Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 117
Filtrar
1.
Dev Biol ; 333(2): 348-58, 2009 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-19607824

RESUMO

Lbh is thought to act as a transcriptional cofactor and is highly conserved among species. Here we show that Lbh is expressed in chondrocytes, cells of the perichondrium, and the primary spongiosa in fetal growth plates of mice and chickens. Lbh overexpression in chick wings, using the RCAS-retroviral vector strategy, results in shortened skeletal elements and delayed hypertrophic chondrocyte maturation and bone formation. Additionally, osteoclast and endothelial cell invasion are delayed in the Lbh-overexpressing bones. Finally, we find a dramatic suppression of Runx2 and VEGF mRNAs in chondrocytes and osteoblasts that overexpress Lbh. Strikingly, this abnormal bone development in infected limbs can be rescued by concurrent overexpression of Runx2. These results suggest that during endochondral bone formation, Lbh may negatively regulate vascular invasion and formation of the early ossification center at least in part by interfering with Runx2 and/or VEGF expression.


Assuntos
Osso e Ossos/metabolismo , Condrócitos/citologia , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Neovascularização Patológica , Proteínas Nucleares/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Células 3T3 , Animais , Desenvolvimento Ósseo , Proteínas de Ciclo Celular , Embrião de Galinha , Condrócitos/metabolismo , Células Endoteliais/citologia , Hibridização In Situ , Camundongos , Modelos Biológicos , Proteínas Nucleares/genética , Osteoclastos/metabolismo , Fatores de Transcrição
2.
Nature ; 425(6960): 841-6, 2003 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-14574413

RESUMO

Stem cell fate is influenced by specialized microenvironments that remain poorly defined in mammals. To explore the possibility that haematopoietic stem cells derive regulatory information from bone, accounting for the localization of haematopoiesis in bone marrow, we assessed mice that were genetically altered to produce osteoblast-specific, activated PTH/PTHrP receptors (PPRs). Here we show that PPR-stimulated osteoblastic cells that are increased in number produce high levels of the Notch ligand jagged 1 and support an increase in the number of haematopoietic stem cells with evidence of Notch1 activation in vivo. Furthermore, ligand-dependent activation of PPR with parathyroid hormone (PTH) increased the number of osteoblasts in stromal cultures, and augmented ex vivo primitive haematopoietic cell growth that was abrogated by gamma-secretase inhibition of Notch activation. An increase in the number of stem cells was observed in wild-type animals after PTH injection, and survival after bone marrow transplantation was markedly improved. Therefore, osteoblastic cells are a regulatory component of the haematopoietic stem cell niche in vivo that influences stem cell function through Notch activation. Niche constituent cells or signalling pathways provide pharmacological targets with therapeutic potential for stem-cell-based therapies.


Assuntos
Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Osteoblastos/citologia , Osteoblastos/metabolismo , Transdução de Sinais , Animais , Osso e Ossos/citologia , Osso e Ossos/metabolismo , Proteínas de Ligação ao Cálcio , Contagem de Células , Divisão Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Meio Ambiente , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intercelular , Proteína Jagged-1 , Ligantes , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , Osteoblastos/efeitos dos fármacos , Hormônio Paratireóideo/farmacologia , Proteínas/metabolismo , Ratos , Receptor Tipo 1 de Hormônio Paratireóideo/metabolismo , Receptores Notch , Receptores de Hormônios Paratireóideos/metabolismo , Proteínas Serrate-Jagged , Transdução de Sinais/efeitos dos fármacos , Células Estromais/citologia , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo
3.
J Cell Biol ; 96(4): 1117-9, 1983 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-6300144

RESUMO

Eucaryotic endoplasmic reticulum and bacterial inner cell membranes use strikingly similar mechanisms to transport proteins from one cellular compartment to another. Using a mammalian linked transcription-translation system, we show here that canine pancreatic microsomes accurately cleave off the signal sequence of beta-lactamase, a secreted bacterial protein. We suggest that the apparent differences between bacterial and eukaryotic protein transport may involve only minor modulations of a profoundly conserved mechanism.


Assuntos
Proteínas de Bactérias/metabolismo , Precursores Enzimáticos/metabolismo , Microssomos/metabolismo , beta-Lactamases/metabolismo , Animais , Cães , Precursores Enzimáticos/genética , Vetores Genéticos , Pâncreas/ultraestrutura , Plasmídeos , Coelhos , Reticulócitos/metabolismo , beta-Lactamases/genética
4.
Science ; 273(5275): 613-22, 1996 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-8662546

RESUMO

Proper regulation of chondrocyte differentiation is necessary for the morphogenesis of skeletal elements, yet little is known about the molecular regulation of this process. A chicken homolog of Indian hedgehog (Ihh), a member of the conserved Hedgehog family of secreted proteins that is expressed during bone formation, has now been isolated. Ihh has biological properties similar to those of Sonic hedgehog (Shh), including the ability to regulate the conserved targets Patched (Ptc) and Gli. Ihh is expressed in the prehypertrophic chondrocytes of cartilage elements, where it regulates the rate of hypertrophic differentiation. Misexpression of Ihh prevents proliferating chondrocytes from initiating the hypertrophic differentiation process. The direct target of Ihh signaling is the perichondrium, where Gli and Ptc flank the expression domain of Ihh. Ihh induces the expression of a second signal, parathyroid hormone-related protein (PTHrP), in the periarticular perichondrium. Analysis of PTHrP (-/-) mutant mice indicated that the PTHrP protein signals to its receptor in the prehypertrophic chondrocytes, thereby blocking hypertrophic differentiation. In vitro application of Hedgehog or PTHrP protein to normal or PTHrP (-/-) limb explants demonstrated that PTHrP mediates the effects of Ihh through the formation of a negative feedback loop that modulates the rate of chondrocyte differentiation.


Assuntos
Desenvolvimento Ósseo , Cartilagem/citologia , Lâmina de Crescimento/citologia , Osteogênese , Proteínas/fisiologia , Transativadores , Animais , Sequência de Bases , Cartilagem/metabolismo , Diferenciação Celular , Divisão Celular , Embrião de Galinha , Clonagem Molecular , Técnicas de Cultura , Extremidades/embriologia , Retroalimentação , Regulação da Expressão Gênica , Lâmina de Crescimento/metabolismo , Proteínas Hedgehog , Camundongos , Dados de Sequência Molecular , Morfogênese , Hormônio Paratireóideo , Proteína Relacionada ao Hormônio Paratireóideo , Fenótipo , Proteínas/farmacologia , Receptor Tipo 1 de Hormônio Paratireóideo , Receptores de Hormônios Paratireóideos/fisiologia , Transdução de Sinais
5.
Science ; 254(5034): 1024-6, 1991 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-1658941

RESUMO

The complementary DNA encoding a 585-amino acid parathyroid hormone-parathyroid hormone-related peptide (PTH-PTHrP) receptor with seven potential membrane-spanning domains was cloned by COS-7 expression using an opossum kidney cell complementary DNA (cDNA) library. The expressed receptor binds PTH and PTHrP with equal affinity, and both ligands equivalently stimulate adenylate cyclase. Striking homology with the calcitonin receptor and lack of homology with other G protein-linked receptors indicate that receptors for these calcium-regulating hormones are related and represent a new family.


Assuntos
Receptores de Superfície Celular/genética , Sequência de Aminoácidos , Animais , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Dados de Sequência Molecular , Gambás , Hormônio Paratireóideo/metabolismo , Proteína Relacionada ao Hormônio Paratireóideo , Proteínas/metabolismo , Receptores de Superfície Celular/química , Receptores de Superfície Celular/metabolismo , Receptores de Hormônios Paratireóideos , Alinhamento de Sequência , Solubilidade
6.
Science ; 273(5275): 663-6, 1996 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-8662561

RESUMO

The PTH/PTHrP receptor binds to two ligands with distinct functions: the calcium-regulating hormone, parathyroid hormone (PTH), and the paracrine factor, PTH-related protein (PTHrP). Each ligand, in turn, is likely to activate more than one receptor. The functions of the PTH/PTHrP receptor were investigated by deletion of the murine gene by homologous recombination. Most PTH/PTHrP receptor (-/-) mutant mice died in mid-gestation, a phenotype not observed in PTHrP (-/-) mice, perhaps because of the effects of maternal PTHrP. Mice that survived exhibited accelerated differentiation of chondrocytes in bone, and their bones, grown in explant culture, were resistant to the effects of PTHrP and Sonic hedgehog. These results suggest that the PTH/PTHrP receptor mediates the effects of Indian Hedgehog and PTHrP on chondrocyte differentiation.


Assuntos
Desenvolvimento Ósseo , Cartilagem/citologia , Lâmina de Crescimento/citologia , Osteogênese , Receptores de Hormônios Paratireóideos/fisiologia , Transativadores , Animais , Cartilagem/metabolismo , Diferenciação Celular , Divisão Celular , Clonagem Molecular , Técnicas de Cultura , Retroalimentação , Deleção de Genes , Marcação de Genes , Lâmina de Crescimento/metabolismo , Proteínas Hedgehog , Camundongos , Camundongos Endogâmicos C57BL , Osteoblastos/citologia , Hormônio Paratireóideo , Proteína Relacionada ao Hormônio Paratireóideo , Biossíntese de Proteínas , Proteínas/farmacologia , Proteínas/fisiologia , Receptor Tipo 1 de Hormônio Paratireóideo , Receptores de Hormônios Paratireóideos/genética , Células-Tronco
7.
J Dent Res ; 97(11): 1260-1267, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29746183

RESUMO

Craniofacial development requires a set of patterning codes that define the identities of postmigratory mesenchymal cells in a region-specific manner, in which locally expressed morphogens, including fibroblast growth factors (FGFs) and bone morphogenetic proteins (BMPs), provide instructive cues. Msx2, a bona fide target of BMP signaling, is a transcription factor regulating Runx2 and osterix (Osx), whose mutations are associated with cranial deformities in humans. Here we show that Msx2 defines osteo-chondro precursor cells in specific regions of the craniofacial mesenchyme at the postmigratory stage, particularly in the mandibular process and the posterior cranial vault. Analysis of Msx2-creER mice revealed that early mesenchymal cells in proximity to the BMP4-expressing mesenchyme were marked upon tamoxifen injection, and their descendants contributed to diverse types of mesenchymal cells in the later stage, such as chondrocytes and perichondrial cells of the transient cartilage, as well as osteoblasts and suture mesenchymal cells. By contrast, Osx-creER marked osteoblast precursors at the later stage, and their descendants continued to become osteoblasts well into the postnatal stage. Therefore, Msx2 marks spatially restricted populations of mesenchymal precursor cells with diverse differentiation potential, suggesting that extrinsic molecular cues can dictate the nature of postmigratory mesenchymal cells in craniofacial development.


Assuntos
Proteínas de Homeodomínio/fisiologia , Mandíbula/crescimento & desenvolvimento , Células-Tronco Mesenquimais/fisiologia , Crânio/crescimento & desenvolvimento , Animais , Cartilagem/embriologia , Cartilagem/crescimento & desenvolvimento , Diferenciação Celular , Feminino , Proteínas de Homeodomínio/metabolismo , Hibridização In Situ , Masculino , Mandíbula/embriologia , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Transgênicos , Osteoblastos/metabolismo , Crânio/embriologia
8.
J Clin Invest ; 104(4): 399-407, 1999 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10449432

RESUMO

Parathyroid hormone (PTH) and parathyroid hormone-related peptide (PTHrP) bind to and activate the same PTH/PTHrP receptor. Deletion of either the PTHrP gene or the PTH/PTHrP receptor gene leads to acceleration of differentiation of growth plate chondrocytes. To explore further the functional relationships of PTHrP and the PTH/PTHrP receptor, bones of knockout mice were analyzed early in development, and the phenotypes of double-knockout mice were characterized. One early phenotype is shared by both knockouts. Normally, the first chondrocytes to become hypertrophic are located in the centers of long bones; this polarity is greatly diminished in both these knockouts. The PTH/PTHrP receptor-deficient (PTH/PTHrP-R(-/-)) mice exhibited 2 unique phenotypes not shared by the PTHrP(-/-) mice. During intramembranous bone formation in the shafts of long bones, only the PTH/PTHrP-R(-/-) bones exhibit a striking increase in osteoblast number and matrix accumulation. Furthermore, the PTH/PTHrP-R(-/-) mice showed a dramatic decrease in trabecular bone formation in the primary spongiosa and a delay in vascular invasion of the early cartilage model. In the double-homozygous knockout mice, the delay in vascular invasion did not occur. Thus, PTHrP must slow vascular invasion by a mechanism independent of the PTH/PTHrP receptor.


Assuntos
Desenvolvimento Ósseo/genética , Desenvolvimento Ósseo/fisiologia , Osso e Ossos/anormalidades , Proteínas/genética , Proteínas/fisiologia , Receptores de Hormônios Paratireóideos/genética , Receptores de Hormônios Paratireóideos/fisiologia , Animais , Osso e Ossos/irrigação sanguínea , Feminino , Masculino , Camundongos , Camundongos Knockout , Osteoblastos/patologia , Proteína Relacionada ao Hormônio Paratireóideo , Fenótipo , Gravidez , Receptor Tipo 1 de Hormônio Paratireóideo
9.
J Clin Invest ; 107(3): 295-304, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11160153

RESUMO

Vertebrate skeletogenesis requires a well-coordinated transition from chondrogenesis to osteogenesis. Hypertrophic chondrocytes in the growth plate play a pivotal role in this transition. Parathyroid hormone-related peptide (PTHrP), synthesized in the periarticular growth plate, regulates the site at which hypertrophy occurs. By comparing PTH/PTHrP receptor(-/-)/wild-type (PPR(-/-)/wild-type) chimeric mice with IHH(-/-);PPR(-/-)/wild-type chimeric and IHH(-/-)/wild-type chimeric mice, we provide in vivo evidence that Indian hedgehog (IHH), synthesized by prehypertrophic and hypertrophic chondrocytes, regulates the site of hypertrophic differentiation by signaling to the periarticular growth plate and also determines the site of bone collar formation in the adjacent perichondrium. By providing crucial local signals from prehypertrophic and hypertrophic chondrocytes to both chondrocytes and preosteoblasts, IHH couples chondrogenesis to osteogenesis in endochondral bone development.


Assuntos
Desenvolvimento Ósseo/fisiologia , Proteínas/fisiologia , Transativadores , Animais , Cartilagem/embriologia , Cartilagem/crescimento & desenvolvimento , Diferenciação Celular , Condrócitos/metabolismo , Condrócitos/fisiologia , Condrogênese , Indução Embrionária , Lâmina de Crescimento/embriologia , Lâmina de Crescimento/crescimento & desenvolvimento , Proteínas Hedgehog , Hibridização In Situ , Camundongos , Osteogênese , Proteína Relacionada ao Hormônio Paratireóideo , Biossíntese de Proteínas , Proteínas/genética , Proteínas/metabolismo , RNA Mensageiro/análise , Transdução de Sinais , Tíbia/embriologia , Tíbia/crescimento & desenvolvimento
10.
J Clin Invest ; 86(4): 1084-7, 1990 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-2212001

RESUMO

Preproparathyroid hormone (preproPTH) gene mutation has been proposed as a cause of familial isolated hypoparathyroidism (FIH). We cloned the preproPTH alleles of a patient with autosomal dominant FIH and sequenced the coding regions, 5' flanking regions, and splice junctions. The putatively abnormal (based on previous linkage studies) allele differed from the other allele's normal sequence at only one nucleotide. This T to C point mutation changes the codon for position 18 of the 31 amino acid prepro sequence from cysteine to arginine, disrupting the hydrophobic core of the signal sequence. Because the hydrophobic core is required by secreted proteins for efficient translocation across the endoplasmic reticulum, the mutant protein is likely to be inefficiently processed. Indeed, in vitro studies demonstrated dramatically impaired processing of the mutant preproPTH to proPTH. In summary, we observed a point mutation in the signal peptide-encoding region of a preproPTH gene in one FIH kindred and demonstrated a functional defect caused by the mutation. Mutation of the signal sequence constitutes a novel pathophysiologic mechanism in man, and further study may yield important insights both into this form of hormone deficiency and into the role of signal sequences in human physiology.


Assuntos
Hipoparatireoidismo/genética , Mutação , Hormônio Paratireóideo/genética , Precursores de Proteínas/genética , Sinais Direcionadores de Proteínas/genética , Sequência de Aminoácidos , Sequência de Bases , Feminino , Humanos , Masculino , Dados de Sequência Molecular
11.
J Clin Invest ; 107(8): 1007-15, 2001 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-11306604

RESUMO

We used Hoxa3 knockout mice and other mouse models to study the role of the fetal parathyroids in fetal calcium homeostasis. Hoxa3-null fetuses lack parathyroid glands, and absence of parathyroid hormone (PTH) was confirmed with a rodent PTH immunoradiometric assay. The ionized calcium level of Hoxa3-null fetuses was significantly lower than that of wild-type or heterozygous littermates or of the mother. Both the rate of placental calcium transfer and the plasma PTHrP level were normal in Hoxa3 mutants and their heterozygous siblings. Because we had previously observed an increase in placental calcium transfer in PTH/PTHrP receptor 1-null (Pthr1-null) fetuses, we assayed plasma PTHrP in those mice. Pthr1-null fetuses had plasma PTHrP levels 11-fold higher than those of their littermates. Northern analysis, immunohistochemical, and in situ hybridization studies of Pthr1-null fetuses indicated that liver and placenta had increased expression of PTHRP: In summary, loss of fetal parathyroids in Hoxa3-null fetuses caused marked hypocalcemia but did not alter placental calcium transfer or the circulating PTHrP level. The findings in the Pthr1-null fetuses indicate that several tissues may contribute to the circulating PTHrP level in fetal mice.


Assuntos
Cálcio/metabolismo , Glândulas Paratireoides/fisiologia , Hormônio Paratireóideo/metabolismo , Placenta/metabolismo , Animais , Transporte Biológico , Calcitonina/metabolismo , Proteínas de Homeodomínio/genética , Camundongos , Camundongos Knockout , Glândulas Paratireoides/metabolismo , Proteína Relacionada ao Hormônio Paratireóideo , Proteínas/genética , Proteínas/metabolismo , Receptor Tipo 1 de Hormônio Paratireóideo , Receptores de Detecção de Cálcio , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo , Receptores de Hormônios Paratireóideos/genética , Receptores de Hormônios Paratireóideos/metabolismo , Distribuição Tecidual
12.
J Clin Invest ; 101(12): 2812-20, 1998 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-9637715

RESUMO

The calcium-sensing receptor (CaSR) regulates PTH secretion to control the extracellular calcium concentration in adults, but its role in fetal life is unknown. We used CaSR gene knockout mice to investigate the role of the CaSR in regulating fetal calcium metabolism. The normal calcium concentration in fetal blood is raised above the maternal level, an increase that depends upon PTH-related peptide (PTHrP). Heterozygous (+/-) and homozygous (-/-) disruption of the CaSR caused a further increase in the fetal calcium level. This increase was modestly blunted by concomitant disruption of the PTHrP gene and completely reversed by disruption of the PTH/ PTHrP receptor gene. Serum levels of PTH and 1, 25-dihydroxyvitamin D were substantially increased above the normal low fetal levels by disruption of the CaSR. The free deoxypyridinoline level was increased in the amniotic fluid (urine) of CaSR-/- fetuses; this result suggests that fetal bone resorption is increased. Placental calcium transfer was reduced, and renal calcium excretion was increased, by disruption of the CaSR. These studies indicate that the CaSR normally suppresses PTH secretion in the presence of the normal raised (and PTHrP-dependent) fetal calcium level. Disruption of the CaSR causes fetal hyperparathyroidism and hypercalcemia, with additional effects on placental calcium transfer.


Assuntos
Cálcio/metabolismo , Feto/metabolismo , Troca Materno-Fetal , Hormônio Paratireóideo/metabolismo , Placenta/metabolismo , Proteínas , Receptores de Superfície Celular/metabolismo , Animais , Feminino , Camundongos , Camundongos Knockout , Proteína Relacionada ao Hormônio Paratireóideo , Fragmentos de Peptídeos/metabolismo , Gravidez , Receptores de Detecção de Cálcio , Receptores de Superfície Celular/genética
13.
J Clin Invest ; 83(6): 2034-40, 1989 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-2723071

RESUMO

Parathyroid adenomas are common benign neoplasms for which no chromosomal defects have been described. We recently found two parathyroid adenomas bearing clonal restriction fragment abnormalities involving the PTH locus, and now show that in one of these tumors: (a) a DNA rearrangement occurred at the PTH locus; (b) the rearrangement separated the PTH gene's 5' flanking region from its coding exons, conceivably placing a newly adjacent gene under the influence of PTH regulatory elements; (c) the DNA that recombined with PTH normally maps to 11q13, the known chromosomal location of several oncogenes and the gene for multiple endocrine neoplasia type I; and (d) the rearrangement was a reciprocal, conservative recombination of the locus on 11q13 (Human Gene Mapping Library assignment D11S287) with PTH (on 11p15). These data provide molecular cytogenetic evidence for the clonal occurrence of a major chromosome 11 aberrancy in this benign parathyroid tumor. The D11S287 clone could prove useful in genetic linkage analyses, in determining precise 11q13 breakpoints in other neoplasms, and in identifying a gene on chromosome 11 that may participate in parathyroid tumor development.


Assuntos
Adenoma/genética , Mapeamento Cromossômico , Clonagem Molecular , Rearranjo Gênico , Hormônio Paratireóideo/genética , Neoplasias das Paratireoides/genética , Idoso , Southern Blotting , Cromossomos Humanos Par 11 , Sondas de DNA , Feminino , Genes , Humanos
14.
J Clin Invest ; 107(3): 277-86, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11160151

RESUMO

Parathyroid hormone (PTH), an important regulator of calcium homeostasis, targets most of its complex actions in bone to cells of the osteoblast lineage. Furthermore, PTH is known to stimulate osteoclastogenesis indirectly through activation of osteoblastic cells. To assess the role of the PTH/PTH-related protein receptor (PPR) in mediating the diverse actions of PTH on bone in vivo, we generated mice that express, in cells of the osteoblastic lineage, one of the constitutively active receptors described in Jansen's metaphyseal chondrodysplasia. In these transgenic mice, osteoblastic function was increased in the trabecular and endosteal compartments, whereas it was decreased in the periosteum. In trabecular bone of the transgenic mice, there was an increase in osteoblast precursors, as well as in mature osteoblasts. Osteoblastic expression of the constitutively active PPR induced a dramatic increase in osteoclast number in both trabecular and compact bone in transgenic animals. The net effect of these actions was a substantial increase in trabecular bone volume and a decrease in cortical bone thickness of the long bones. These findings, for the first time to our knowledge, identify the PPR as a crucial mediator of both bone-forming and bone-resorbing actions of PTH, and they underline the complexity and heterogeneity of the osteoblast population and/or their regulatory microenvironment.


Assuntos
Remodelação Óssea , Osso e Ossos/metabolismo , Osteoblastos/metabolismo , Hormônio Paratireóideo/fisiologia , Receptores de Hormônios Paratireóideos/genética , Fatores Etários , Animais , Osso e Ossos/citologia , Osso e Ossos/efeitos dos fármacos , Hibridização In Situ , Camundongos , Camundongos Transgênicos , Mutação , Osteoblastos/efeitos dos fármacos , Receptor Tipo 1 de Hormônio Paratireóideo , Receptores de Hormônios Paratireóideos/biossíntese , Transdução de Sinais , Tíbia/citologia , Tíbia/efeitos dos fármacos , Tíbia/metabolismo
15.
Mol Cell Biol ; 6(5): 1830-3, 1986 May.
Artigo em Inglês | MEDLINE | ID: mdl-3785180

RESUMO

The promoter region of the human parathyroid hormone gene was fused to the Escherichia coli neo gene and introduced into GH4C1 rat pituitary and human HeLa cells. Both TATA boxes of the human parathyroid hormone gene accurately directed transcription in GH4C1 cells; the parathyroid hormone promoter was inactive in HeLa cells.


Assuntos
Genes , Hormônio Paratireóideo/genética , Neoplasias Hipofisárias/metabolismo , Regiões Promotoras Genéticas , Transcrição Gênica , Animais , Linhagem Celular , Genes Bacterianos , Células HeLa/metabolismo , Humanos , Plasmídeos , Ratos
16.
Oncogene ; 8(2): 519-21, 1993 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-8426754

RESUMO

PRAD1 (cyclin D1) is a recently identified member of the family of cyclin genes, believed to play roles in regulating transitions through the cell cycle. The PRAD1 gene, located at 11q13, has been implicated in the pathogenesis of a variety of tumors, including parathyroid adenomas, t(11;14) bearing B-lymphoid tumors (particularly centrocytic lymphomas) where it is highly likely to be the BCL1 oncogene, and possibly in breast carcinomas and squamous cell cancers of the head and neck as well. PRAD1's tumorigenic influence appears to be effected through overexpression of its normal-sized transcript, but it has not been established whether the transcript's coding sequence is normal or contains oncogenic mutations. We have sequenced the coding region of the overexpressed PRAD1 transcript from two primary tumors with clonal PRAD1 region rearrangements: a benign parathyroid adenoma and a malignant centrocytic lymphoma. Each sequence is identical to the normal PRAD1 cDNA sequence, and presumably encodes normal PRAD1 protein. Thus, PRAD1 likely functions as a direct-acting oncogene whose rearrangement in tumors leads to overexpression or deregulated expression of its normal protein product.


Assuntos
Adenoma/genética , Ciclinas/genética , Expressão Gênica , Linfoma/genética , Proteínas Oncogênicas/genética , Oncogenes , Neoplasias das Paratireoides/genética , Sequência de Bases , Ciclina D1 , Rearranjo Gênico , Humanos , Dados de Sequência Molecular
17.
Oncogene ; 6(3): 449-53, 1991 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-2011400

RESUMO

We report the detailed molecular characterization of a human parathyroid adenoma with a clonal parathyroid hormone gene rearrangement. This rearrangement is similar to one we characterized recently in an independent adenoma. In these two, plus a third partially characterized adenoma, one allele of the PTH gene, on 11p15, is rearranged with DNA from the D11S287 region on 11q13. This region contains a transcribed sequence, D11S287E, distinct from known 11q13 oncogenes, that is expressed in all parathyroid tissues examined, but is overexpressed dramatically in all three tumours with PTH gene-D11S287 rearrangements. These findings suggest that overexpression of D11S287E, perhaps driven by the misplaced PTH gene's regulatory elements, contributed to the development of these benign tumors. D11S287E is a new candidate oncogene with potential importance in parathyroid adenomas and perhaps other tumors with 11q13 abnormalities.


Assuntos
Adenoma/genética , Cromossomos Humanos Par 11 , Regulação Neoplásica da Expressão Gênica , Rearranjo Gênico , Oncogenes , Neoplasias das Paratireoides/genética , Northern Blotting , Southern Blotting , DNA de Neoplasias/análise , DNA de Neoplasias/genética , Humanos , Hibridização de Ácido Nucleico , Hormônio Paratireóideo/genética , RNA Neoplásico/análise , RNA Neoplásico/genética , Mapeamento por Restrição
18.
Mech Dev ; 81(1-2): 151-61, 1999 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10330492

RESUMO

A number of studies suggest a role for PTHrP and the classical PTH/PTHrP receptor (type I) in one of the first differentiation processes in mouse embryogenesis, i.e. the formation of parietal endoderm (PE). We previously reported that although in type I receptor (-/-) embryos PE formation seemed normal, the embryos were smaller from at least day 9.5 p.c. and 60% had died before day 12.5 p.c. Here we show that the observed growth defect commences even earlier, at day 8.5 p.c. Using two novel antibodies, we show that the expression of the type I receptor protein at this stage is confined to extraembryonic endoderm only. In addition, we show that large amounts of PTHrP protein are present in the adjacent trophoblast giant cells, suggesting a paracrine interaction of PTHrP and the type I PTH/PTHrP receptor in PE formation. The involvement in PE differentiation of other recently described receptors for PTHrP would explain a possible redundancy for the type I receptor in PE formation. However, deletion of the type I PTH/PTHrP receptor in ES cells by homologous recombination completely prevents PTHrP-induced PE differentiation. Based upon these observations, we propose that PTHrP and the type I PTH/PTHrP receptor, although not required for the initial formation of PE, are required for its proper differentiation and/or functioning.


Assuntos
Ectoderma/fisiologia , Proteínas/fisiologia , Receptores de Hormônios Paratireóideos/fisiologia , Animais , Western Blotting , Células COS , Células Cultivadas , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Feminino , Imunofluorescência , Regulação da Expressão Gênica no Desenvolvimento , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Proteína Relacionada ao Hormônio Paratireóideo , Proteínas/análise , Receptor Tipo 1 de Hormônio Paratireóideo , Receptores de Hormônios Paratireóideos/análise , Células-Tronco/metabolismo , Trombomodulina/metabolismo , Fatores de Tempo , Transfecção
19.
Mol Endocrinol ; 4(1): 69-74, 1990 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-2325669

RESUMO

The differential expression of mRNAs between the closely related rat osteosarcoma cell lines ROS 17/2.8 and ROS 25/1 was used to identify genes whose expression is associated with the osteoblast phenotype. Thymosin beta 4 cDNA was cloned from an ROS 17/2.8 complimentary DAN library on the basis of its differential hybridization with radiolabeled cDNA prepared from ROS 17/2.8 and ROS 25/1 cells. Northern blot analysis confirmed that thymosin beta 4, hitherto a putative immunodulatory hormone, was indeed differentially expressed. Steady state mRNA levels were severalfold higher in ROS 17/2.8 cells exhibiting an osteoblast-like phenotype, compared with the less osteoblast-like ROS 25/1. Thymosin beta 4 transcripts were also detected in rat UMR 106 osteosarcoma cells and in intact neonatal and fetal rat calvaria. Sequence analysis of the cDNA indicated that thymosin beta 4 transcripts may arise by processing at a more distal polyadenylation signal. Treatment of ROS 17/2.8 cells with dexamethasone increased, while addition of 1,25-dihydroxyvitamin D3 decreased thymosin beta 4 mRNA. The phenotype-dependent expression in the ROS cells and the response to steroid hormone suggest that thymosin beta 4 expression contributes to the osteoblast phenotype.


Assuntos
Neoplasias Ósseas/genética , Osteossarcoma/genética , Timosina/análogos & derivados , Sequência de Aminoácidos , Animais , Sequência de Bases , Neoplasias Ósseas/metabolismo , Calcitriol/farmacologia , DNA/biossíntese , Dexametasona/farmacologia , Regulação para Baixo/efeitos dos fármacos , Dados de Sequência Molecular , Osteossarcoma/metabolismo , RNA Mensageiro/biossíntese , Ratos , Timosina/biossíntese , Timosina/genética , Células Tumorais Cultivadas , Regulação para Cima/efeitos dos fármacos
20.
Mol Endocrinol ; 3(1): 60-7, 1989 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-2536893

RESUMO

PTH activates multiple acute intracellular signals within responsive target cells, but the importance of cAMP vs. other second messenger signals in mediating different biological responses to PTH is not known. To address these questions, we developed a genetic approach to block activation of the cAMP-dependent protein kinase (PK-A) in PTH-responsive cell lines. Clonal rat osteosarcoma cells (UMR 106-01) were stably transfected with REV-I, a plasmid that directs synthesis of a mutant cAMP-resistant form of the type I regulatory subunit of PK-A. In the transfected bone cells, most of the catalytic subunits of PK-A were associated with the mutant regulatory subunit, and activation of PK-A by cAMP was correspondingly inhibited. We have characterized one such mutant (UMR 4-7) that expressed large amounts of mutant mRNA and exhibited inducible blockade of PK-A via the REV-1 metallothionein promoter. In the absence of metallothionein induction, these cells exhibited nearly normal PTH responsiveness, but after REV-1 induction by Zn2+, they were resistant to PTH-induced activation of PK-A and regulation of membrane phospholipid synthesis by both PTH and cAMP analogs. The mutant UMR 4-7 cell provides a model system in which the consequences of cAMP production by PTH or other agonists that activate adenylate cyclase in osteoblasts may be specifically inhibited by brief exposure to Zn2+. Such mutant cell lines will facilitate further investigation of the linkage between early signalling events and subsequent biological responses in the action of PTH and other agonists on target cells in bone.


Assuntos
AMP Cíclico/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Isoenzimas/genética , Osteoblastos/metabolismo , Hormônio Paratireóideo/farmacologia , Proteínas Quinases/genética , Animais , Northern Blotting , Ativação Enzimática/efeitos dos fármacos , Isoenzimas/biossíntese , Metalotioneína/genética , Mutação , Osteoblastos/efeitos dos fármacos , Osteossarcoma , Fosfatidiletanolaminas/biossíntese , Plasmídeos , Regiões Promotoras Genéticas , Proteínas Quinases/biossíntese , RNA Mensageiro/genética , Ratos , Transcrição Gênica , Transfecção , Células Tumorais Cultivadas , Zinco/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA