence growth and development through negative regulatory effects on GH/IGF-1 pathway. So, we evaluate the potential influence of SOCS2 single nucleotide polymorphisms (SNPs) on clinical and laboratorial characteristics of a large cohort of Brazilian patients with acromegaly. METHODS: Four SOCS2 SNPs (rs3782415, rs3816997, rs3825199 and rs11107116) were selected and genotyped by real-time PCR using specific Taqman probe assays. A total of 186 patients (116 women, age range 26-88 years) were evaluated. RESULTS: No association of SOCS2 genotypes was observed with none of the following clinical and laboratorial characteristics: age, sex, body mass index, comorbidities, basal GH, oral glucose tolerance test GH nadir, IGF-I, ULNR-IGF-I. CONCLUSION: Despite of the key role of SOCS2 in the regulation of GH receptor signaling, we did not find any significant association between SOCS2 polymorphisms and acromegaly.


Asunto(s)
Acromegalia/genética , Polimorfismo Genético/genética , Proteínas Supresoras de la Señalización de Citocinas/genética , Acromegalia/patología , Índice de Masa Corporal , Femenino , Genotipo , Humanos , Factor I del Crecimiento Similar a la Insulina , Masculino , Fenotipo , Transducción de Señal/genética
5.
Pituitary ; 18(5): 666-73, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25552351

RESUMEN

BACKGROUND: The association of GHR-exon 3 and -202 A/C IGFBP3 polymorphisms with clinical presentation, biochemical measurements and response to therapies in acromegaly have been suggested. OBJECTIVE: To evaluate the presence of these polymorphisms in acromegaly and their influence on clinical and laboratorial characteristics of patients at diagnosis and after treatment in a large cohort of acromegalic patients. PATIENTS AND METHODS: This is a cross-sectional study developed in a single tertiary reference center. Clinical data were obtained from the medical records of 186 acromegalic patients (116 women, age range 21-88 years). GH and IGF1 levels and GHR-exon 3 and -202 A/C IGFBP3 polymorphisms were evaluated in the same hospital. RESULTS: At diagnosis, serum GH concentrations were lower in patients with GHR-d3 genotype than those with GHR-fl, whereas an association of lower IGFBP3 levels with d3 allele was observed only after neurosurgical or medical treatments. However, these associations were not confirmed in posterior statistical analysis. CONCLUSION: Our results suggest that GHR-exon 3 and -202 A/C IGFBP3 polymorphisms did not show any consistent association on clinical and laboratorial features of acromegalic patients even after treatment.


Asunto(s)
Acromegalia/genética , Acromegalia/terapia , Proteínas Portadoras/genética , Exones , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Polimorfismo Genético , Acromegalia/sangre , Acromegalia/diagnóstico , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores/sangre , Brasil , Estudios Transversales , Femenino , Predisposición Genética a la Enfermedad , Hormona de Crecimiento Humana/sangre , Humanos , Masculino , Persona de Mediana Edad , Fenotipo , Valor Predictivo de las Pruebas , Centros de Atención Terciaria , Resultado del Tratamiento , Adulto Joven
6.
J Clin Endocrinol Metab ; 108(4): 876-887, 2023 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-36273993

RESUMEN

CONTEXT: Invasive and somatostatin receptor ligand (SRL)-resistant pituitary tumors represent a challenge in the clinical practice of endocrinologists. Efforts have been made to elucidate reliable makers for both. Survivin and eukaryotic translation initiation factor-binding protein 1 (4EBP1) are upregulated in several cancers and involved in apoptosis and cell proliferation. OBJECTIVE: We explored the role of these markers in somatotropinomas. METHODS: Immunostains for survivin and 4EBP1, and also for somatostatin receptor type 2 (SSTR2), Ki-67, and cytokeratin 18, were analyzed in tissue microarrays containing 52 somatotropinoma samples. Tumor invasiveness was evaluated in all samples while drug resistance was evaluated in 34 patients who received SRL treatment. All these parameters were correlated with first-generation SRL (fg-SRL) responsiveness and tumor invasiveness. RESULTS: Low survivin expression (P = 0.04), hyperintense signal on T2 weighted image (T2WI) (P = 0.01), younger age (P = 0.01), sparsely granular adenomas (SGA) (P = 0.04), high postoperative growth hormone (GH) and insulin-like growth factor-1 (IGF-1) levels (P = 0.049 and P < 0.001, respectively), and large postoperative tumor size (P = 0.02) were associated with resistance to fg-SRL. Low survivin and SSTR2 expression and high 4EBP1 expression were associated with SGA (P = 0.04, P = 0.01, and P = 0.001, respectively). Younger age (P = 0.03), large tumor pre- and postoperative (P = 0.04 and P = 0.006, respectively), low SSTR2 expression (P = 0.03), and high baseline GH and IGF-1 (P = 0.01 and P = 0.02, respectively) were associated with tumor invasiveness. However, survivin, 4EBP1, Ki-67, and granulation patterns were not associated with tumor invasion. CONCLUSION: This study suggests that low survivin expression is predictive of resistance to fg-SRL in somatotropinomas, but not of tumor invasiveness.


Asunto(s)
Acromegalia , Adenoma , Hormona de Crecimiento Humana , Neoplasias Hipofisarias , Humanos , Receptores de Somatostatina/metabolismo , Somatostatina/uso terapéutico , Factor I del Crecimiento Similar a la Insulina , Acromegalia/tratamiento farmacológico , Survivin/uso terapéutico , Antígeno Ki-67 , Adenoma/metabolismo , Hormona de Crecimiento Humana/uso terapéutico , Neoplasias Hipofisarias/tratamiento farmacológico , Hormona del Crecimiento/uso terapéutico
7.
N Engl J Med ; 358(7): 709-15, 2008 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-18272894

RESUMEN

Gonadotropin-dependent, or central, precocious puberty is caused by early maturation of the hypothalamic-pituitary-gonadal axis. In girls, this condition is most often idiopathic. Recently, a G protein-coupled receptor, GPR54, and its ligand, kisspeptin, were described as an excitatory neuroregulator system for the secretion of gonadotropin-releasing hormone (GnRH). In this study, we have identified an autosomal dominant GPR54 mutation--the substitution of proline for arginine at codon 386 (Arg386Pro)--in an adopted girl with idiopathic central precocious puberty (whose biologic family was not available for genetic studies). In vitro studies have shown that this mutation leads to prolonged activation of intracellular signaling pathways in response to kisspeptin. The Arg386Pro mutant appears to be associated with central precocious puberty.


Asunto(s)
Mutación Puntual , Pubertad Precoz/genética , Receptores Acoplados a Proteínas G/genética , Proteínas Supresoras de Tumor/metabolismo , Secuencia de Aminoácidos , Niño , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Genes Dominantes , Heterocigoto , Humanos , Fosfatos de Inositol/biosíntesis , Kisspeptinas , Datos de Secuencia Molecular , Fosforilación , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Kisspeptina-1 , Análisis de Secuencia de ADN , Transducción de Señal/genética
8.
J Clin Endocrinol Metab ; 106(9): 2690-2706, 2021 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-33871626

RESUMEN

CONTEXT: Acromegaly can impair bone integrity, increasing the risk of vertebral fractures (VFs). OBJECTIVE: To evaluate the impact of isolated GH/IGF-I hypersecretion on bone turnover markers, Wnt inhibitors, bone mineral density (BMD), microarchitecture, bone strength and vertebral fractures in female patients with acromegaly (Acro), compared with healthy control group (HC). DESIGN, SETTING, AND PATIENTS: Cross-sectional study including 83 premenopausal women without any pituitary deficiency:18 acromegaly in remission (AcroR), 12 in group with active acromegaly (AcroA), and 53 HC. Serum procollagen type 1 N-terminal propeptide, ß-carboxy-terminal crosslinked telopeptide of type 1 collagen, osteocalcin, sclerostin, and DKK1 were measured in blood samples. dual-energy X-ray absorptiometry, high-resolution peripheral quantitative computed tomography (HR-pQCT) and vertebral fractures evaluation were also assessed simultaneously. MAIN OUTCOME AND RESULTS: AcroA showed significantly lower sclerostin and higher DKK1 compared with HC. On HR-pQCT of tibia and radius, Acro showed impairment of trabecular (area and trabecular number), increased cortical porosity, and increased cortical area and cortical thickness compared with HC. The only significant correlation found with HR-pQCT parameters was a positive correlation between cortical porosity and serum DKK1 (R = 0.45, P = 0.044). Mild VFs were present in approximately 30% of patients. CONCLUSIONS: Eugonadal women with acromegaly without any pituitary deficiency showed increased cortical BMD, impairment of trabecular bone microstructure, and increased VF. Sclerostin was not correlated with any HR-pQCT parameters; however, DKK1 was correlated with cortical porosity in tibia (P = 0.027). Additional studies are needed to clarify the role of Wnt inhibitors on bone microarchitecture impairment in acromegaly.


