Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Ann Oncol ; 31(6): 815-821, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32165204

RESUMEN

BACKGROUND: The term germline is commonly used to refer to any non-tumor control sample analyzed in tumor-normal paired sequencing experiments. Blood is the most commonly utilized control, and variants found in both tumor and blood are considered germline. However, somatic variants accumulate within an organism from embryogenesis throughout life. The resultant mosaicism is extensive and calls into question the assumption that blood, or any somatic tissue, represents the germline. Misclassification of germline and somatic variants has critical consequences for individual patient care and enormous impact on our health care system, given potential screening, counseling, and treatment implications of misidentifying germline variants. PATIENTS AND METHODS: Whole-exome sequencing was performed on six separate specimens from each of two patients with papillary thyroid carcinoma, and three specimens each from eight additional patients forming a validation cohort. Tumor variants were compared with each individual non-tumor control and with composite control sets generated as approximations of true germline. For the index patient, parental blood was also sequenced to assess whether patient-only samples could approximate a trio-derived germline. RESULTS: Using different non-tumor control tissues results in altered germline-somatic designation of tumor variants. In patient 1, 82% of variants are labeled germline using blood control, compared with 75.8%, 61.5%, and 49.6% using lymph node, thyroid, and thymus, respectively. In patient 2, the thyroid control resulted in the greatest percentage of germline calls (70.0%), followed by thymus (56.0%), lymph node (50.1%), and blood (44.1%). Composite control sets built from multiple samples can approximate the germline, even in the absence of parental DNA. CONCLUSIONS: Misclassification of germline-somatic origin has potential consequences for patient care, informing screening, trial eligibility, prophylactic interventions, and family planning. This study demonstrates the need for caution in interpreting germline-somatic designation if these data are to inform clinical decisions and suggests that improved design of controls can overcome current limitations.


Asunto(s)
Células Germinativas , Neoplasias de la Tiroides , Estudios de Cohortes , Mutación de Línea Germinal , Humanos , Cáncer Papilar Tiroideo/genética , Neoplasias de la Tiroides/genética , Secuenciación del Exoma
2.
J Intern Med ; 280(6): 540-550, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27864864

RESUMEN

The recently available genomic sequencing techniques have led to breakthroughs in understanding of the underlying genetic mechanisms in adrenocortical tumours. Disease-causing mutations have been described for aldosterone-producing adenomas, cortisol-producing adenomas and adrenocortical carcinomas. Further, knowledge gained from transcriptome analyses and methylation arrays has provided new insights into the development of these tumours. Elucidation of the genomic landscape of adrenocortical tumours and improved techniques may in the future be useful for early diagnosis through the detection of mutated DNA in the circulation. Moreover, compounds that bind specifically to altered proteins may be used as screening targets or therapeutic agents. Regulation of cortisol release by interaction with an altered subunit in adenylate cyclase may be more complex, but may provide a new option for regulating steroid release. Information about derangements in adrenocortical carcinoma is already helpful for determining patient prognosis. With further knowledge, we may be able to identify novel biomarkers that effectively and noninvasively help in differentiating between benign and malignant disease. It is clear that the next few years will provide much novel information that hopefully will aid in the treatment of patients with adrenocortical tumours.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/genética , Adenoma/genética , Adenoma/metabolismo , Carcinoma/genética , Análisis Mutacional de ADN , Genómica , Glucocorticoides/metabolismo , Humanos , Hiperaldosteronismo/genética
3.
ESMO Open ; 9(7): 103617, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38935991

RESUMEN

BACKGROUND: Adrenocortical carcinoma (ACC) is one of the most lethal endocrine malignancies and there is a lack of clinically useful markers for prognosis and patient stratification. Therefore our aim was to identify clinical and genetic markers that predict outcome in patients with ACC. METHODS: Clinical and genetic data from a total of 162 patients with ACC were analyzed by combining an independent cohort consisting of tumors from Yale School of Medicine, Karolinska Institutet, and Düsseldorf University (YKD) with two public databases [The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO)]. We used a novel bioinformatical pipeline combining differential expression and messenger RNA (mRNA)- and DNA-dependent survival. Data included reanalysis of previously conducted whole-exome sequencing (WES) for the YKD cohort, WES and RNA data for the TCGA cohort, and RNA data for the GEO cohort. RESULTS: We identified 3903 significant differentially expressed genes when comparing ACC and adrenocortical adenoma, and the mRNA expression levels of 461/3903 genes significantly impacted survival. Subsequent analysis revealed 45 of these genes to be mutated in patients with significantly worse survival. The relationship was significant even after adjusting for stage and age. Protein-protein interaction showed previously unexplored interactions among many of the 45 proteins, including the cancer-related proteins DNA polymerase delta 1 (POLD1), aurora kinase A (AURKA), and kinesin family member 23 (KIF23). Furthermore 14 of the proteins had significant interactions with TP53 which is the most frequently mutated gene in the germline of patients with ACC. CONCLUSIONS: Using a multiparameter approach, we identified 45 genes that significantly influenced survival. Notably, many of these genes have protein interactions not previously implicated in ACC. These findings may lay the foundation for improved prognostication and future targeted therapies.


