Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Acta Endocrinol (Buchar) ; 19(1): 115-124, 2023.
Article in English | MEDLINE | ID: mdl-37601716

ABSTRACT

Background: Molecular defects in the SHOX gene including deletions, duplications or pathogenic point mutations are responsible for well-known pathologies involving short stature as a clinical manifestation: Léri-Weill dyschondrosteosis, Langer mesomelic dysplasia, Turner syndrome or idiopathic short stature. Duplications flanking the SHOX gene (upstream or downstream of the intact SHOX gene involving conserved non-coding cis-regulatory DNA elements - CNEs) have been described but their clinical involvement is still difficult to understand. Results: We describe two cases with short stature and normal GH-IGF1 status. Multiplex ligation-dependent probe amplification (MLPA) and array comparative genomic hybridization (arrayCGH) identified in both cases heterozygous duplications involving downstream regions of SHOX gene, within CNEs (CNE8, CNE9 and CNE4, CNE5, CNE6, ECR1, CNE8, CNE9 and surrounding areas, respectively). One of the cases showed a maternally inherited duplication. Although every case has several particularities, we consider that duplications in these non-coding regions of SHOX gene may explain the short stature phenotype. Conclusion: To our knowledge, these are the first Romanian-reported cases of ISS with a large duplication of downstream SHOX enhancers CNEs region. The spectrum of phenotypic consequences and the exact mechanism of the presumed clinical expression of these genetic alterations still needs to be evaluated and described.

2.
Thyroid ; 11(1): 37-9, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11272095

ABSTRACT

Decrease or loss of the sodium iodide (Na+/I-) symporter (NIS) activity influences the suitability of using radioiodine to detect and treat metastatic thyroid tissues. In previous studies, the presence of the NIS transcript, albeit at lower expression levels, has been shown in most thyroid differentiated carcinomas. In this study we searched for point mutations or other genetic alterations that may be responsible for an altered function of the NIS protein in tumors that still express NIS transcripts. Tumoral cDNAs derived from seven differentiated thyroid carcinomas (DTC), five papillary and two follicular, were analyzed by direct sequencing after polymerase chain reaction (PCR) amplification of the structural gene of the Na+/I- symporter. Neither mutations nor other genetic abnormalities were detected in any tumor sample examined. The data indicate that mutations or other genetic alterations of the NIS structural gene are not a major cause of the reduced iodide uptake in DTC.


Subject(s)
Carrier Proteins/genetics , Membrane Proteins/genetics , Mutation , Symporters , Thyroid Neoplasms/genetics , Adenocarcinoma, Follicular/genetics , Carcinoma, Papillary/genetics , Cloning, Molecular , DNA, Complementary/chemistry , Gene Expression , Humans , Immunoblotting , Immunohistochemistry , Iodine Radioisotopes/metabolism , Point Mutation , Polymerase Chain Reaction , RNA, Messenger/analysis , Reverse Transcriptase Polymerase Chain Reaction , Sequence Analysis, DNA
3.
J Clin Endocrinol Metab ; 86(3): 1072-7, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11238488

ABSTRACT

Thyroid tumors are about 3 times more frequent in females than in males. Epidemiological studies suggest that the use of estrogens may contribute to the pathogenesis of thyroid tumors. In a very recent study a direct growth stimulatory effect of 17beta-estradiol was demonstrated in FRTL-5 rat thyroid cells. In this work the presence of estrogen receptors alpha and beta in thyroid cells derived from human goiter nodules and in human thyroid carcinoma cell line HTC-TSHr was demonstrated. There was no difference between the expression levels of estrogen receptor alpha in males and females, but there was a significant increase in expression levels in response to 17beta-estradiol. Stimulation of benign and malignant thyroid cells with 17beta-estradiol resulted in an increased proliferation rate and an enhanced expression of cyclin D1 protein, which plays a key role in the regulation of G(1)/S transition in the cell cycle. In malignant tumor cells maximal cyclin D1 expression was observed after 3 h, whereas in benign cells the effect of 17beta-estradiol was delayed. ICI 182780, a pure estrogen antagonist, prevented the effects of 17beta-estradiol. In addition, 17beta-estradiol was found to modulate activation of mitogen-activated protein (MAP) kinase, whose activity is mainly regulated by growth factors in thyroid carcinoma cells. In response to 17beta-estradiol, both MAP kinase isozymes, extracellular signal-regulated protein kinases 1 and 2, were strongly phosphorylated in benign and malignant thyroid cells. Treatment of the cells with 17beta-estradiol and MAP kinase kinase 1 inhibitor, PD 098059, prevented the accumulation of cyclin D1 and estrogen-mediated mitogenesis. Our data indicate that 17beta-estradiol is a potent mitogen for benign and malignant thyroid tumor cells and that it exerts a growth-promoting effect not only by binding to nuclear estrogen receptors, but also by activation of the MAP kinase pathway.


Subject(s)
Adenocarcinoma/pathology , Cell Division/drug effects , Estradiol/pharmacology , Thyroid Neoplasms/pathology , Adenocarcinoma/chemistry , Adenocarcinoma/metabolism , Blotting, Western , Cyclin D1/analysis , Estrogen Receptor alpha , Estrogen Receptor beta , Goiter/metabolism , Goiter/pathology , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Receptors, Estrogen/analysis , Reverse Transcriptase Polymerase Chain Reaction , Thyroid Gland/chemistry , Thyroid Neoplasms/chemistry , Thyroid Neoplasms/metabolism , Tumor Cells, Cultured
4.
Exp Clin Endocrinol Diabetes ; 106 Suppl 4: S6-9, 1998.
Article in English | MEDLINE | ID: mdl-9867188

ABSTRACT

In iodine deficiency areas, activating mutations in the TSH receptor and Gs-alpha gene are found in the majority of toxic thyroid adenomas and in some toxic goiter nodules. Since TSH receptor and Gs-alpha gene mutations are very rare in areas with high iodine supply, iodine deficiency has been suspected to favor the occurrence of these mutations by yet unknown pathways. However, TSH receptor and Gs-alpha gene mutations alone are not sufficient to cause toxic adenomas and nodules. There is compelling evidence that other secondary and cAMP-independent mechanisms, including enhanced expression of various growth factors, their corresponding receptors and of signaling proteins, may affect the mutated cell and thus promote cell proliferation and in turn generation of the tumor.


Subject(s)
Adenoma/etiology , GTP-Binding Proteins/genetics , Iodine/deficiency , Mutation , Receptors, Thyrotropin/genetics , Thyroid Neoplasms/etiology , Thyroid Nodule/etiology , Adenoma/genetics , Animals , Cell Transformation, Neoplastic , Goiter, Nodular/etiology , Goiter, Nodular/genetics , Growth Substances/metabolism , Humans , Oncogenes , Thyroid Neoplasms/genetics , Thyroid Nodule/genetics
5.
J Clin Endocrinol Metab ; 81(5): 1898-904, 1996 May.
Article in English | MEDLINE | ID: mdl-8626855

ABSTRACT

In toxic thyroid adenomas, mutations in the TSH receptor (TSH-R) gene or the gene encoding the alpha-subunit of the stimulatory guanine nucleotide-binding protein (Gs alpha) have been demonstrated to constitutively activate the cAMP cascade, which subsequently stimulates the growth and function of these tumors. However, the widely varying thyroid phenotypes in patients with TSH-R germline mutations, ranging from only slightly enlarged diffuse to multinodular goiters, suggest that additional mechanisms may be effective in the pathogenesis of toxic adenomas. We have investigated the levels of stimulatory and inhibitory G protein alpha-subunits together with basal and TSH-stimulated adenylate cyclase (AC) activity in toxic adenomas with or without activating mutations and in nodular and extranodular tissues of a toxic goiter due to a germline mutation in the TSH-R gene. Augmented expression of Gs alpha protein was detected in all toxic adenomas, independent of the presence of mutations, and in the nodular tissue of the toxic goiter, but not in the nonnodular hyperplastic tissue of the toxic goiter with the mutated TSH-R. Analogously, the expression of the alpha-subunit of the inhibitory G protein (Gi alpha) was also increased in all adenomas and the nodular tissue of the goiter, but, again, not in the hyperplastic goiter tissue. Basal AC activity was high in all tissues with mutations, but was only slightly increased in adenomas without detected mutations. No correlation was detectable between basal or TSH-stimulated AC activity and the levels of Gs alpha and Gi alpha. Our data suggest that mutational activation of the cAMP cascade may not be sufficient to generate toxic nodules and adenomas, but far more complex mechanisms, including alterations of G protein signaling, may be effective in the pathogenesis of these tumors.


Subject(s)
Adenoma/enzymology , Adenylyl Cyclases/metabolism , GTP-Binding Proteins/metabolism , Thyroid Neoplasms/enzymology , Base Sequence , DNA Mutational Analysis , Enzyme Activation/drug effects , GTP-Binding Proteins/genetics , Gene Expression , Humans , Molecular Sequence Data , Point Mutation , Receptors, Thyrotropin/genetics , Thyrotropin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...