Asunto(s)
Acromegalia/patología , Huesos/ultraestructura , Vía de Señalización Wnt/fisiología , Adulto , Densidad Ósea , Huesos/metabolismo , Estudios Transversales , Femenino , Análisis de Elementos Finitos , Humanos , Péptidos y Proteínas de Señalización Intercelular/sangre , Persona de Mediana Edad , Premenopausia , Fracturas de la Columna Vertebral/diagnóstico por imagen , Fracturas de la Columna Vertebral/etiología
9.
Ann Endocrinol (Paris) ; 80(2): 96-100, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30318257

RESUMEN

OBJECTIVES: This study analyzed the KISS1 c.-145delA (rs5780218) promoter polymorphism in a cohort of patients with growth hormone secreting pituitary adenoma (somatotropinoma) and controls, to investigate its role in the incidence of acromegaly and to assess patient/tumor characteristics. Material and methods rs5780218 allelic and genotypic distributions were compared between 49 somatotropinoma patients and 167 healthy controls. rs5780218 was also assessed in relation to patient characteristics and tumor aggressiveness, as characterized by tumor invasion and resistance to conventional therapy. The relationship between KISS1 mRNA expression and the rs5780218 genotype was also assessed in available pituitary tumor samples. RESULTS: The homozygous -/- variant genotype was associated with high rates of somatotropinoma (P<0.01), but not with tumor invasiveness, patient characteristics or hormonal remission. KISS1 mRNA expression was much lower in somatotropinomas carrying the deleted allele than in homozygous wild type AA. CONCLUSIONS: In this pilot study, the rs5780218 promoter polymorphism was evaluated in pituitary adenoma, and showed a possible association with the incidence of somatotropinoma but not with tumor progression.


Asunto(s)
Adenoma/genética , Adenoma Hipofisario Secretor de Hormona del Crecimiento/genética , Kisspeptinas/genética , Polimorfismo de Nucleótido Simple , Regiones Promotoras Genéticas/genética , Adenoma/epidemiología , Adenoma/patología , Adulto , Estudios de Casos y Controles , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Adenoma Hipofisario Secretor de Hormona del Crecimiento/epidemiología , Adenoma Hipofisario Secretor de Hormona del Crecimiento/patología , Humanos , Incidencia , Masculino , Persona de Mediana Edad , Proyectos Piloto , Polimorfismo de Nucleótido Simple/fisiología , Factores de Riesgo , Adulto Joven
10.
Am J Clin Oncol ; 42(2): 221-227, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30540568

RESUMEN

BACKGROUND: Complete tumor removal by transsphenoidal surgery is usually difficult for large nonfunctioning pituitary adenomas (NFPAs). A validated medical treatment may be useful for their management. This study evaluates the clinical efficacy of the dopaminergic agonist cabergoline for residual NFPA. DESIGN, SETTING, AND PARTICIPANTS: We conducted a randomized, parallel, open-label clinical trial that compared cabergoline with nonintervention in patients with residual NFPA after transsphenoidal surgery over 2 years. The primary outcome was clinical efficacy (tumor reduction). The secondary outcome was the relationship between tumor dopamine D2 receptor (D2R) expression and clinical responsiveness. Tumor measurements and clinical evaluations were performed every 6 months. RESULTS: In total, 59 and 57 individuals were randomly assigned to the study and control groups, respectively. At the end of the study, residual tumor shrinkage, stabilization, and enlargement were observed in 28.8%, 66.1%, and 5.1% of patients, respectively, in the medical-therapy group and in 10.5%, 73.7%, and 15.8% of patients, respectively, in the control group (P=0.01). The progression-free survival rate was 23.2 and 20.8 months for the study and control groups, respectively (P=0.01). D2R was not associated with cabergoline responsiveness. No major side effects were related to cabergoline use. CONCLUSIONS: Cabergoline was an effective drug for treating residual NFPA, and its use was associated with a high rate of tumor shrinkage (ClinicalTrials.gov NCT03271918).


Asunto(s)
Adenoma/tratamiento farmacológico , Antiparkinsonianos/uso terapéutico , Cabergolina/uso terapéutico , Neoplasia Residual/tratamiento farmacológico , Neoplasias Hipofisarias/tratamiento farmacológico , Adenoma/metabolismo , Adenoma/patología , Manejo de la Enfermedad , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Neoplasia Residual/metabolismo , Neoplasia Residual/patología , Neoplasias Hipofisarias/metabolismo , Neoplasias Hipofisarias/patología , Pronóstico , Receptores de Dopamina D2/metabolismo , Tasa de Supervivencia
11.
J Endocr Soc ; 3(9): 1682-1692, 2019 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-31528828

RESUMEN

CONTEXT: Von Hippel-Lindau (VHL) disease is an autosomal dominant syndrome caused by germline mutations in the VHL gene. Guidelines recommend pheochromocytoma (PHEO) biochemical screening should start at age 5 years. OBJECTIVE: Genotype-phenotype correlations in VHL, focusing on PHEO penetrance in children, were studied. DESIGN: We retrospectively evaluated 31 individuals (median age at diagnosis was 26 years) with diagnosed VHL disease. RESULTS: PHEO was diagnosed in six children with VHL. A large PHEO (5 cm) was detected in a 4-year-old boy with p.Gly114Ser mutation. PHEO penetrance was 55% starting at age 4 years. VHL missense mutations were identified in 11 of 22 families (50%), frameshift mutations in four (18.2%), stop codon in three (13.6%), splicing site in two (9.1%), and large gene deletion in two (9.1%). The codon 167 (n = 10) was a hotspot for VHL mutations and was significantly associated with PHEO (90% vs. 38%; P = 0.007). PHEOs and pancreatic neuroendocrine tumors (PNETs) were strongly associated with VHL missense mutations compared with other mutations (89.5% vs. 0% and 73.7% vs. 16.7%; P = 0.0001 and 0.002, respectively). In contrast, pancreatic cysts (91.7% vs. 26.3%; P = 0.0001), renal cysts (66.7% vs. 26.3%; P = 0.027), and central nervous system hemangioblastomas (91.7% vs. 47.3%; P = 0.012) were more frequent in VHL with nonmissense mutations. CONCLUSION: VHL missense mutations were highly associated with PHEO and PNETs. Our data support that in children with VHL harboring missense mutations, biochemical screening for PHEO should be initiated at diagnosis.

12.
J Endocrinol ; 187(3): 361-8, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16423815

RESUMEN

We report the results of molecular analysis in a series of twelve Kallmann syndrome (KS) and five normosmic hypogonadotropic hypogonadism (nHH) Brazilian patients. Kallman syndrome 1 (KAL-1) gene analysis was performed in all patients and the gonadotrophin releasing hormone receptor (GnRH-R) gene was investigated in nHH patients using PCR analysis with exon-flanking primers followed by automated sequencing techniques. Two-point mutations at the KAL-1 locus were found in two KS patients. One case exhibited a novel C deletion (del1956C) in exon 12 leading to a premature stop codon at position 617. The second case, a C to T transition at exon 5, showed a stop codon at aminoacid 191 (Arg191X). Renal agenesis and bimanual synkinesis, which are frequently found in patients with the KAL-1 mutation, were observed in these cases. Among the KS patients, two previously reported cases had intragenic deletions of exons 5-10, while a third patient had a KAL-1 gene microdeletion detected by fluorescence in situ hybridization. For the nHH patients, no abnormalities were observed at the exonic and flanking sequences of the KAL-1 or GnRH-R genes. Nasal embryonic LHRH factor (NELF) and early B-cell factor 2 (EBF2) exons were evaluated in KAL-1/GnRH-R mutation-negative cases (seven KS and five nHH) by sequence analysis but no mutations were identified in the coding regions in these patients. In conclusion, this report includes the description of a novel point mutation of the KAL-1 gene and suggests that the KAL-1 mutations and deletions might be more prevalent in KS Brazilian patients than previously described in other series. NELF and EBF2 genes have been considered good candidates for HH and a large number of patients need to be studied to assess their contribution to reproductive function.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Proteínas de la Matriz Extracelular/genética , Hipogonadismo/genética , Síndrome de Kallmann/genética , Proteínas del Tejido Nervioso/genética , Receptores LHRH/genética , Factores de Transcripción/genética , Aminoácidos/genética , Exones/genética , Eliminación de Gen , Humanos , Masculino , Mutación Puntual/genética , Polimorfismo Genético/genética , Análisis de Secuencia de ADN/métodos
13.
Med Sci Monit Basic Res ; 21: 179-82, 2015 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-26319784