Asunto(s)
Neoplasias de la Corteza Suprarrenal , Carcinoma Corticosuprarrenal , Humanos , Carcinoma Corticosuprarrenal/genética , Carcinoma Corticosuprarrenal/mortalidad , Neoplasias de la Corteza Suprarrenal/genética , Neoplasias de la Corteza Suprarrenal/mortalidad , Femenino , Masculino , Persona de Mediana Edad , Adulto , Pronóstico , Genómica/métodos , Secuenciación del Exoma , Perfil Genético , Anciano , Biomarcadores de Tumor/genética , Regulación Neoplásica de la Expresión Génica , Mutación , Perfilación de la Expresión Génica
4.
Nat Med ; 1(12): 1309-11, 1995 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-7489414

RESUMEN

Vitamin D and parathyroid hormone (PTH) constitute the main regulators of systemic calcium homeostasis. As well as its calcaemic effects, active vitamin D3(1,25(OH)2D3) has a direct regulatory role on parathyroid cells. Active vitamin D3 acts via its receptor (VDR), and binding of the ligand-receptor complex to specific promoter regions of the PTH gene inhibits transcription. Active vitamin D3 constitutes a principal regulator of parathyroid cell growth, and polymorphism in the VDR gene has recently been related to bone mineral density and suggested as predisposing to osteoporosis. Impaired effects of active vitamin D3 may contribute to the relatively enhanced secretion and cell proliferation seen in hyperparathyroidism (HPT). Indeed, VDR dysfunction, of essentially unknown character, has been demonstrated in the pathological parathyroid tissue of primary HPT as well as HPT secondary to uraemia. Consistent with the essential role of active vitamin D3 in parathyroid regulation, the VDR gene polymorphism was studied in 90 postmenopausal women with primary hyperparathyroidism. The VDR genotype bb was found in 60.0% of HPT patients and in 33.3% of the postmenopausal female controls (P < 0.001). As the b allele has been linked to decreased transcriptional activity or messenger RNA stability, reduced VDR expression may impede regulatory actions of vitamin D and may contribute to parathyroid tumorigenesis in these patients.


Asunto(s)
Hiperparatiroidismo/genética , Receptores de Calcitriol/genética , Anciano , Secuencia de Bases , Calcitriol/fisiología , Cartilla de ADN , Femenino , Genotipo , Humanos , Hiperparatiroidismo/sangre , Hiperparatiroidismo/metabolismo , Datos de Secuencia Molecular , Glándulas Paratiroides/metabolismo , Hormona Paratiroidea/sangre , Hormona Paratiroidea/genética , Polimorfismo Genético , Posmenopausia , Receptores de Calcitriol/fisiología
5.
Cancer Res ; 61(22): 8094-9, 2001 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-11719434

RESUMEN

The retinoblastoma protein-interacting zinc finger gene RIZ1 is a tumor suppressor gene and a member of a nuclear histone/protein methyltransferase superfamily. RIZ1 inactivation is commonly found in many types of human cancers and occurs through loss of mRNA expression, frameshift mutation, chromosomal deletion, and missense mutation. RIZ1 is also a tumor susceptibility gene in mice. We now show that loss of RIZ1 mRNA in human cancers is associated with DNA methylation of its promoter CpG island. Methylation of the RIZ1 promoter strongly correlated with lost or decreased RIZ1 mRNA expression in breast, liver, colon, and lung cancer cell lines as well as in liver cancer tissues. Treatment with the methylation inhibitor 5-aza-2'-deoxycytidine activated RIZ1 mRNA expression in cancer cells. Furthermore, methylation was found in 11 of 25 (44%) breast cancer specimens and 20 of 32 (62%) liver cancer specimens. Our results suggest that DNA methylation is a common mechanism in inactivating the RIZ1 tumor suppressor gene in human liver and breast cancers.


Asunto(s)
Azacitidina/análogos & derivados , Metilación de ADN , Proteínas de Unión al ADN , Genes Supresores de Tumor , N-Metiltransferasa de Histona-Lisina , Metiltransferasas/genética , Neoplasias/genética , Proteínas Nucleares/genética , Factores de Transcripción , Animales , Azacitidina/farmacología , Secuencia de Bases , Clonación Molecular , Islas de CpG/genética , ADN de Neoplasias/genética , Decitabina , Inhibidores Enzimáticos/farmacología , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Histona Metiltransferasas , Humanos , Metiltransferasas/biosíntesis , Datos de Secuencia Molecular , Neoplasias/enzimología , Neoplasias/metabolismo , Proteínas Nucleares/biosíntesis , Regiones Promotoras Genéticas , Proteína Metiltransferasas , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Ratas , Proteína de Retinoblastoma/genética , Proteína de Retinoblastoma/metabolismo , Dedos de Zinc/genética
6.
Cancer Res ; 58(3): 377-9, 1998 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-9458074

RESUMEN

Endocrine pancreatic tumors are rare neoplasms that occur sporadically or as part of a multiple endocrine neoplasia type 1 (MEN1) syndrome. Germ-line mutations of the MEN1 gene, located at 11q13, have been demonstrated in MEN1 kindreds, and loss of heterozygosity (LOH) on 11q13 together with somatic MEN1 mutations have been detected in 20% of nonfamilial parathyroid tumors. Here, we examine 11 non-MEN1 malignant tumors of the endocrine pancreas, 9 nonfunctioning tumors, and 2 glucagonomas. LOH of at least one informative locus on 11q13 was found in 70% of the tumors. Three tumors displayed somatic mutations of the MEN1 gene together with LOH on 11q13, whereas the corresponding germ-line DNA was normal. These findings support the hypothesis that MEN1 gene mutations contribute to the tumorigenesis of nonfamilial, malignant endocrine pancreatic tumors.


Asunto(s)
Glucagonoma/genética , Proteínas de Neoplasias/genética , Neoplasias Pancreáticas/genética , Proteínas Proto-Oncogénicas , Adulto , Anciano , Cromosomas Humanos Par 11/genética , ADN de Neoplasias/genética , Femenino , Humanos , Pérdida de Heterocigocidad , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/deficiencia , Reacción en Cadena de la Polimerasa
8.
Trends Endocrinol Metab ; 12(2): 53-8, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11167122

RESUMEN

Primary hyperparathyroidism (pHPT), generally caused by a monoclonal parathyroid adenoma, is a common endocrinopathy. Until recently, the genesis of the disease was poorly understood but during the past decade the molecular pathology of parathyroid tumor development has begun to be unveiled. This review summarizes recent advances in our understanding of genetic predisposition to pHPT, and the role of vitamin D receptor gene (VDR) variants in development of the disease. It has been shown that the multiple endocrine neoplasia tumor suppressor gene (MEN1) is mutated in parathyroid adenomas, and overexpression of the cyclin D1 oncogene [PRAD1 (parathyroid adenoma 1)] seems to contribute to parathyroid tumorigenesis. Several familial hyperparathyroid disorders have been studied, and the identification and characterization of the disease-causing genes have contributed to our understanding of parathyroid physiology and pathophysiology.


Asunto(s)
Neoplasias de las Paratiroides/genética , Adenoma/genética , Ciclina D1/genética , Predisposición Genética a la Enfermedad , Humanos , Hiperplasia , Neoplasia Endocrina Múltiple Tipo 1/genética , Mutación , Glándulas Paratiroides/patología , Receptores de Calcitriol/genética
9.
J Clin Endocrinol Metab ; 83(7): 2255-9, 1998 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-9661591

RESUMEN

Calcitriol, via its receptor (VDR) is a main regulator of PTH secretion and parathyroid cell proliferation. Recently, marked overrepresentation of the polymorphic VDR alleles b, a, and T was found in patients with primary hyperparathyroidism (pHPT), which suggests pathogenic importance in the disease. Using the ribonuclease protection assay, relative VDR and PTH messenger ribonucleic acid (mRNA) levels of parathyroid adenomas from 42 patients with sporadic pHPT were related to these VDR polymorphisms. The tumors of patients homozygous for the b, a, or T alleles demonstrated significantly lower VDR and higher PTH mRNA levels than those exhibiting the BB, AA, or tt genotypes (P < 0.0001-0.02), whereas heterozygotes had intermediate values. A similar discrepancy was found when comparing the baT and non-baT haplotypes (0.042 +/- 0.005 vs. 0.064 +/- 0.004 for VDR; 34.4 +/- 3.7 vs. 21.6 +/- 2.2 for PTH; both P < 0.005). The lower VDR mRNA levels associated with the b, a, and T alleles may affect the calcitriol-mediated control of parathyroid function and thereby contribute to the development of sporadic pHPT.


Asunto(s)
Adenoma/genética , Hormona Paratiroidea/genética , Neoplasias de las Paratiroides/genética , Polimorfismo Genético , ARN Mensajero/metabolismo , Receptores de Calcitriol/genética , Adenoma/metabolismo , Anciano , Alelos , Calcitriol/fisiología , Femenino , Haplotipos , Heterocigoto , Homocigoto , Humanos , Masculino , Neoplasias de las Paratiroides/metabolismo
10.
J Clin Endocrinol Metab ; 85(5): 2000-3, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10843188

RESUMEN

Vitamin D, via its receptor (VDR), inhibits the hormone secretion and proliferation of parathyroid cells. Vitamin D deficiency and reduced parathyroid VDR expression has been associated with development of hyperparathyroidism (HPT) secondary to uremia. VDR polymorphisms may influence VDR messenger RNA (mRNA) levels and have been coupled to an increased risk of parathyroid adenoma of primary HPT. VDR mRNA relative to glyceraldehyde-3-phosphate dehydrogenase mRNA levels were determined by RNase protection assay in 42 single parathyroid adenomas of patients with primary HPT, 23 hyperplastic glands of eight patients with uremic HPT, and 15 normal human parathyroid glands. The adenomas and hyperplasias demonstrated similar VDR mRNA levels, which were reduced (42 +/- 2.8% and 44 +/- 4.0%) compared with the normal glands (P < 0.0001). Comparison of parathyroid adenoma with a normal-sized parathyroid gland of the same individual (n = 3 pairs) showed a 20-58% reduction in the tumor. Nodularly enlarged glands represent a more advanced form of secondary HPT and showed greater reduction in the VDR mRNA levels than the diffusely enlarged glands (P < 0.005). The reduced VDR expression is likely to impair the 1,25(OH)2D3-mediated control of parathyroid functions, and to be of importance for the pathogenesis of not only uremic but also primary HPT. Circulating factors like calcium, PTH, and 1,25(OH)2D3 seem to be less likely candidates mediating the decreased VDR gene expression in HPT.


Asunto(s)
Adenoma/genética , Hiperparatiroidismo Secundario/genética , Hiperparatiroidismo/genética , Glándulas Paratiroides/metabolismo , Neoplasias de las Paratiroides/genética , Receptores de Calcitriol/genética , Adenoma/patología , Adenoma/cirugía , Anciano , Calcio/sangre , Humanos , Hiperparatiroidismo/metabolismo , Hiperparatiroidismo Secundario/metabolismo , Persona de Mediana Edad , Hormona Paratiroidea/sangre , Neoplasias de las Paratiroides/patología , Neoplasias de las Paratiroides/cirugía , ARN Mensajero/genética
11.
J Clin Endocrinol Metab ; 82(6): 1772-5, 1997 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-9177380

RESUMEN

Calcitriol acts via its receptor (VDR) and inhibits PTH secretion and parathyroid cell proliferation. Increased prevalence of the polymorphic VDR alleles b, a, and T has been demonstrated in sporadic primary hyperparathyroidism. Sixty-two patients with primary hyperparathyroidism due to parathyroid adenoma (mean age, 69.5 +/- 1.4 yr) were genotyped for these VDR polymorphisms. Dispersed cells of the adenomas were exposed to increasing concentrations of extracellular Ca2+ and analyzed for PTH release and cytoplasmic Ca2+ concentrations. Ca2+-mediated PTH inhibition exhibited higher ED50 and less suppression in the cells of patients who were homozygous for the b, a, and T alleles (P < 0.05-0.10). When analyzing haplotypes, the patients with baT demonstrated a ED50 of 1.81 +/- 0.15 vs. 1.29 +/- 0.10 for BAt (P < 0.05). As VDR alleles were unrelated to parathyroid intracellular Ca2+, influences of polymorphic VDR alleles on PTH secretion seem to involve mechanisms other than the Ca2+-sensing protein of the parathyroid cell surface.


Asunto(s)
Hiperparatiroidismo/genética , Hiperparatiroidismo/fisiopatología , Glándulas Paratiroides/fisiopatología , Polimorfismo Genético , Receptores de Calcitriol/genética , Adenoma/complicaciones , Adenoma/patología , Anciano , Alelos , Calcio/farmacología , Femenino , Genotipo , Heterocigoto , Humanos , Hiperparatiroidismo/etiología , Masculino , Glándulas Paratiroides/patología , Hormona Paratiroidea/antagonistas & inhibidores , Neoplasias de las Paratiroides/complicaciones , Neoplasias de las Paratiroides/patología
12.
J Clin Endocrinol Metab ; 84(5): 1690-4, 1999 May.
Artículo en Inglés | MEDLINE | ID: mdl-10323401

RESUMEN

Vitamin D regulates parathyroid cell proliferation and secretion of PTH. Increased prevalence of the polymorphic vitamin D receptor (VDR) alleles b, a, and T has been reported in sporadic primary hyperparathyroidism (PHPT), suggesting that these genetic variants may predispose to the disease. Recently, another polymorphism in the VDR gene was related to bone mineral density, and this VDR-FokI polymorphism causes different lengths of the VDR, implying possible functional consequences. The VDR-FokI polymorphism was studied in 182 postmenopausal women with sporadic PHPT and in matched controls. No significant differences in distribution of the VDR-FokI genotypes could be detected between the groups, although there was a tendency toward overrepresentation of the F allele in the PHPT patients (P = 0.05). There were no significant associations with age, serum calcium, serum PTH, bone mineral density, or parathyroid tumor weight. The VDR genotypes were unrelated to VDR and PTH messenger ribonucleic acid levels in the parathyroid adenomas of 42 PHPT patients. In 23 PHPT patients, the Ca2+-PTH set-points were determined in vivo and were unrelated to the VDR alleles. We suggest that the VDR-FokI polymorphism has at most a minor pathogenic importance in the development of PHPT.


Asunto(s)
Codón Iniciador/genética , Hiperparatiroidismo/genética , Glándulas Paratiroides/metabolismo , Polimorfismo Genético , ARN Mensajero/biosíntesis , Receptores de Calcitriol/genética , Adulto , Anciano , Anciano de 80 o más Años , Calcio/sangre , Calcio/metabolismo , Citratos , Desoxirribonucleasas de Localización Especificada Tipo II/biosíntesis , Desoxirribonucleasas de Localización Especificada Tipo II/genética , Femenino , Genotipo , Humanos , Hiperparatiroidismo/fisiopatología , Masculino , Persona de Mediana Edad , Glándulas Paratiroides/fisiopatología , Hormona Paratiroidea/biosíntesis , Hormona Paratiroidea/genética , Receptores de Calcitriol/biosíntesis
13.
J Clin Endocrinol Metab ; 85(5): 2042-7, 2000 May.
Artículo en Inglés | MEDLINE | ID: mdl-10843194

RESUMEN

Familial hyperparathyroidism (HPT), characterized by hypercalcemia and hypercalciuria, and familial benign hypocalciuric hypercalcemia (FHH) are the most common causes of hereditary hypercalcemia. The calcium-sensing receptor (CaR) regulates PTH secretion and renal calcium excretion. Heterozygous inactivating mutations of the gene cause FHH, whereas CaR gene mutations have not been demonstrated in HPT. In a kindred with 20 affected individuals, the hypercalcemic disorder segregated with inappropriately higher serum PTH and magnesium levels and urinary calcium levels than in unaffected members. Subtotal parathyroidectomy revealed parathyroid gland hyperplasia/adenoma and corrected the biochemical signs of the disorder in seven of nine individuals. Linkage analysis mapped the condition to markers flanking the CaR gene on chromosome 3q. Sequence analysis revealed a mutation changing phenylalanine to leucine at codon 881 of the CaR gene, representing the first identified point mutation located within the cytoplasmic tail of the CaR. A construct of the mutant receptor (F881L) was expressed in human embryonic kidney cells (HEK 293), and demonstrated a right-shifted dose-response relationship between the extracellular and intracellular calcium concentrations. The hypercalcemic disorder of the present family is caused by an inactivating point mutation in the cytoplasmic tail of the CaR and displays clinical characteristics atypical of FHH and primary HPT.


Asunto(s)
Trastornos del Metabolismo del Calcio/genética , Proteínas de Unión al Calcio/genética , Cromosomas Humanos Par 3 , Hipercalcemia/genética , Mutación Puntual , Adulto , Anciano , Sustitución de Aminoácidos , Calcio/orina , Línea Celular , Mapeo Cromosómico , ADN/sangre , Exones , Femenino , Marcadores Genéticos , Heterocigoto , Humanos , Masculino , Persona de Mediana Edad , Mutagénesis Sitio-Dirigida , Linaje , Proteínas Recombinantes/metabolismo , Transfección
14.
J Clin Endocrinol Metab ; 89(12): 6173-8, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15579774

RESUMEN

Low bone mineral density (BMD) is a major risk factor for osteoporotic fracture, and the trait is under genetic control by a large number of genes. It is recognized that estrogen plays an important role in the maintenance of bone mass by binding to estrogen receptor alpha (ERalpha). RIZ1 has previously been shown to be a specific ERalpha coactivator and strongly enhances its function both in vivo and in vitro. We performed in vitro studies comparing the abilities of RIZ1 P704 polymorphic variants (homozygous presence, P704+; absence, P704-; heterozygosity P704(+/-) of a proline at position 704) to coactivate the ERalpha and also examined the polymorphism associated to BMD of 343 Swedish women, aged 20-39 yr. The expression vector containing P704- RIZ1 showed an impaired response in coactivating ERalpha in a ligand- and dose-dependent manner compared with P704+ RIZ (P < 0.0001). The genotype frequencies were 19% (P704+), 32% (P704-), and 49% (P704(+/-)) and were in Hardy-Weinberg equilibrium. BMD at the heel was higher in the P704+ genotype group than in the P704(+/-) group (P = 0.02), which was evident also after corrections for fat and lean mass (P = 0.03). We conclude that RIZ1 may be a new candidate gene for involvement in the variation seen in BMD.


Asunto(s)
Densidad Ósea/genética , Proteínas de Unión al ADN/genética , Receptor alfa de Estrógeno/metabolismo , Eliminación de Gen , Proteínas Nucleares/genética , Polimorfismo Genético , Factores de Transcripción/genética , Adulto , Estudios de Cohortes , Femenino , Genotipo , N-Metiltransferasa de Histona-Lisina , Humanos , Distribución Aleatoria , Suecia
15.
J Clin Endocrinol Metab ; 83(8): 2960-3, 1998 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9709976

RESUMEN

Biochemical signs and severity of symptoms of primary hyperparathyroidism (pHPT) differ among patients, and little is known of any coupling of clinical characteristics of nonfamilial pHPT to genetic abnormalities in the parathyroid tumors. Mutations in the recently identified MEN1 gene at chromosome 11q13 have been found in parathyroid tumors of nonfamilial pHPT. Using microsatellite analysis for loss of heterozygosity (LOH) at 11q13 and DNA sequencing of coding exons, the MEN1 gene was studied in 49 parathyroid lesions of patients with divergent symptoms, operative findings, histopathological diagnosis, and biochemical signs of nonfamilial pHPT. Allelic loss at 11q13 was detected in 13 tumors, and 6 of them demonstrated previously unrecognized somatic missense and frameshift deletion mutations of the MEN1 gene. Many of the detected mutations would most likely result in a nonfunctional menin protein, consistent with a tumor suppressor mechanism. Clinical and biochemical characteristics of HPT were apparently unrelated to the presence or absence of LOH and the MEN1 gene mutations. However, the demonstration of LOH at 11q13 and MEN1 gene mutations in small parathyroid adenomas of patients with slight hypercalcemia and normal serum PTH levels suggest that altered MEN1 gene function may also be important for the development of mild sporadic pHPT.


Asunto(s)
Adenoma/genética , Hiperparatiroidismo/genética , Neoplasia Endocrina Múltiple Tipo 1/genética , Mutación , Neoplasias de las Paratiroides/genética , Anciano , Anciano de 80 o más Años , Cromosomas Humanos Par 11 , Femenino , Mutación del Sistema de Lectura , Eliminación de Gen , Humanos , Pérdida de Heterocigocidad , Masculino , Repeticiones de Microsatélite , Persona de Mediana Edad , Hormona Paratiroidea/sangre
16.
J Histochem Cytochem ; 45(3): 383-92, 1997 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-9071320

RESUMEN

We used riboprobes and monoclonal antibodies to characterize tissue distribution of the human 550-kD homologue to gp330/megalin, primarily identified in the rat kidney. Human gp330/megalin mRNA and protein are readily identified in human parathyroid cells, placental cytotrophoblasts, kidney proximal tubule cells, and epididymal epithelial cells. The immunoreactivity is found on the surface of the cells and is heterogeneously downregulated in parathyroid hyperplasia and adenomas. Cells of the proximal kidney tubule and epididymis express the protein on their luminal aspect. Moreover, the protein is expressed in Type II pneumocytes, mammary epithelial and thyroid follicular cells, and the ciliary body of the eye. Sequence analysis of cDNA fragments, obtained by RT-PCR, revealed identical nucleotide sequences in parathyroid, kidney, placenta, epididymis, and lung. Immunohistochemistry for parathyroid hormone-related protein (PTHrP) revealed partial co-expression with human gp330/megalin in parathyroid, placenta, and mammary gland. The findings substantiate human gp330/megalin expression in a variety of human tissues expected to possess calcium-sensing functions. It may constitute a protein of utmost importance to adult and fetal calcium homeostasis, although other important functions may also be coupled to this exceptionally large protein with highly restricted tissue distribution.


Asunto(s)
Calcio/metabolismo , Glicoproteínas de Membrana/metabolismo , Secuencia de Aminoácidos , Animales , Secuencia de Bases , ADN Complementario , Complejo Antigénico de Nefritis de Heymann , Humanos , Técnicas para Inmunoenzimas , Hibridación in Situ , Glicoproteínas de Membrana/genética , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Pruebas de Precipitina , Distribución Tisular
17.
Histol Histopathol ; 15(2): 619-27, 2000 04.
Artículo en Inglés | MEDLINE | ID: mdl-10809383

RESUMEN

Parathyroid gland is the overall regulatory organ within the systemic calcium homeostasis. Through cell surface bound calcium-sensing receptors external calcium inversely regulates release of parathyroid hormone (PTH). This mechanism, which is voltage independent and most sensitive around physiologic calcium concentrations, is regulated through a 120 kDa calcium sensing receptor, CaR. Inherited inactivation of this receptor is the cause for familial hypocalciuric hypercalcemia (FHH). Parallel research identified the 550 kDa glycoprotein megalin, which also is expressed on the parathyroid cell surface, as another potential calcium sensing protein. Although this protein expresses numerous calcium binding sites on its external domain, its main function may be calcium sensitive binding and uptake of steroid hormones, such as 25-OH-vitamin D3 (bound to vitamin D binding protein) and retinol. In hyperparathyroidism (HPT), excessive PTH is secreted and the calcium sensitivity of the cells reduced, i.e. the set-point, defined as the external calcium concentration at which half-maximal inhibition of PTH release occurs, shifted to the right. Pathological cells have reduced expression of both CaR and megalin, and reduced amount of intracellular lipids, possibly including stored steroid hormones. A number of possible genetic disturbances have been identified, indicating multifactorial reasons for the disease. In postmenopausal women, however, the individual group with highest incidence of disease, a causal relation to reduced effect of vitamin D is possible. An incipient renal insufficiency with age, lack of sunshine in the Northern Hemisphere, and an association to the baT haplotype of the vitamin D receptor supports this theory. This review summarizes data on regulation of PTH release, dysregulation in HPT, as well as proliferation of parathyroid cells.


Asunto(s)
Hiperparatiroidismo/fisiopatología , Animales , Femenino , Humanos , Hiperparatiroidismo/metabolismo , Hormona Paratiroidea/metabolismo , Ratas
18.
Surgery ; 122(6): 1101-5; discussion 1106, 1997 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9426425

RESUMEN

BACKGROUND: Estrogen and its receptor play a key role in calcium homeostasis, with implications in both the development and treatment of primary hyperparathyroidism. METHODS: Leukocyte DNA was isolated from 101 postmenopausal female patients with primary hyperparathyroidism and age-matched female control subjects (n = 101). Polymorphic estrogen receptor alleles denoted P/p and X/x were detected by polymerase chain reaction and restriction digestion with Pvu II and Xba 1 RESULTS: Allelic prevalences were similar in the patients and control subjects. In contrast to normocalcemic control subjects, patients with primary hyperthyroidism homozygous for alleles P and X displayed less elevated serum calcium concentrations (mean, 2.63 and 2.66 mmol/L) versus those with Pp and pp (2.72 mmol/L; p < 0.01) and Xx and xx (2.71; p < 0.05), respectively, as well as trends for higher intact serum parathyroid hormone values (p = 0.09). The PP genotype was accompanied by lower bone mineral density in the lumbar spine (p < 0.05). CONCLUSIONS: Estrogen receptor gene polymorphisms are presumably not directly involved in the parathyroid tumorigenesis. However, they seem to interact with the extent of biochemical derangements of primary hyperthyroidism by possible influences on both the peripheral action of parathyroid hormone and calcium regulation of its secretion.


Asunto(s)
Hiperparatiroidismo/genética , Receptores de Estrógenos/genética , Anciano , Femenino , Genotipo , Humanos , Persona de Mediana Edad , Polimorfismo Genético , Posmenopausia
19.
Surgery ; 118(6): 924-30; discussion 930-1, 1995 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-7491535

RESUMEN

BACKGROUND: Hyperparathyroidism affects most patients with multiple endocrine neoplasia type 1 (MEN 1). This study investigates expression of the candidate MEN1 gene phospholipase C beta 3 (PLC beta 3) and expression and function of a putative calcium sensing protein (CAS) in hyperparathyroidism of MEN 1. METHODS: In 31 parathyroid glands from 17 patients with MEN 1, CAS distribution was studied immunohistochemically and parallel sections were explored for PLC beta 3 mRNA expression by in situ hybridization. Enzymatically dispersed parathyroid cells were analyzed for cytoplasmic calcium concentrations [Ca2+]i and parathyroid hormone (PTH) release. RESULTS: All glands exhibited a heterogeneously reduced CAS immunoreactivity, especially meager in nodularly assembled parathyroid cells. Calcium regulated [Ca2+]i and PTH release tended to be more deranged in the glands possessing the lowest immunostaining. Parathyroid PLC beta 3 invariably was homogeneously expressed, and this included even MEN 1 patients with reduced PLC beta 3 expression in endocrine pancreatic tumors. CONCLUSIONS: The findings support variable calcium insensitivity of [Ca2+]i and PTH release in hyperparathyroidism of MEN 1, apparently coupled to heterogeneously reduced CAS expression. For clarification of the role of PLC beta 3 in MEN 1 parathyroid tumorigenesis further study of this protein is required.


Asunto(s)
Expresión Génica , Hiperparatiroidismo/genética , Neoplasia Endocrina Múltiple Tipo 1/genética , Receptores de Superficie Celular/genética , Fosfolipasas de Tipo C/genética , Adulto , Calcio/metabolismo , Femenino , Humanos , Hiperparatiroidismo/complicaciones , Inmunohistoquímica , Hibridación in Situ , Masculino , Persona de Mediana Edad , Neoplasia Endocrina Múltiple Tipo 1/complicaciones , Glándulas Paratiroides/química , Glándulas Paratiroides/metabolismo , Hormona Paratiroidea/metabolismo , ARN Mensajero/análisis , Receptores Sensibles al Calcio , Receptores de Superficie Celular/análisis , Fosfolipasas de Tipo C/análisis
20.
Lakartidningen ; 98(18): 2198-200, 2001 May 02.
Artículo en Sueco | MEDLINE | ID: mdl-11402599

RESUMEN

Primary hyperparathyroidism (pHPT) is commonly seen in postmenopausal women. Along with the clinical characterisation of the disease, studies of molecular genetics have contributed to increased understanding of the etiology of pHPT. Genetic association studies have revealed that certain vitamin D receptor polymorphisms relate to the development of sporadic pHPT. A new type of familial pHPT was recently discovered. Studies of parathyroid adenomas have demonstrated that the tumor suppressor gene MEN1 and the oncogene cyclinD1 are of importance for the tumorigenesis.


Asunto(s)
Predisposición Genética a la Enfermedad , Hiperparatiroidismo/genética , Posmenopausia , Anciano , Calcio/metabolismo , Femenino , Genes bcl-1 , Humanos , Hiperparatiroidismo/diagnóstico , Persona de Mediana Edad , Hormona Paratiroidea/genética , Factores de Riesgo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA