Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 160
Filtrar
1.
Biochim Biophys Acta ; 1849(2): 171-80, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24939836

RESUMO

Expression of the vitamin D receptor (VDR) is widespread but may vary depending on the developmental stage of the animal, and therefore may differentially influence phenotypic function. Thus, the major role of the 1,25-dihydroxyvitamin D [1,25(OH)2D]/VDR system is to regulate mineral and skeletal homeostasis, although mainly after birth. Post-natally, under conditions of low dietary calcium, the 1,25(OH)2D/VDR system enhances intestinal transcellular transport of calcium and possibly paracellular calcium entry by regulating genes that are critical for these functions. This process, by providing adequate calcium, is essential for normal development of the skeletal growth plate and mineralization of bone. Furthermore, blood calcium and phosphorus homeostasis is maintained by an interplay between feedback loops of the 1,25(OH)2D/VDR system with parathyroid hormone and with fibroblast-growth factor (FGF) 23 respectively. The 1,25(OH)2D/VDR system can also modulate the expression of genes involved in both bone formation and resorption post-natally. Ligand independent activity of the VDR normally influences mammalian hair cycling after birth by potentiating Wnt and bone morphogenetic protein (BMP) signaling. Nevertheless ligand bound VDR may also modulate epidermal cell proliferation/differentiation by regulating the balance in function of c-MYC and its antagonist the transcriptional repressor MAD1/MXD1 in skin epithelia. The 1,25(OH)2D/VDR system can also modulate innate immune cells and promote a more tolerogenic immunological status and may therefore influence inflammation and the development of autoimmunity; whether this impacts the fetus is uncertain. This article is part of a Special Issue entitled: Nuclear receptors in animal development.


Assuntos
Crescimento e Desenvolvimento/genética , Receptores de Calcitriol/fisiologia , Vitamina D/análogos & derivados , Animais , Epiderme/crescimento & desenvolvimento , Epiderme/metabolismo , Fator de Crescimento de Fibroblastos 23 , Regulação da Expressão Gênica no Desenvolvimento , Crescimento e Desenvolvimento/efeitos dos fármacos , Humanos , Sistema Imunitário/fisiologia , Osteogênese/genética , Osteogênese/fisiologia , Transdução de Sinais/genética , Fenômenos Fisiológicos da Pele/genética , Vitamina D/metabolismo , Vitamina D/farmacologia , Vitamina D/fisiologia
2.
Horm Metab Res ; 43(3): 183-7, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21264795

RESUMO

Menin promotes the commitment of pluripotent mesenchymal stem cells to the osteoblast lineage by interacting with the BMP-2 signaling molecules Smad1/5, and Runx2. However, the relationship between menin and the Wnt-ß-catenin pathway in bone is unclear. Reduction of menin expression by transfection of a menin antisense construct did not alter the levels of ß-catenin in mouse mesenchymal C2C12 and osteoblastic MC3T3-E1 cells. However, menin co-immunoprecipitated with ß-catenin as well as LEF-1 in C2C12 and MC3T3-E1 cells. Reduction of menin expression by antisense menin transfection antagonized ß-catenin-induced transcriptional activity of the pGL3-OT luciferase reporter construct in C2C12 and MC3T3-E1 cells. Antisense menin transfection antagonized the BMP-2 and ß-catenin-stimulated increases in Runx2 and alkaline phosphatase levels in C2C12 cells. The data show that menin interacts with ß-catenin in mouse mesenchymal and osteoblastic cells, and suggest that the interaction is important for osteoblast differentiation.


Assuntos
Diferenciação Celular , Osteoblastos/citologia , Osteoblastos/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , beta Catenina/metabolismo , Animais , Proteína Morfogenética Óssea 2/metabolismo , Células Cultivadas , Células-Tronco Mesenquimais/metabolismo , Camundongos , Ligação Proteica , Proteínas Proto-Oncogênicas/genética , beta Catenina/genética
3.
Kidney Int ; 69(2): 218-23, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16408109

RESUMO

Recent studies in mice using genetic approaches have shed new light on the physiological effects of 1,25-dihydroxyvitamin D (1,25(OH)(2)D) and the vitamin D receptor (VDR) in skeletal and mineral homeostasis, and on their interaction with calcium. These studies in mice with targeted deletion of the 25-hydroxyvitamin D-1alpha-hydroxylase (1alpha(OH)ase), and of the VDR or of double mutants, have shown the discrete effects of calcium in inhibiting parathyroid hormone secretion and in enhancing bone mineralization, but overlapping effects of calcium and 1,25(OH)(2)D on inhibiting parathyroid growth and on normal development of the cartilaginous growth plate. The 1,25(OH)(2)D/VDR system is essential, however, in enhancing intestinal calcium absorption and in optimally increasing osteoclastic activation. In addition, the 1,25(OH)(2)D/VDR system has important anabolic effects on bone, thus defining a dual role for this system in bone turnover. These studies are revealing functions of the vitamin D/VDR system which have relevance for new concepts of the pathophysiology of renal bone disease and, in particular, of the adynamic bone disorder, and for the development of new analogs of the active form of vitamin D, which have less calcemic activity and greater skeletal anabolic effects.


Assuntos
Remodelação Óssea , Calcificação Fisiológica , Calcitriol/fisiologia , Receptores de Calcitriol/fisiologia , Animais , Calcitriol/uso terapêutico , Distúrbio Mineral e Ósseo na Doença Renal Crônica/etiologia , Humanos , Nefropatias/tratamento farmacológico , Nefropatias/metabolismo , Camundongos , Hormônio Paratireóideo/sangue , Diálise Renal
4.
Horm Metab Res ; 37(6): 375-9, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16001330

RESUMO

PITUITARY: Menin is a Smad3-interacting protein; inactivation of menin blocks transforming growth factor (TGF)-beta and activin signaling, antagonizing their growth-inhibitory properties in anterior pituitary cells. Menin is also required for the activin-induced inhibition of prolactin expression mediated by the Smads and the transcription factor, Pit-1. The interaction between menin and Smad3 is direct. PARATHYROID: In cultured parathyroid cells from uremic hemodialysis patients, in which the menin signaling pathways are probably still intact, menin inactivation achieved by menin antisense oligonucleotides leads to loss of TGF-beta inhibition of parathyroid cell proliferation and parathyroid hormone (PTH) secretion. Moreover, TGF-beta does not affect the proliferation and PTH production of parathyroid cells from multiple endocrine neoplasia type 1 (MEN1) patients. OSTEOBLAST: Men1-null mouse fetuses that die at day 12 or earlier have cranial/facial hypoplasias implicating menin in bone development. Menin is required for the commitment of multipotential mesenchymal stem cells into the osteoblast lineage. This is achieved by menin interacting physically and functionally with bone morphogenetic protein (BMP)-2 regulated Smads, such as Smad1 and Smad5, and the key osteoblast regulator, Runx2. These interactions are lost as the committed osteoblasts differentiate further at which time menin interacts with Smad3, mediating the negative regulation of Runx2 by TGF-beta. Menin also suppresses osteoblast maturation, partly by inhibiting the differentiation actions of JunD.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Osteoblastos/metabolismo , Hipófise/metabolismo , Transdução de Sinais , Glândula Tireoide/metabolismo , Transativadores/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Diferenciação Celular/genética , Células Cultivadas , Humanos , Camundongos , Camundongos Mutantes , Neoplasia Endócrina Múltipla Tipo 1/genética , Neoplasia Endócrina Múltipla Tipo 1/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Smad , Proteína Smad1 , Glândula Tireoide/citologia
6.
J Mol Endocrinol ; 27(3): 357-66, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11719288

RESUMO

The human growth hormone receptor (hGHR) is encoded by exons 2-10 of the hGHR gene on chromosome 5p13.1-p12. There are several different 5' untranslated region (5'UTR) variants of hGHR mRNA (V1-V9) that all encode the same protein. We have recently mapped the V1-V9 5'UTR sequences within 40 kb of the 5' flanking region of the hGHR gene. Seven of the exons are clustered within two small modules, module A (V2-V9-V3) and module B (V7-V1-V4-V8), approximately 38 kb and approximately 18 kb respectively upstream of exon 2 of the coding region; V6 lies midway between the two modules and V5 is adjacent to exon 2. We now report the existence of two subvariant V3 exons, one upstream of module A (exon V3b) and one midway between module B and exon 2 (exon V3a/b). Both have sequences homologous to Alu elements. In addition, we determined the alternative splicing mechanisms that produce three different mRNAs from these exons: V3c (from the V3 exon in module A) or V3a and V3b (from a combination of exon V3 and the Alu-containing V3 subvariant exons). hGHR expression is under developmental- and tissue-specific regulation: module A-derived mRNAs are widely expressed in human tissues, while module B-derived mRNAs are only detectable in postnatal liver. Expression of the variant V3 mRNAs is similar to those from module A, being produced ubiquitously in human fetal and postnatal tissues, with V3c always the major variant detected. The Alu-containing mRNAs (V3a and V3b) are also detectable in baboon and rhesus tissues, in accordance with the finding of Alu elements throughout the primate genome. In summary, we have mapped the relative locations of two new 5'UTR exons within the 5' flanking region of the hGHR gene and described the derivation and expression patterns for two variant hGHR mRNAs from these primate-specific exons. The introduction of Alu elements has contributed to the evolution of the primate GHR gene as a highly complex transcriptional unit.


Assuntos
Elementos Alu , Proteínas de Membrana/genética , Regiões 5' não Traduzidas , Adulto , Sequência de Bases , Mapeamento Cromossômico , Clonagem Molecular , DNA/genética , Éxons , Feto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Variação Genética , Humanos , Dados de Sequência Molecular , RNA Mensageiro/genética , Homologia de Sequência do Ácido Nucleico , Distribuição Tecidual
7.
Hum Mutat ; 18(5): 411-21, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11668634

RESUMO

We studied family members of a large kindred expressing both familial hypocalciuric hypercalcemia (FHH) and neonatal severe hyperparathyroidism (NSHPT) and found, by PCR amplification of the extracellular calcium-sensing receptor (CASR) gene exons and flanking intronic sequences, that FHH individuals were heterozygous for a g to t substitution in the last nucleotide of intron 2 (IVS2-1G>T). Defects in messenger RNA splicing were investigated by illegitimate transcription of the CASR gene in lymphoblastoid cells from an FHH affected individual, as well as by transfection of a CASR minigene harboring this mutation into HEK293 cells. The mutation resulted predominantly in exon III skipping causing a shift in exon IV reading frame and introduction of a premature stop codon leading to a predicted truncated protein of 153 amino acids. Interestingly, it was noted that exon III splicing is not 100% efficient in parathyroid, thyroid, and kidney; an exon III-deleted transcript is produced approximately 15% of the time. This is the first description of a splice site mutation in the CASR gene and provides an explanation of the clinical phenotype of the patients.


Assuntos
Processamento Alternativo/genética , Hipercalcemia/genética , Hiperparatireoidismo/genética , Mutação/genética , Sítios de Splice de RNA/genética , Receptores de Superfície Celular/genética , Sequência de Bases , Linhagem Celular , Códon sem Sentido/genética , Análise Mutacional de DNA , Éxons/genética , Feminino , Humanos , Recém-Nascido , Íntrons/genética , Masculino , Ensaios de Proteção de Nucleases , Fases de Leitura Aberta/genética , Linhagem , RNA Mensageiro/análise , RNA Mensageiro/genética , Receptores de Detecção de Cálcio , Receptores de Superfície Celular/química , Receptores de Superfície Celular/metabolismo , Deleção de Sequência/genética , Transcrição Gênica/genética , Transfecção
8.
J Clin Endocrinol Metab ; 86(10): 4627-9, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11600515

RESUMO

Pseudohypoparathyroid type 1b patients are characterized by renal resistance to PTH in the absence of Albright's hereditary osteodystrophy or other endocrine abnormalities. Kindred studies have suggested that the cause of this resistance is a specific decrease in Galphas activity in renal proximal tubules due to paternal imprinting of Galphas. To test this, allelic expression of Galphas was analyzed in human fetal kidney cortex samples by RT-PCR assays. The results showed that, in contrast to the parent-specific expression of exon 1A and XLalphas (paternal) or NESP (maternal) mRNAs, Galphas transcripts are biallelically expressed in human kidney cortex. These data implicate abnormal imprinting of alternative regions within the GNAS1 locus as a more likely cause of pseudohypoparathyroid type 1b.


Assuntos
Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Proteínas Heterotriméricas de Ligação ao GTP/genética , Córtex Renal/metabolismo , Pseudo-Hipoparatireoidismo/metabolismo , RNA Mensageiro/análise , Impressão Genômica , Humanos
9.
Mol Cell Probes ; 15(4): 239-42, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11513560

RESUMO

A polymorphic GT dinucleotide repeat sequence has been identified in the 5' flanking region of the human growth hormone receptor (hGHR) gene on chromosome 5p13.1-p12, within the promoter region of the V9 5'UTR exon. Thirteen alleles have been identified in 50 non-related individuals, with an observed heterozygosity of 52%. The major allele contains 24 repeats, although a range of 19-32 repeats has been observed. Codominant segregation was demonstrated in five two-generation and two three-generation families. This marker may be useful in analysing the role of the hGHR gene in pre- and postnatal growth disorders.


Assuntos
Regiões 5' não Traduzidas/genética , Repetições de Dinucleotídeos/genética , Proteínas de Membrana/genética , Polimorfismo Genético/genética , Alelos , Feminino , Frequência do Gene , Marcadores Genéticos/genética , Transtornos do Crescimento/genética , Heterozigoto , Humanos , Masculino
10.
J Biol Chem ; 276(44): 41229-36, 2001 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-11514554

RESUMO

SOX9 is a transcription factor that is essential for chondrocyte differentiation and cartilage formation. We stably overexpressed SOX9 cDNA in the rat chondrocytic cell line CFK2. Compared with the vector control, a greater proportion of SOX9-transfected cells accumulated in the G0/G1 phase. This was associated with an increase in mRNA and protein expression of p21(cip1), an inhibitor of cyclin-dependent kinase activity. SOX9 enhanced p21(cip1) promoter activity in a luciferase reporter assay. CFK2 cells overexpressing SOX9 became more elongated and adhesive and demonstrated a shift in cytoplasmic F-actin distribution. N-cadherin mRNA levels were elevated in the SOX9-transfected cells, and SOX9 enhanced N-cadherin promoter activity. By electrophoretic mobility shift assay, nuclear extracts of SOX9-transfected CFK2 cells specifically bound an oligonucleotide comprising an N-cadherin promoter region containing a consensus SOX9-binding motif. The transcriptional activity of SOX9 depended upon nuclear localization signals required for SOX9 nuclear entry. Differentiation of transfected CFK2 cells was accelerated as evidenced by more rapid accumulation of alkaline phosphatase activity, increased production of proteoglycans, and increased calcium accumulation, and this was associated with decreased ERK1 expression. These studies demonstrate that SOX9 alters the rate of cell cycle progression of chondrocytes and their differentiation by enhancing or inhibiting the expression of selected genes, including p21(cip1) and ERK1, and that N-cadherin is an additional direct target of this transcriptional regulator.


Assuntos
Ciclo Celular/fisiologia , Diferenciação Celular/fisiologia , Condrócitos/citologia , Regulação da Expressão Gênica/fisiologia , Proteínas de Grupo de Alta Mobilidade/fisiologia , Fatores de Transcrição/fisiologia , Transcrição Gênica/fisiologia , Animais , Sequência de Bases , Caderinas/genética , Linhagem Celular , Primers do DNA , Proteínas de Grupo de Alta Mobilidade/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Fatores de Transcrição SOX9 , Fatores de Transcrição/genética , Transfecção
11.
J Biol Chem ; 276(36): 33995-4005, 2001 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-11404353

RESUMO

During endochondral bone formation and fracture healing, cells committed to chondrogenesis undergo a temporally restricted program of differentiation that is characterized by sequential changes in their phenotype and gene expression. This results in the manufacture, remodeling, and mineralization of a cartilage template on which bone is laid down. Articular chondrocytes undergo a similar but restricted differentiation program that does not proceed to mineralization, except in pathologic conditions such as osteoarthritis. The pathogenesis of disorders of cartilage development and metabolism, including osteochondrodysplasia, fracture non-union, and osteoarthritis remain poorly defined. We used the CFK2 model to examine the potential roles of phosphate and calcium ions in the regulatory pathways that mediate chondrogenesis and cartilage maturation. Differentiation was monitored over a 4-week period using a combination of morphological, biochemical, and molecular markers that have been characterized in vivo and in vitro. CFK2 cells expressed the type III sodium-dependent phosphate transporters Glvr-1 and Ram-1, as well as a calcium-sensing mechanism. Regulated expression and activity of Glvr-1 by extracellular phosphate and parathyroid hormone-related protein was restricted to an early stage of CFK2 differentiation, as evidenced by expression of type II collagen, proteoglycan, and Ihh. On the other hand, regulated expression and activity of a calcium-sensing receptor by extracellular calcium was most evident after 2 weeks of differentiation, concomitant with an increase in type X collagen expression, alkaline phosphatase activity and parathyroid hormone/parathyroid hormone-related protein receptor expression. On the basis of these temporally restricted changes in the sensing and transport of phosphate and calcium, we predict that extracellular phosphate plays a role in the commitment of chondrogenic cells to differentiation, whereas extracellular calcium plays a role at a later stage in their differentiation program.


Assuntos
Cálcio/metabolismo , Condrócitos/citologia , Fosfatos/metabolismo , Fosfatase Alcalina/metabolismo , Animais , Southern Blotting , Desenvolvimento Ósseo , Diferenciação Celular , Linhagem Celular , Colágeno/metabolismo , Concentração de Íons de Hidrogênio , Immunoblotting , Rim/metabolismo , Glândulas Paratireoides/metabolismo , Proteína Relacionada ao Hormônio Paratireóideo , Fenótipo , Transporte Proteico , Proteínas/metabolismo , Proteoglicanas/metabolismo , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ribonucleases/metabolismo , Sódio/farmacologia , Fatores de Tempo
12.
Proc Natl Acad Sci U S A ; 98(13): 7498-503, 2001 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-11416220

RESUMO

The active form of vitamin D, 1alpha,25-dihydroxyvitamin D [1alpha,25(OH)2D], is synthesized from its precursor 25 hydroxyvitamin D [25(OH)D] via the catalytic action of the 25(OH)D-1alpha-hydroxylase [1alpha(OH)ase] enzyme. Many roles in cell growth and differentiation have been attributed to 1,25(OH)2D, including a central role in calcium homeostasis and skeletal metabolism. To investigate the in vivo functions of 1,25(OH)2D and the molecular basis of its actions, we developed a mouse model deficient in 1alpha(OH)ase by targeted ablation of the hormone-binding and heme-binding domains of the 1alpha(OH)ase gene. After weaning, mice developed hypocalcemia, secondary hyperparathyroidism, retarded growth, and the skeletal abnormalities characteristic of rickets. These abnormalities are similar to those described in humans with the genetic disorder vitamin D dependent rickets type I [VDDR-I; also known as pseudovitamin D-deficiency rickets (PDDR)]. Altered non-collagenous matrix protein expression and reduced numbers of osteoclasts were also observed in bone. Female mutant mice were infertile and exhibited uterine hypoplasia and absent corpora lutea. Furthermore, histologically enlarged lymph nodes in the vicinity of the thyroid gland and a reduction in CD4- and CD8-positive peripheral T lymphocytes were observed. Alopecia, reported in vitamin D receptor (VDR)-deficient mice and in humans with VDDR-II, was not seen. The findings establish a critical role for the 1alpha(OH)ase enzyme in mineral and skeletal homeostasis as well as in female reproduction and also point to an important role in regulating immune function.


Assuntos
25-Hidroxivitamina D3 1-alfa-Hidroxilase/deficiência , 25-Hidroxivitamina D3 1-alfa-Hidroxilase/genética , Transtornos do Crescimento/genética , Hiperparatireoidismo/genética , Infertilidade Feminina/genética , Raquitismo/genética , 25-Hidroxivitamina D3 1-alfa-Hidroxilase/metabolismo , Animais , Doenças Ósseas/genética , Calcifediol/sangue , Calcitriol/sangue , Cálcio/deficiência , Modelos Animais de Doenças , Feminino , Heterozigoto , Homozigoto , Humanos , Masculino , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Raquitismo/fisiopatologia
13.
Endocrinology ; 142(5): 1923-34, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11316758

RESUMO

Previous studies have identified eight variant human GH receptor (hGHR) messenger RNA (mRNAs; V1-V8), that differ in their 5'-untranslated regions (5'UTRs) but splice into the same site just upstream of the translation start site in exon 2; thus, they encode the same protein. Here we report a novel variant, V9, and describe the mapping of all nine 5'UTR sequences within 40 kb upstream of exon 2. A cluster of three sequences, V2-V9-V3 (termed module A), lies furthest 5', and approximately 16 kb downstream is a second cluster of four exons, V7-V1-V4-V8 (module B). V6 is midway between modules A and B. Module B is about 18 kb upstream of V5, which lies adjacent to exon 2. hGHR expression is under developmental- and tissue-specific regulation, and expression of the variant mRNAs is related to their position within the 5'-flanking region; whereas module A (V2,V9,V3) and V5 variants are widely expressed, module B (V7,V1,V4,V8) and V6 variant mRNAs are detectable only in postnatal liver. Transcriptional start sites for V1 and V9 (representing the two different modules) were identified, showing that postnatal liver-specific expression of V1 is driven from two TATA boxes, whereas the ubiquitous V9 transcript has a single start site and a TATA-less promoter. V9 promoter activity was shown by in vivo and in vitro transfection assays, and an NF-Y binding site was demonstrated by electromobility shift assay. Thus, the regulatory regions of the hGHR gene are complex, and the clustering of seven 5'UTR exons within two modules with distinctly different mRNA expression patterns is the most striking feature.


Assuntos
Regiões 5' não Traduzidas , Receptores da Somatotropina/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Sequência de Bases , Evolução Biológica , Criança , Pré-Escolar , Clonagem Molecular , Humanos , Lactente , Pessoa de Meia-Idade , Dados de Sequência Molecular , Especificidade de Órgãos , Regiões Promotoras Genéticas , RNA Mensageiro/análise , TATA Box , Transcrição Gênica
14.
Am J Physiol Renal Physiol ; 280(5): F868-78, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11292630

RESUMO

The distal convoluted tubule plays a significant role in renal magnesium conservation. Although the cells of the distal convoluted tubule possess the vitamin D receptor, little is known about the effects of 1alpha,25-dihydroxyvitamin D [1,25(OH)(2)D(3)] on magnesium transport. In this study, we examined the effect of 1,25(OH)(2)D(3) on distal cellular magnesium uptake and the modulation of this response by extracellular Ca2+ and Mg2+ in an immortalized mouse distal convoluted tubule (MDCT) cell line. MDCT cells possess the divalent cation-sensing receptor (CaSR) that responds to elevation of extracellular Ca2+ and Mg2+ concentrations to diminish peptide hormone-stimulated Mg2+ uptake. Mg2+ uptake rates were determined by microfluorescence in Mg2+ -depleted MDCT cells. Treatment of MDCT cells with 1,25(OH)(2)D(3) for 16-24 h stimulated basal Mg2+ uptake in a concentration-dependent manner from basal levels of 164 +/- 5 to 210 +/- 11 nM/s, representing a 28 +/- 3% change. Pretreatment with actinomycin D or cycloheximide abolished 1,25(OH)(2)D(3)-stimulated(.)Mg2+ uptake (154 +/- 18 nM/s), suggesting that 1,25(OH)(2)D(3) stimulates Mg2+ uptake through gene activation and protein synthesis. Elevation of extracellular Ca2+ inhibited 1,25(OH)(2)D(3)-stimulated Mg2+ uptake (143 +/- 5 nM/s). Preincubation of the cells with an antibody to the CaSR prevented the inhibition by elevated extracellular Ca2+ of 1,25(OH)(2)D(3)-stimulated Mg2+ uptake (202 +/- 8 nM/s). Treatment with an antisense CaSR mRNA oligodeoxynucleotide also abolished the effects of extracellular Ca2+ on 1,25(OH)(2)D(3)-responsive Mg2+ entry. This showed that elevated extracellular calcium modulates 1,25(OH)(2)D-mediated responses through the CaSR. In summary, 1,25(OH)(2)D(3) stimulated Mg2+ uptake in MDCT cells, and this is dependent on de novo protein synthesis. Elevation of extracellular Ca2+, acting via the CaSR, inhibited 1,25(OH)(2)D(3)-stimulated Mg2+ entry. These data indicate that 1,25(OH)(2)D(3) has important effects on the control of magnesium entry in MDCT cells and these responses can be modulated by extracellular divalent cations.


Assuntos
Calcitriol/farmacologia , Cálcio/fisiologia , Túbulos Renais Distais/metabolismo , Magnésio/metabolismo , Animais , Anticorpos Bloqueadores/farmacologia , Western Blotting , Bovinos , Linhagem Celular , AMP Cíclico/metabolismo , Citoplasma/metabolismo , Túbulos Renais Distais/efeitos dos fármacos , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Retículo Sarcoplasmático/efeitos dos fármacos , Retículo Sarcoplasmático/metabolismo , Estimulação Química
15.
Proc Natl Acad Sci U S A ; 98(7): 3837-42, 2001 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-11274402

RESUMO

Multiple endocrine neoplasia type 1 (MEN1) is an autosomal dominant disorder characterized by endocrine tumors of parathyroids, pancreatic islets, and anterior pituitary. The MEN1 gene encodes a nuclear protein called menin. In MEN1 carriers inactivating mutations give rise to a truncated product consistent with menin acting as a tumor suppressor gene. However, the role of menin in tumorigenesis and its physiological functions are not known. Here, we show that menin inactivation by antisense RNA antagonizes transforming growth factor type beta-mediated cell growth inhibition. Menin interacts with Smad3, and antisense menin suppresses transforming growth factor type beta-induced and Smad3-induced transcriptional activity by inhibiting Smad3/4-DNA binding at specific transcriptional regulatory sites. These results implicate a mechanism of tumorigenesis by menin inactivation.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas Proto-Oncogênicas , RNA Antissenso/farmacologia , Transativadores/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Animais , Células CHO , Células COS , Divisão Celular/efeitos dos fármacos , Cricetinae , DNA/efeitos dos fármacos , DNA/metabolismo , DNA Complementar/biossíntese , Proteínas de Ligação a DNA/fisiologia , Humanos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Adeno-Hipófise/citologia , Adeno-Hipófise/fisiologia , Ratos , Transdução de Sinais , Proteína Smad3 , Transativadores/fisiologia , Ativação Transcricional/efeitos dos fármacos , Células Tumorais Cultivadas
16.
J Clin Endocrinol Metab ; 86(3): 1394-7, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11238537

RESUMO

Hypocalcemia and hyperphosphatemia caused by PTH resistance are the only discernible abnormalities in pseudohypoparathyroidism type 1b (PHP-1b). Because of the selective resistance toward PTH, inactivating mutations in its receptor, the PTH/PTH-related peptide receptor (PTHR1), were thought to be responsible for PHP-1b. However, gene abnormalities responsible for PHP-1b have not been identified in the coding region and well conserved promoters (P1 and P2) of the PTHR1 gene. The purpose of the present study was to analyze the structure of the P3 promoter, the main promoter of the human PTHR1 gene in kidney, in patients with PHP-1b. Southern analysis of genomic DNA from lymphoblastoid cell lines of eight nonfamilial patients with PHP-1b revealed neither gross rearrangements nor methylation abnormalities in the P3 promoter region of the PTHR1 gene. Sequencing revealed no abnormalities in the P3 promoter region, although one patient was homozygous for an (AAAG)n polymorphic variant. In conclusion, despite the selective resistance toward PTH in the kidney, which mainly uses the PTHR1 P3 promoter, PHP-1b in eight cases is not associated with structural abnormalities in this promoter. This study also indicates that inactivation of the P3 promoter is not achieved by methylation as tested in patients' genomic DNA from lymphoblastoid cell lines. The influence of alterations in the polymorphic A-rich repeat sequence on promoter activity warrants further study.


Assuntos
DNA/análise , Regiões Promotoras Genéticas/genética , Pseudo-Hipoparatireoidismo/genética , Receptores de Hormônios Paratireóideos/genética , Adolescente , Sequência de Bases , Southern Blotting , Criança , Feminino , Homozigoto , Humanos , Japão , Masculino , Dados de Sequência Molecular , Reação em Cadeia da Polimerase , Polimorfismo Genético , Polimorfismo Conformacional de Fita Simples , Splicing de RNA , Receptor Tipo 1 de Hormônio Paratireóideo , Mapeamento por Restrição , Análise de Sequência de DNA
17.
Mol Genet Metab ; 72(2): 168-74, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11161843

RESUMO

Serum calcium is under tight physiological control, but it is also a quantitative trait with substantial genetic regulation. Mutations of the CASR gene cause familial hypocalciuric hypercalcemia or autosomal dominant hypoparathyroidism, depending on whether they decrease or increase, respectively, ligand binding to the receptor protein. We described an association between ionized calcium and a common polymorphism (A986S) found in the cytoplasmic tail of this G protein-coupled receptor. We report here on an independent study of 387 healthy young women. Genotyping was performed by allele-specific amplification and serum chemistries were measured by automated clinical assay. Frequencies of SS, AS, and AA genotypes were 6, 107, and 274, respectively, yielding a 986S allele frequency of 15.4%. Mean total serum calcium (Ca(T)) was significantly higher in the SS (9.88 +/- 0.29 mg/dL, P = 0.015) and AS groups (9.45 +/- 0.05 mg/dL, P = 0.002), than in the AA group (9.23 +/- 0.04 mg/dL). In multiple regression modeling, the A986S genotype remained an independently significant predictor of Ca(T) (P < 0.0001) when serum albumin, globulin, inorganic phosphate, and creatinine covariates were included. These data are the first to show significant association between a common polymorphism and concentrations of a serum electrolyte. The A986S polymorphism is also a potential predisposing factor in disorders of bone and mineral metabolism.


Assuntos
Cálcio/sangue , Polimorfismo Genético , Receptores de Superfície Celular/genética , Adolescente , Adulto , Fatores Etários , Alelos , Estatura , Peso Corporal , Osso e Ossos/metabolismo , Citoplasma/metabolismo , Feminino , Genótipo , Humanos , Análise Multivariada , Receptores de Detecção de Cálcio , Análise de Regressão
18.
J Bone Miner Res ; 16(1): 46-56, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11149489

RESUMO

The murine homologue of the 25-hydroxyvitamin D [25(OH)D] 1alpha-hydroxylase gene [1alpha(OH)ase; Cyp27bl], which is mutated in humans with vitamin D-dependent rickets type I (VDDR-I; also known as pseudovitamin D-deficiency rickets [PDDR]) was cloned and characterized. Like the human, the mouse gene has nine exons, and the exon-intron organization is well conserved. By interspecific backcross analysis, the Cyp27bl gene was mapped to 70.5 cM on mouse Chr 10. This is in a region syntenic with human Chr 12q13.1-q13.3 to which the human 1alpha(OH)ase gene was previously mapped. Kidney expression of the 1alpha(OH)ase was localized to cortical tubules and was higher in the adult mouse than in the fetus, consistent with the increased role of its product as a circulating hormone postnatally. Prenatally, the 1alpha(OH)ase gene, together with the vitamin D receptor (VDR) gene, was expressed in embryonic stem cells, and expression of 1alpha(OH)ase in bone and intestine was higher in the fetus than in the adult. These observations suggest that 1,25-dihydroxyvitamin D [1,25(OH)2D] plays a role in fetal development. In view of the fact that humans lacking 1alpha(OH)ase have apparently normal prenatal development, this may point to functional redundancy in the fetal vitamin D system, which now can be explored further in mouse models in which the 1alpha(OH)ase gene has been deleted.


Assuntos
25-Hidroxivitamina D3 1-alfa-Hidroxilase/genética , Éxons/genética , Regulação da Expressão Gênica no Desenvolvimento , Mapeamento Físico do Cromossomo , 25-Hidroxivitamina D3 1-alfa-Hidroxilase/química , Envelhecimento/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Osso e Ossos/embriologia , Osso e Ossos/metabolismo , Linhagem Celular , Clonagem Molecular , Sequência Conservada/genética , Feto/metabolismo , Perfilação da Expressão Gênica , Humanos , Hibridização In Situ , Mucosa Intestinal/metabolismo , Intestinos/embriologia , Íntrons/genética , Rim/embriologia , Rim/metabolismo , Camundongos , Camundongos Endogâmicos , Dados de Sequência Molecular , Oligonucleotídeos Antissenso , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de Calcitriol/genética , Receptores de Calcitriol/metabolismo , Mapeamento por Restrição , Células-Tronco/metabolismo
19.
J Biol Chem ; 276(6): 4070-9, 2001 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-11071898

RESUMO

Liver cells respond to changes in Ca(2+)(o). The hepatic functions affected include bile secretion, metabolic activity, liver regeneration, and the response to xenobiotics. In the present study, we demonstrate the presence, in the liver, of the extracellular calcium-sensing receptor (CASR), described previously in the parathyroid and thyroid glands and kidney. CASR mRNA was specifically expressed in hepatocytes and was absent in nonparenchymal liver cells (stellate, endothelial, and Kupffer cells). Western blot analysis using a specific CASR antibody showed staining in both whole liver and hepatocyte extracts. Immunohistochemistry and in situ hybridization of rat liver sections showed expression of CASR protein and mRNA by a subset of hepatocytes. The known agonists of the CASR, gadolinium (Gd(3+); 0.5-3.0 mm) and spermine (1.25-20 mm), in the absence of Ca(2+)(o), elicited dose-related increases in Ca(2+)(i) in isolated rat hepatocytes loaded with Fura-2/acetoxymethyl ester. There was a greatly attenuated response to a second challenge with either agonist. The response was also abrogated when inositol 1,4,5-trisphosphate (IP(3))-sensitive calcium pools had been depleted by pretreatment with either thapsigargin or phenylephrine, an alpha(1)-adrenergic receptor agonist known to mobilize Ca(2+)(i) from IP(3)-sensitive pools. Addition of the deschloro-phenylalkylamine compound, NPS R-467, but not the S enantiomer, NPS S-467, increased the sensitivity of the Ca(2+)(i) mobilization response to 1.25 mm spermine. Bile flow ceased after Ca(2+)(o) withdrawal, and its recovery was enhanced by spermine in isolated perfused liver preparations. The CASR agonists Ca(2+) and Gd(3+) increased bile flow, and the response to a submaximal Ca(2+) concentration was enhanced by NPS R-467 but not the S compound. Thus, the data indicate that rat hepatocytes harbor a CASR capable of mobilizing Ca(2+)(i) from IP(3)-sensitive stores and that activation of the CASR stimulates bile flow.


Assuntos
Bile/metabolismo , Cálcio/metabolismo , Hepatócitos/metabolismo , Receptores de Superfície Celular/metabolismo , Compostos de Anilina/farmacologia , Animais , Sequência de Bases , Linhagem Celular , Primers do DNA , Espaço Extracelular/metabolismo , Feminino , Hepatócitos/efeitos dos fármacos , Hibridização In Situ , Inositol 1,4,5-Trifosfato/metabolismo , Camundongos , RNA Mensageiro/genética , Ratos , Ratos Sprague-Dawley , Receptores de Detecção de Cálcio , Receptores de Superfície Celular/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Espermina/farmacologia
20.
Hum Mutat ; 16(4): 281-96, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11013439

RESUMO

The calcium-sensing receptor (CASR) is a plasma membrane G protein coupled receptor that is expressed in the parathyroid hormone (PTH) producing chief cells of the parathyroid gland and the cells lining the kidney tubule. By virtue of its ability to sense small changes in circulating calcium concentration ([Ca(2+)](o)) and to couple this information to intracellular signaling pathways that modify PTH secretion or renal cation handling, the CASR plays an essential role in maintaining mineral ion homeostasis. Inherited abnormalities of the CASR gene located on chromosome 3p13.3-21 can cause either hypercalcemia or hypocalcemia depending upon whether they are inactivating or activating, respectively. Heterozygous loss-of-function mutations give rise to familial (benign) hypocalciuric hypercalcemia (FHH) in which the lifelong hypercalcemia is asymptomatic. The homozygous condition manifests itself as neonatal severe hyperparathyroidism (NSHPT), a rare disorder characterized by extreme hypercalcemia and the bony changes of hyperparathyroidism which occur in infancy. The disorder autosomal dominant hypocalcemia (ADH) is due to gain-of-function mutations in the CASR gene. ADH may be asymptomatic or present with neonatal or childhood seizures. A common polymorphism in the intracellular tail of the CASR, Ala to Ser at position 986, has a modest effect on the serum calcium concentration in healthy individuals.


Assuntos
Cálcio/sangue , Cálcio/urina , Hipercalcemia/genética , Hiperparatireoidismo/genética , Hipocalcemia/genética , Mutação/genética , Receptores de Superfície Celular/genética , Humanos , Hipercalcemia/sangue , Hipercalcemia/urina , Hiperparatireoidismo/sangue , Hiperparatireoidismo/urina , Hipocalcemia/sangue , Hipocalcemia/urina , Recém-Nascido , Receptores de Detecção de Cálcio , Receptores de Superfície Celular/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...