RESUMEN

BACKGROUND DREAM (Downstream Regulatory Element Antagonistic Modulator) is a neuronal calcium sensor that was suggested to modulate TSH receptor activity and whose overexpression provokes an enlargement of the thyroid gland in transgenic mice. The aim of this study was to investigate somatic mutations and DREAM gene expression in human multinodular goiter (MNG). MATERIAL AND METHODS DNA and RNA samples were obtained from hyperplastic thyroid glands of 60 patients (54 females) with benign MNG. DREAM mutations were evaluated by PCR and direct automatic sequencing, whereas relative quantification of mRNA was performed by real-time PCR. Over- and under-expression were defined as a 2-fold increase and decrease in comparison to normal thyroid tissue, respectively. RQ M (relative quantification mean); SD (standard deviation). RESULTS DREAM expression was detected in all nodules evaluated. DREAM mRNA was overexpressed in 31.7% of MNG (RQ M=6.26; SD=5.08), whereas 53.3% and 15% had either normal (RQ M=1.16; SD=0.46) or underexpression (RQ M=0.30; SD=0.10), respectively. Regarding DREAM mutations analysis, only previously described intronic polymorphisms were observed. CONCLUSIONS We report DREAM gene expression in the hyperplastic thyroid gland of MNG patients. However, DREAM expression did not vary significantly, and was somewhat underexpressed in most patients, suggesting that DREAM upregulation does not significantly affect nodular development in human goiter.


Asunto(s)
Bocio Nodular/genética , Proteínas de Interacción con los Canales Kv/genética , Proteínas Represoras/genética , Adulto , Anciano , Anciano de 80 o más Años , Animales , ADN/genética , ADN/metabolismo , Femenino , Expresión Génica , Bocio Nodular/metabolismo , Humanos , Proteínas de Interacción con los Canales Kv/biosíntesis , Masculino , Ratones , Persona de Mediana Edad , Mutación , ARN/genética , ARN/metabolismo , Proteínas Represoras/biosíntesis , Adulto Joven
14.
Endocr Connect ; 4(2): 100-7, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25759380

RESUMEN

The genetic aetiology of congenital hypopituitarism (CH) is not entirely elucidated. FGFR1 and PROKR2 loss-of-function mutations are classically involved in hypogonadotrophic hypogonadism (HH), however, due to the clinical and genetic overlap of HH and CH; these genes may also be involved in the pathogenesis of CH. Using a candidate gene approach, we screened 156 Brazilian patients with combined pituitary hormone deficiencies (CPHD) for loss-of-function mutations in FGFR1 and PROKR2. We identified three FGFR1 variants (p.Arg448Trp, p.Ser107Leu and p.Pro772Ser) in four unrelated patients (two males) and two PROKR2 variants (p.Arg85Cys and p.Arg248Glu) in two unrelated female patients. Five of the six patients harbouring the variants had a first-degree relative that was an unaffected carrier of it. Results of functional studies indicated that the new FGFR1 variant p.Arg448Trp is a loss-of-function variant, while p.Ser107Leu and p.Pro772Ser present signalling activity similar to the wild-type form. Regarding PROKR2 variants, results from previous functional studies indicated that p.Arg85Cys moderately compromises receptor signalling through both MAPK and Ca(2) (+) pathways while p.Arg248Glu decreases calcium mobilization but has normal MAPK activity. The presence of loss-of-function variants of FGFR1 and PROKR2 in our patients with CPHD is indicative of an adjuvant and/or modifier effect of these rare variants on the phenotype. The presence of the same variants in unaffected relatives implies that they cannot solely cause the phenotype. Other associated genetic and/or environmental modifiers may play a role in the aetiology of this condition.

15.
J Pediatr (Rio J) ; 90(5): 500-5, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24818524

RESUMEN

OBJECTIVES: To evaluate the association between 3111T/C polymorphism of the CLOCK gene and the presence of obesity and sleep duration in children aged 6-13 years. In adults, this genetic variant has been associated with duration of sleep, ghrelin levels, weight, and eating habits. Although short sleep duration has been linked to obesity in children, no study has aimed to identify the possible molecular mechanisms of this association to date. METHODS: Weight, height, and circumferences were transformed into Z-scores for age and gender. Genotyping was performed using TaqMan methodology. A questionnaire regarding hours of sleep was provided to parents. The appropriate statistical tests were performed. RESULTS: This study evaluated 370 children (45% males, 55% females, mean age 8.5 ± 1.5 years). The prevalence of overweight was 18%. The duration of sleep was, on average, 9.7hours, and was inversely related to age (p<0.001). Genotype distribution was: 4% CC, 31% CT, and 65% TT. There was a trend toward higher prevalence of overweight in children who slept less than nine hours (23%) when compared to those who slept more than ten hours (16%, p=0.06). Genotype was not significantly correlated to any of the assessed outcomes. CONCLUSIONS: The CLOCK 3111T/C polymorphism was not significantly associated with overweight or sleep duration in children in this city.


Asunto(s)
Proteínas CLOCK/genética , Obesidad/genética , Sobrepeso/genética , Polimorfismo Genético/genética , Sueño/genética , Adolescente , Factores de Edad , Distribución de la Grasa Corporal , Niño , Estudios Transversales , Femenino , Frecuencia de los Genes , Genotipo , Humanos , Masculino , Estudiantes , Encuestas y Cuestionarios , Factores de Tiempo
16.
J Clin Endocrinol Metab ; 99(9): E1808-13, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24905066

RESUMEN

BACKGROUND: There is great interindividual variability in the response to GH therapy. Ascertaining genetic factors can improve the accuracy of growth response predictions. Suppressor of cytokine signaling (SOCS)-2 is an intracellular negative regulator of GH receptor (GHR) signaling. OBJECTIVE: The objective of the study was to assess the influence of a SOCS2 polymorphism (rs3782415) and its interactive effect with GHR exon 3 and -202 A/C IGFBP3 (rs2854744) polymorphisms on adult height of patients treated with recombinant human GH (rhGH). DESIGN AND PATIENTS: Genotypes were correlated with adult height data of 65 Turner syndrome (TS) and 47 GH deficiency (GHD) patients treated with rhGH, by multiple linear regressions. Generalized multifactor dimensionality reduction was used to evaluate gene-gene interactions. RESULTS: Baseline clinical data were indistinguishable among patients with different genotypes. Adult height SD scores of patients with at least one SOCS2 single-nucleotide polymorphism rs3782415-C were 0.7 higher than those homozygous for the T allele (P < .001). SOCS2 (P = .003), GHR-exon 3 (P= .016) and -202 A/C IGFBP3 (P = .013) polymorphisms, together with clinical factors accounted for 58% of the variability in adult height and 82% of the total height SD score gain. Patients harboring any two negative genotypes in these three different loci (homozygosity for SOCS2 T allele; the GHR exon 3 full-length allele and/or the -202C-IGFBP3 allele) were more likely to achieve an adult height at the lower quartile (odds ratio of 13.3; 95% confidence interval of 3.2-54.2, P = .0001). CONCLUSION: The SOCS2 polymorphism (rs3782415) has an influence on the adult height of children with TS and GHD after long-term rhGH therapy. Polymorphisms located in GHR, IGFBP3, and SOCS2 loci have an influence on the growth outcomes of TS and GHD patients treated with rhGH. The use of these genetic markers could identify among rhGH-treated patients those who are genetically predisposed to have less favorable outcomes.


Asunto(s)
Trastornos del Crecimiento/tratamiento farmacológico , Trastornos del Crecimiento/genética , Hormona de Crecimiento Humana/uso terapéutico , Proteínas Supresoras de la Señalización de Citocinas/genética , Síndrome de Turner/tratamiento farmacológico , Síndrome de Turner/genética , Adolescente , Adulto , Niño , Femenino , Pruebas Genéticas , Genotipo , Hormona del Crecimiento/uso terapéutico , Hormona de Crecimiento Humana/deficiencia , Humanos , Masculino , Polimorfismo de Nucleótido Simple , Resultado del Tratamiento
17.
Fertil Steril ; 102(3): 838-846.e2, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25016926

RESUMEN

OBJECTIVE: To analyze the GNRHR in patients with normosmic isolated hypogonadotropic hypogonadism (IHH) and constitutional delay of growth and puberty (CDGP). DESIGN: Molecular analysis and in vitro experiments correlated with phenotype. SETTING: Academic medical center. PATIENT(S): A total of 110 individuals with normosmic IHH (74 male patients) and 50 with CDGP. INTERVENTION(S): GNRHR coding region was amplified and sequenced. MAIN OUTCOME MEASURE(S): Novel variants were submitted to in vitro analysis. Frequency of mutations and genotype-phenotype correlation were analyzed. Microsatellite markers flanking GNRHR were examined in patients carrying the same mutation to investigate a possible founder effect. RESULT(S): Eleven IHH patients (10%) carried biallelic GNRHR mutations. In vitro analysis of novel variants (p.Y283H and p.V134G) demonstrated complete inactivation. The founder effect study revealed that Brazilian patients carrying the p.R139H mutation shared the same haplotype. Phenotypic spectrum in patients with GNRHR mutations varied from complete GnRH deficiency to partial and reversible IHH, with a relatively good genotype-phenotype correlation. One boy with CDGP was heterozygous for the p.Q106R variant, which was not considered to be pathogenic. CONCLUSION(S): GNRHR mutations are a frequent cause of congenital normosmic IHH and should be the first candidate gene for genetic screening in this condition, especially in autosomal recessive familial cases. The founder effect study suggested that the p.R139H mutation arises from a common ancestor in the Brazilian population. Finally, mutations in GNRHR do not appear to be involved in the pathogenesis of CDGP.


Asunto(s)
Trastornos del Crecimiento/genética , Mutación , Pubertad Tardía/genética , Receptores LHRH/genética , Adolescente , Animales , Células COS , Chlorocebus aethiops , Análisis Mutacional de ADN , Femenino , Frecuencia de los Genes , Estudios de Asociación Genética , Trastornos del Crecimiento/complicaciones , Humanos , Masculino , Polimorfismo de Nucleótido Simple , Pubertad Tardía/complicaciones
18.
Fertil Steril ; 100(3): 854-9, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23721716

RESUMEN

OBJECTIVE: To investigate the role of KAL1 abnormalities in Brazilian patients with Kallmann syndrome. DESIGN: In vitro experiments. SETTING: Academic medical center. PATIENT(S): One hundred fifteen Brazilian patients (98 men) with Kallmann syndrome. INTERVENTION(S): Peripheral blood leukocytes were used to obtain DNA. MAIN OUTCOME MEASURE(S): Direct sequencing and multiplex ligation-dependent probe amplification were used to identify KAL1 abnormalities. RESULT(S): We identified four KAL1 mutations (p.Met1?, p.Ala33Glyfs, p.Arg257*, and p.Trp462*) and two multiple exon deletions (exons 1-2 and 3-14) in six new male patients. Overall, 17 KAL1 defects (14.8%) were identified in the entire cohort of patients with Kallmann syndrome, including previously studied cases. KAL1-mutated patients presented with a more severe reproductive and nonreproductive phenotype (synkinesia, renal malformations, cryptorchidism, and anatomic olfactory abnormalities) in comparison with patients without KAL1 mutations. Intragenic deletions were one of the most often encountered defects (29.4%). These deletions can be missed by polymerase chain reaction (PCR) due to Yq11.2 KAL1 pseudogene (KALP) spurious amplification. CONCLUSION(S): These results indicate that intragenic multiexon deletions are one of the most frequent KAL1 abnormalities, which can be more accurately detected by multiplex ligation-dependent probe amplification. In addition, KAL1 sequencing results should be interpreted with caution, and stringency conditions of the PCR reaction should be adjusted to avoid pseudogene amplification.


Asunto(s)
Análisis Mutacional de ADN/métodos , Proteínas de la Matriz Extracelular/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Síndrome de Kallmann/genética , Reacción en Cadena de la Polimerasa Multiplex , Proteínas del Tejido Nervioso/genética , Adulto , Automatización , Secuencia de Bases , Análisis Mutacional de ADN/instrumentación , Femenino , Frecuencia de los Genes , Secuenciación de Nucleótidos de Alto Rendimiento/instrumentación , Humanos , Síndrome de Kallmann/diagnóstico , Síndrome de Kallmann/epidemiología , Masculino , Mutación/genética , Mutación/fisiología , Prevalencia , Seudogenes/genética
19.
Arq Bras Endocrinol Metabol ; 56(8): 519-24, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23295292

RESUMEN

Maturity-onset diabetes of the young (MODY) is characterized by an autosomal dominant mode of inheritance, early onset of hyperglycemia, and defects of insulin secretion. MODY subtypes described present genetic, metabolic, and clinical differences. MODY 2 is characterized by mild asymptomatic fasting hyperglycemia, and rarely requires pharmacological treatment. Hence, precise diagnosis of MODY is important for determining management and prognosis. We report two heterozygous GCK mutations identified during the investigation of short stature. Case 1: a prepubertal 14-year-old boy was evaluated for constitutional delay of growth and puberty. During follow-up, he showed abnormal fasting glucose (113 mg/dL), increased level of HbA1c (6.6%), and negative ß-cell antibodies. His father and two siblings also had slightly elevated blood glucose levels. The mother had normal glycemia. A GCK heterozygous missense mutation, p.Arg191Trp, was identified in the proband. Eighteen family members were screened for this mutation, and 11 had the mutation in heterozygous state. Case 2: a 4-year-old boy investigated for short stature revealed no other laboratorial alterations than elevated glycemia (118 mg/dL); ß-cell antibodies were negative. His father, a paternal aunt, and the paternal grandmother also had slightly elevated glycemia, whereas his mother had normal glycemia. A GCK heterozygous missense mutation, p.Glu221Lys, was identified in the index patient and in four family members. All affected patients had mild elevated glycemia. Individuals with normal glycemia did not harbor mutations. GCK mutation screening should be considered in patients with chronic mild early-onset hyperglycemia, family history of impaired glycemia, and negative ß-cell antibodies.


Asunto(s)
Diabetes Mellitus Tipo 2/genética , Glucoquinasa/genética , Heterocigoto , Hiperglucemia/genética , Adolescente , Preescolar , Enfermedad Crónica , Diabetes Mellitus Tipo 2/enzimología , Humanos , Hiperglucemia/enzimología , Masculino
20.
Arq Bras Endocrinol Metabol ; 56(8): 540-4, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23295295

RESUMEN

We report a novel GNRHR mutation in a male with normosmic isolated hypogonadotropic hypogonadism (nIHH). The coding region of the GNRHR gene was amplified and sequenced. Three variants p.[Asn10Lys;Gln11Lys]; [Tyr283His] were identified in the GNRHR coding region in a male with sporadic complete nIHH. The three variants were absent in the controls (130 normal adults). Familial segregation showed that the previously described p.Asn10Lys and p.Gln11Lys are in the same allele, in compound heterozygozity with the novel variant p.Tyr283His. The p.[Asn10Lys;Gln11Lys] are known inactivating mutations. The p.Tyr283His affects a well-conserved residue, and in silico analysis suggested it is a deleterious variant. We describe a novel GNRHR mutation in a male with nIHH. Absence of the mutation in the control group, conservation among species, in silico analysis, and familial segregation suggest that p.Tyr283His, which was identified in compound heterozygozity with the p.[Asn10Lys;Gln11Lys] variants, is an inactivating mutation.


Asunto(s)
Hipogonadismo/genética , Mutación/genética , Receptores LHRH/genética , Adolescente , Andrógenos/administración & dosificación , Estudios de Casos y Controles , Humanos , Hipogonadismo/tratamiento farmacológico , Masculino , Testosterona/administración & dosificación , Testosterona/análogos & derivados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA