Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Osteoporos Int ; 30(8): 1607-1616, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31053927

RESUMO

Glucorticoid (GC) therapy is the commonest cause of secondary osteoporosis. Ovariectomized rabbits receiving the GC methylprednisolone for 6 weeks exhibited relatively lower vertebral and femoral bone mass. Treatment with the PTH receptor agonist abaloparatide for 12 weeks during ongoing methylprednisolone administration increased cortical and trabecular bone mass and femur bending strength. INTRODUCTION: Abaloparatide, an osteoanabolic PTHrP analog, increases bone mineral density (BMD) and reduces fracture risk in women with postmenopausal osteoporosis. This study assessed abaloparatide effects on BMD and bone strength in ovariectomized (OVX) rabbits with glucocorticoid (GC)-induced osteopenia. METHODS: Thirty-two rabbits underwent OVX and 8 underwent sham surgery. One day later, 24 OVX rabbits began daily s.c. GC injections (methylprednisolone, 1 mg/kg/day) for 6 weeks, while 8 OVX and 8 sham controls received no GC. GC-challenged rabbits (8/group) then received GC (0.5 mg/kg/day) along with daily s.c. vehicle (GC-OVX), abaloparatide 5 µg/kg/day (ABL5), or 25 µg/kg/day (ABL25) for 12 weeks, and the no-GC OVX and sham controls received daily vehicle. RESULTS: GC-OVX rabbits showed significant deficits in vertebral and proximal femur areal BMD, lower cortical area, thickness and volumetric BMD of the femur diaphysis, and reduced trabecular bone volume and volumetric BMD in the vertebra and distal femur versus sham controls. These deficits were significantly reversed in the ABL25 group, which also showed enhanced trabecular micro-architecture versus GC-OVX controls. Destructive bending tests showed significantly lower femur diaphysis ultimate load and bending rigidity of the femoral diaphysis in the GC-OVX group versus sham controls, whereas these parameters were similar in the ABL25 group vs sham controls. CONCLUSIONS: Abaloparatide 25 µg/kg/day mitigated the adverse effects of GC administration on cortical and trabecular bone and improved femoral strength in OVX rabbits. These results suggest potential promise for abaloparatide as an investigational therapy for glucocorticoid-induced osteoporosis.


Assuntos
Conservadores da Densidade Óssea/uso terapêutico , Densidade Óssea/efeitos dos fármacos , Doenças Ósseas Metabólicas/tratamento farmacológico , Proteína Relacionada ao Hormônio Paratireóideo/uso terapêutico , Absorciometria de Fóton , Animais , Fenômenos Biomecânicos , Conservadores da Densidade Óssea/farmacologia , Doenças Ósseas Metabólicas/induzido quimicamente , Doenças Ósseas Metabólicas/fisiopatologia , Feminino , Fêmur/fisiopatologia , Glucocorticoides , Vértebras Lombares/fisiopatologia , Metilprednisolona , Ovariectomia , Proteína Relacionada ao Hormônio Paratireóideo/farmacologia , Coelhos , Microtomografia por Raio-X
2.
Anat Rec (Hoboken) ; 293(7): 1214-26, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20583265

RESUMO

Fibroblast growth factor-23 (FGF23) is a hormone that modulates circulating phosphate (P(i)) levels by controlling P(i) reabsorption from the kidneys. When FGF23 levels are deficient, as in tumoral calcinosis patients, hyperphosphatemia ensues. We show here in a murine model that Fgf23 ablation disrupted morphology and protein expression within the dentoalveolar complex. Ectopic matrix formation in pulp chambers, odontoblast layer disruption, narrowing of periodontal ligament space, and alteration of cementum structure were observed in histological and electron microscopy sections. Because serum P(i) levels are dramatically elevated in Fgf23(-/-), we assayed for apoptosis and expression of members from the small integrin-binding ligand, N-linked glycoprotein (SIBLING) family, both of which are sensitive to elevated P(i) in vitro. Unlike X-linked hypophosphatemic (Hyp) and wild-type (WT) specimens, numerous apoptotic osteocytes and osteoblasts were detected in Fgf23(-/-) specimens. Further, in comparison to Hyp and WT samples, decreased bone sialoprotein and elevated dentin matrix protein-1 protein levels were observed in cementum of Fgf23(-/-) mice. Additional dentin-associated proteins, such as dentin sialoprotein and dentin phosphoprotein, exhibited altered localization in both Fgf23(-/-) and Hyp samples. Based on these results, we propose that FGF23 and (P(i)) homeostasis play a significant role in maintenance of the dentoalveolar complex.


Assuntos
Processo Alveolar/patologia , Fatores de Crescimento de Fibroblastos/genética , Hiperfosfatemia/patologia , Animais , Proteínas da Matriz Extracelular/metabolismo , Raquitismo Hipofosfatêmico Familiar/genética , Raquitismo Hipofosfatêmico Familiar/metabolismo , Fator de Crescimento de Fibroblastos 23 , Doenças Genéticas Ligadas ao Cromossomo X , Humanos , Hiperfosfatemia/genética , Hiperfosfatemia/metabolismo , Camundongos , Camundongos Knockout , Osteócitos/metabolismo , Fosfatos/metabolismo
3.
J Dent Res ; 89(4): 349-54, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20200412

RESUMO

Indian hedgehog (Ihh) is essential for embryonic mandibular condylar growth and disc primordium formation. To determine whether it regulates those processes during post-natal life, we ablated Ihh in cartilage of neonatal mice and assessed the consequences on temporomandibular joint (TMJ) growth and organization over age. Ihh deficiency caused condylar disorganization and growth retardation and reduced polymorphic cell layer proliferation. Expression of Sox9, Runx2, and Osterix was low, as was that of collagen II, collagen I, and aggrecan, thus altering the fibrocartilaginous nature of the condyle. Though a disc formed, it exhibited morphological defects, partial fusion with the glenoid bone surface, reduced synovial cavity space, and, unexpectedly, higher lubricin expression. Analysis of the data shows, for the first time, that continuous Ihh action is required for completion of post-natal TMJ growth and organization. Lubricin overexpression in mutants may represent a compensatory response to sustain TMJ movement and function.


Assuntos
Cartilagem Articular/crescimento & desenvolvimento , Proteínas Hedgehog/fisiologia , Côndilo Mandibular/crescimento & desenvolvimento , Articulação Temporomandibular/anatomia & histologia , Articulação Temporomandibular/crescimento & desenvolvimento , Agrecanas/biossíntese , Agrecanas/genética , Animais , Anquilose/genética , Anquilose/metabolismo , Cartilagem Articular/anatomia & histologia , Condrócitos/patologia , Colágeno Tipo II/biossíntese , Colágeno Tipo II/genética , Subunidade alfa 1 de Fator de Ligação ao Core/biossíntese , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Regulação para Baixo , Fibrocartilagem/anatomia & histologia , Fibrocartilagem/crescimento & desenvolvimento , Lâmina de Crescimento/anormalidades , Proteínas Hedgehog/genética , Côndilo Mandibular/anatomia & histologia , Camundongos , Camundongos Knockout , Proteoglicanas/biossíntese , Fatores de Transcrição SOX9/biossíntese , Fatores de Transcrição SOX9/genética , Fator de Transcrição Sp7 , Disco da Articulação Temporomandibular/anatomia & histologia , Disco da Articulação Temporomandibular/crescimento & desenvolvimento , Transtornos da Articulação Temporomandibular/genética , Transtornos da Articulação Temporomandibular/metabolismo , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética
4.
J Periodontol ; 80(8): 1348-54, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19656036

RESUMO

BACKGROUND: Cementogenesis is sensitive to altered local phosphate levels; thus, we hypothesized a cementum phenotype, likely of decreased formation, would be present in the teeth of X-linked hypophosphatemic (Hyp) mice. Mutations in the phosphate-regulating gene with homologies to endopeptidases on the X chromosome (Phex) cause X-linked hypophosphatemia, characterized by rickets, osteomalacia, and hypomineralized dentin formation, a phenotype recapitulated in the Hyp mouse homolog. Here, we report a developmental study of tooth root formation in Hyp mouse molars, focusing on dentin and cementum. METHODS: Light and transmission electron microscopy were used to study molar tissues from wild-type (WT) and Hyp mice. Demineralized and hematoxylin and eosin-stained tissues at developmental stages 23 to 96 days postcoital (dpc) were examined by light microscopy. Immunohistochemistry methods were used to detect bone sialoprotein (BSP) distribution in Hyp and WT mouse molar tissues, and transmission electron microscopy was used to study similar molar tissues in the non-demineralized state. RESULTS: Dentin in Hyp mice exhibited mineralization defects by 33 dpc, as expected, but this defect was partially corrected by 96 dpc. In support of our hypothesis, a cementum phenotype was detected using a combination of immunohistochemistry and transmission electron microscopy, which included thinner BSP-positive staining within the cementum, discontinuous mineralization, and a globular appearance compared to WT controls. CONCLUSION: Mutations in the phosphate-regulating Phex gene of the Hyp mouse resulted in defective cementum development.


Assuntos
Cementogênese/genética , Cemento Dentário/anormalidades , Raquitismo Hipofosfatêmico Familiar/patologia , Doenças Genéticas Ligadas ao Cromossomo X , Animais , Cemento Dentário/patologia , Dentina/anormalidades , Dentina/patologia , Dentinogênese/genética , Feminino , Idade Gestacional , Imuno-Histoquímica , Sialoproteína de Ligação à Integrina , Masculino , Camundongos , Camundongos Mutantes , Microscopia Eletrônica de Transmissão , Dente Molar/anormalidades , Dente Molar/patologia , Mutação/genética , Odontogênese/genética , Endopeptidase Neutra Reguladora de Fosfato PHEX/genética , Fenótipo , Sialoglicoproteínas/análise , Calcificação de Dente/genética , Germe de Dente/anormalidades , Germe de Dente/patologia , Raiz Dentária/anormalidades , Raiz Dentária/patologia
5.
J Pathol ; 216(3): 345-55, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18729070

RESUMO

Genetic ablation of fibroblast growth factor 23 from mice (Fgf-23(-/-)) results in a short lifespan with numerous abnormal biochemical and morphological features. Such features include kyphosis, hypogonadism and associated infertility, osteopenia, pulmonary emphysema, severe vascular and soft tissue calcifications, and generalized atrophy of various tissues. To determine whether these widespread anomalies in Fgf-23(-/-) mice can be ameliorated by genetically restoring the systemic actions of FGF-23, we generated Fgf-23(-/-) mice expressing the human FGF-23 transgene in osteoblasts under the control of the 2.3 kb alpha1(I) collagen promoter (Fgf-23(-/-) /hFGF-23-Tg double mutants). This novel mouse model is completely void of all endogenous Fgf-23 activity, but produces human FGF-23 in bone cells that is subsequently released into the circulation. Our results suggest that lack of Fgf-23 activities results in extensive premature ageing-like features and early mortality of Fgf-23(-/-) mice, while restoring the systemic effects of FGF-23 significantly ameliorates these phenotypes, with the resultant effect being improved growth, restored fertility, and significantly prolonged survival of double mutants. With regard to their serum biochemistry, double mutants reversed the severe hyperphosphataemia, hypercalcaemia, and hypervitaminosis D found in Fgf-23(-/-) littermates; rather, double mutants show hypophosphataemia and normal serum 1,25-dihydroxyvitamin D(3) levels similar to pure FGF-23 Tg mice. These changes were associated with reduced renal expression of NaPi2a and 1 alpha-hydroxylase, compared to Fgf-23(-/-) mice. FGF-23 acts to prevent widespread abnormal features by acting systemically to regulate phosphate homeostasis and vitamin D metabolism. This novel mouse model provides us with an in vivo tool to study the systemic effects of FGF-23 in regulating mineral ion metabolism and preventing multiple abnormal phenotypes without the interference of native Fgf-23.


Assuntos
Senilidade Prematura/genética , Fatores de Crescimento de Fibroblastos/genética , Osteoblastos/metabolismo , Senilidade Prematura/metabolismo , Senilidade Prematura/patologia , Animais , Biomarcadores/sangue , Osso e Ossos/diagnóstico por imagem , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Calcitriol/sangue , Cálcio/sangue , Feminino , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/metabolismo , Expressão Gênica , Engenharia Genética , Genótipo , Membro Posterior , Humanos , Mucosa Intestinal/patologia , Pulmão/patologia , Masculino , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Modelos Animais , Hormônio Paratireóideo/sangue , Fosfatos/sangue , Radiografia , Reação em Cadeia da Polimerase Via Transcriptase Reversa/métodos , Pele/patologia , Transgenes
6.
Cytogenet Genome Res ; 97(1-2): 106-10, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12438747

RESUMO

The vertebrate Patched 2 (Ptch2) gene encodes a putative membrane-embedded protein which may have roles in Hedgehog signaling during development and in tumorigenesis. We determined the genomic structure of the mouse Ptch2 gene and show that Ptch2 is composed of 22 exons spanning approximately 18 kb of genomic DNA. The exon-intron boundaries were found to be conserved within the human and mouse Ptch2 genes. Analysis of the 5' flanking region revealed a CpG island, the putative promoter region and the transcriptional start site while a polyadenylation signal as well as a mRNA destabilizing motif were identified in the 3' flanking region. Single-strand conformation polymorphism analysis was used to map mouse Ptch2 to chromosome 4 between the microsatellite markers D4Mit20 and D4Mit334.


Assuntos
Proteínas de Membrana/genética , Animais , Sequência de Bases , Mapeamento Cromossômico , Ilhas de CpG , DNA/genética , Éxons , Genoma , Humanos , Íntrons , Camundongos , Repetições de Microssatélites , Dados de Sequência Molecular , Receptores Patched , Receptor Patched-2 , Polimorfismo Conformacional de Fita Simples , Receptores de Superfície Celular
7.
Endocrinology ; 142(12): 5303-10, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11713230

RESUMO

The homozygous ablation of the gene encoding the PTH/PTHrP receptor (PPR(-/-)) leads to early lethality and limited developmental defects, including an acceleration of chondrocyte differentiation. In contrast to the findings in homozygous PTHrP-ablated (PTHrP(-/-)) animals, these PPR(-/-) mice show an increase in cortical bone, a decrease in trabecular bone, and a defect in bone mineralization. Opposite observations are made in Jansen's metaphyseal chondrodysplasia, a disorder caused by constitutively active PPR mutants, and in transgenic animals expressing one of these receptor mutants (HKrk-H223R) under control of the type alpha1(I) collagen promoter. Expression of the Jansen transgene under the control of the type alpha1(II) collagen promoter was, furthermore, shown to delay chondrocyte differentiation and to prevent the dramatic acceleration of chondrocyte differentiation in PTHrP(-/-) mice, thus rescuing the early lethality of these animals. In the present study we demonstrated that the type alpha1(II) collagen promoter Jansen transgene restored most of the bone abnormalities in PPR(-/-) mice, but did not prevent their perinatal lethality. These findings suggested that factors other than impaired gas exchange due to an abnormal rib cage contribute to the early death of PPR(-/-) mice.


Assuntos
Exostose Múltipla Hereditária/genética , Expressão Gênica , Marcação de Genes , Receptores de Hormônios Paratireóideos/deficiência , Transgenes , Animais , Condrócitos/fisiologia , Colágeno Tipo II/genética , Embrião de Mamíferos/patologia , Exostose Múltipla Hereditária/mortalidade , Exostose Múltipla Hereditária/patologia , Lâmina de Crescimento/metabolismo , Homozigoto , Camundongos , Camundongos Knockout , Mutação , Fenótipo , Regiões Promotoras Genéticas/fisiologia , Receptor Tipo 1 de Hormônio Paratireóideo , Receptores de Hormônios Paratireóideos/genética , Receptores de Hormônios Paratireóideos/metabolismo , Análise de Sobrevida , Tíbia/embriologia , Tíbia/patologia
8.
Endocrinology ; 142(3): 1260-8, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11181543

RESUMO

Type-1 PTH/PTH-related peptide receptors (PTH1Rs), which activate both adenylyl cyclase and phospholipase C (PLC), control endochondral bone development by regulating chondrocyte differentiation. To directly analyze PTH1R function in such cells, we isolated conditionally transformed clonal chondrocytic cell lines from tibial growth plates of neonatal mice heterozygous for PTH1R gene ablation. Among 104 cell lines isolated, messenger RNAs for PTH1R, collagen II, and collagen X were detected in 28%, 90%, and 29%, respectively. These cell lines were morphologically diverse. Some appeared large, rounded, and enveloped by abundant extracellular matrix; whereas others were smaller, flattened, and elongated. Two PTH1R-expressing clones showed similar PTH1R binding and cAMP responsiveness to PTH and PTH-related peptide but disparate morphologic features, characteristic of hypertrophic (hC1--5) or nonhypertrophic (nhC2--27) chondrocytes, respectively. hC1--5 cells expressed messenger RNAs for collagen II and X, alkaline phosphatase (ALP), and matrix GLA protein, whereas nhC2--27 cells expressed collagen II and Indian hedgehog but not collagen X or ALP. In hC1--5 cells, PTH and cAMP analog, but not phorbol ester, inhibited both ALP and mineralization. PTH1R-null hC1--5 subclones were isolated by in vitro selection and then reconstituted by stable transfection with wild-type PTH1Rs or mutant (DSEL) PTH1Rs defective in PLC activation. ALP and mineralization were inhibited similarly via both forms of the receptor. These results indicate that PLC activation is not required for PTH1R regulation of mineralization or ALP in hypertrophic chondrocytes and are consistent with a major role for cAMP in regulating differentiation of hypertrophic chondrocytes.


Assuntos
Condrócitos/citologia , Lâmina de Crescimento/citologia , Receptores de Hormônios Paratireóideos/fisiologia , Transdução de Sinais/fisiologia , Animais , Diferenciação Celular/fisiologia , Linhagem Celular Transformada , Camundongos , Camundongos Knockout/genética , Receptor Tipo 1 de Hormônio Paratireóideo , Receptores de Hormônios Paratireóideos/genética , Tíbia
9.
Bone ; 25(4): 397-404, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10511105

RESUMO

Bone morphogenetic protein-7, or BMP-7 (OP-1), is highly expressed in the perichondrium of embryonic long bones and is thought to play a role in endochondral ossification. Previously we have shown that BMP-7 inhibits terminal chondrocyte differentiation; that is, chondrocyte hypertrophy and mineralization in cultured explants of embryonic mouse metatarsals. However, the mechanism of this inhibition and the target cells of BMP-7 are still unknown. In this study we show that BMP-7 inhibits terminal chondrocyte differentiation indirectly, via an interaction with the periarticular region of the explants. This region also expresses parathyroid hormone-related peptide (PTHrP). PTHrP regulates terminal chondrocyte differentiation by inhibiting hypertrophic differentiation of prehypertrophic chondrocytes. The differentiating center in turn regulates PTHrP expression via a feedback loop involving Indian hedgehog (Ihh), which is expressed in the prehypertrophic chondrocytes. Ihh is thought to act on perichondrial cells, which in turn start to express an as yet unknown mediator that stimulates PTHrP expression in the periarticular region. It has been suggested that this factor belongs to the BMP-family. We investigated whether the inhibition of terminal chondrocyte differentiation by BMP-7 was due to upregulation of the PTHrP-Ihh feedback loop and whether BMP-7 was the unknown factor in the loop. Here we show that exogenous BMP-7 did not upregulate the mRNA expression of PTHrP, Ihh, or the PTH/PTHrP receptor in cultured wild-type embryonic metatarsals. Furthermore, BMP-7 could still inhibit terminal chondrocyte differentiation in the metatarsals of PTHrP-deficient (PTHrP-/-) mouse embryos. These data indicate that the BMP-7-mediated inhibition of terminal chondrocyte differentiation in vitro is independent of the PTHrP-Ihh feedback loop. We concluded that BMP-7 modulates terminal chondrocyte differentiation and cartilage mineralization of fetal bone explants in vitro via as yet unknown inhibitory factor(s) produced in the periarticular region.


Assuntos
Proteínas Morfogenéticas Ósseas/farmacologia , Condrócitos/citologia , Ossos do Metatarso/citologia , Proteínas/fisiologia , Fator de Crescimento Transformador beta , Animais , Proteína Morfogenética Óssea 7 , Diferenciação Celular/efeitos dos fármacos , Tamanho Celular/efeitos dos fármacos , Condrócitos/efeitos dos fármacos , Condrócitos/fisiologia , Retroalimentação , Ossos do Metatarso/embriologia , Ossos do Metatarso/fisiologia , Camundongos , Técnicas de Cultura de Órgãos , Proteína Relacionada ao Hormônio Paratireóideo , Transdução de Sinais
10.
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
11.
Mol Endocrinol ; 13(7): 1183-96, 1999 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-10406468

RESUMO

We have shown previously that the PTH/PTHrP (PTH-related peptide) receptor mRNA becomes expressed very early in murine embryogenesis, i.e. during the formation of extraembryonic endoderm. Retinoic Acid (RA) is a potent inducer of extraembryonic endoderm formation and PTH/PTHrP-receptor expression in embryonal carcinoma (EC) and embryonal stem (ES) cells. Using the P19 EC cell line, we have characterized promoter elements of the murine PTH/PTHrP-receptor gene that are involved in this RA-induced expression. The data show that RA-induced expression of the PTH/ PTHrP-receptor gene is mediated by the downstream P2 promoter. Analysis of promoter reporter constructs in transiently transfected P19 cells treated with RA identified an enhancer region between nucleotides -2714 and -2702 upstream of the P2 transcription start site that is involved in the RA effect. This region matches a consensus hormone response element consisting of a direct repeat with an interspacing of 1 bp (R-DR1). The R-DR1 efficiently binds retinoic acid receptor-alpha (RARalpha)-retinoid X receptor-alpha (RXRalpha) and chicken ovalbumin upstream promoter (COUP)-transcription factor I (TFI)-RXRalpha heterodimers and RXRalpha and COUP-TFI homodimers in a bandshift assay using extracts of transiently transfected COS-7 cells. RA differentiation of P19 EC cells strongly increases protein binding to the R-DR1 in a band-shift assay. This is caused by increased expression of RXR (alpha, beta, or gamma) and by the induction of expression of RARbeta and COUP TFI/TFII, which bind to the R-DR1 as shown by supershifting antibodies. The presence of RXR (alpha, beta, or gamma) in the complexes binding to the R-DR1 suggests that RXR homodimers are involved in RA-induced expression of the PTH/PTHrP-receptor gene. The importance of the R-DR1 for RA-induced expression of PTH/ PTHrP-receptor was shown by an inactivating mutation of the R-DR1, which severely impairs RA-induced expression of PTH/PTHrP-receptor promoter reporter constructs. Since this mutation does not completely abolish RA-induced expression of PTH/PTHrP-receptor promoter reporter constructs, sequences other than the R-DR1 might also be involved in the RA effect. Finally, we show that the RA-responsive promoter region is also able to induce expression of a reporter gene in extraembryonic endoderm of 7.5 day-old transgenic mouse embryos.


Assuntos
Hormônio Paratireóideo/genética , Proteínas/genética , Receptores de Esteroides , Elementos de Resposta/fisiologia , Tretinoína/metabolismo , Animais , Sequência de Bases , Fatores de Transcrição COUP , Carcinoma Embrionário/genética , Carcinoma Embrionário/metabolismo , Diferenciação Celular/efeitos dos fármacos , Proteínas de Ligação a DNA/metabolismo , Endoderma/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Dados de Sequência Molecular , Mutação , Hormônio Paratireóideo/metabolismo , Proteína Relacionada ao Hormônio Paratireóideo , Regiões Promotoras Genéticas , Proteínas/metabolismo , Receptores do Ácido Retinoico/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Sequências Repetitivas de Ácido Nucleico , Receptores X de Retinoides , Fatores de Transcrição/metabolismo , Transcrição Gênica , Tretinoína/farmacologia , Células Tumorais Cultivadas , beta-Galactosidase/genética , beta-Galactosidase/metabolismo
12.
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
13.
J Bone Miner Res ; 13(12): 1835-45, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9844101

RESUMO

Osteoblasts synthesize and mineralize bone matrix and are principal target cells for parathyroid hormone (PTH). The type 1 PTH/PTH-related protein (PTHrP) receptor (PTH1R), cloned from rat osteoblastic cells, activates multiple intracellular signaling mechanisms. The specific roles of these PTH1R signals, or of responses to other types of PTH receptors that may be expressed, in regulating osteoblast function are incompletely understood. Use of established mammalian osteoblastic cell lines has led to much understanding of PTH action in bone, although such cells are of neoplastic origin or have other characteristics that compromise their validity as models of normal osteoblasts. To examine the role of the PTH1R in osteoblast biology, we have isolated a series of clonal murine calvarial osteoblastic cell lines that are only conditionally immortalized, via expression of a transgene encoding the tsA58 temperature-sensitive SV40 large T antigen, and that lack both functional alleles of the PTH1R gene. When cultured under nontransforming conditions, these cells stopped proliferating, expressed a series of characteristic osteoblastic genes (including the nonfunctional remnant of the PTH1R gene), and, after 3-4 weeks, produced mineralized bone nodules in a manner that was regulated by 1,25-dihydroxyvitamin D3 but not by PTH(1-84). Cyclic AMP measurements revealed no evidence of expression of alternate species of Gs-linked PTH receptors. Stable transfection with PTH1R cDNA reconstituted both PTH binding and adenylyl cyclase activation, increased basal osteocalcin expression, and supported PTH stimulation of c-Fos expression and matrix mineralization. These conditionally transformed, PTH1R(-/-) clonal osteoblastic cell lines should prove useful for studies of the regulation of osteoblast differentiation and function by both endogenous nonclassical species of PTH (or PTHrP) receptors and mutant signal-selective PTH1Rs.


Assuntos
Osteoblastos/fisiologia , Receptores de Hormônios Paratireóideos/deficiência , Adenilil Ciclases/metabolismo , Fosfatase Alcalina/análise , Alelos , Animais , Western Blotting , Calcificação Fisiológica , Bovinos , Linhagem Celular Transformada , Separação Celular , AMP Cíclico/metabolismo , Humanos , Camundongos , Hormônio Paratireóideo/metabolismo , Fenótipo , Reação em Cadeia da Polimerase , Ratos , Receptor Tipo 1 de Hormônio Paratireóideo , Receptores de Hormônios Paratireóideos/genética , Receptores de Hormônios Paratireóideos/fisiologia , Transgenes
14.
Endocrinology ; 139(12): 5194-204, 1998 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-9832460

RESUMO

PTH and PTH-related peptide (PTHrP) have been shown to bind to and activate the same PTH/PTHrP receptor. Recent studies have demonstrated, however, the presence of additional receptors specific for each ligand. We used the PTHrP and PTH/PTHrP receptor gene knock-out models to investigate whether this receptor mediates the actions of both ligands in bone. The similar phenotype of the PTHrP (-/-) and PTH/PTHrP receptor (-/-) animals in the growth plate of the tibia suggests that this receptor mediates the actions of PTHrP. Electron microscopic studies have confirmed the accelerated differentiation and disordered organization of chondrocytes, with the accumulation of large amounts of dispersed glycogen granules in the cytoplasm of proliferative and maturing cells of both genotypes. The contrasting growth plate mineralization patterns of the PTHrP (-/-) and PTH/PTHrP receptor (-/-) mice, however, suggest that the actions of PTHrP and the PTH/PTHrP receptor are not identical. Studies using calvariae from PTH/PTHrP receptor (-/-) embryos demonstrate that this receptor solely mediates the ability of PTH and PTHrP to stimulate adenylate cyclase in bone and to stimulate bone resorption. Furthermore, we show that osteoblasts of PTH/PTHrP receptor (-/-) animals, but not PTHrP (-/-) animals, have decreased levels of collagenase 3, osteopontin, and osteocalcin messenger RNAs. The PTH/PTHrP receptor, therefore, mediates distinct physiologic actions of both PTH and PTHrP.


Assuntos
Osso e Ossos/efeitos dos fármacos , Osso e Ossos/metabolismo , Hormônio Paratireóideo/farmacologia , Proteínas/farmacologia , Receptores de Hormônios Paratireóideos/fisiologia , Animais , Densidade Óssea/fisiologia , Reabsorção Óssea/fisiopatologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Condrócitos/citologia , Condrócitos/ultraestrutura , Lâmina de Crescimento/metabolismo , Ligantes , Camundongos , Camundongos Knockout/genética , Microscopia Eletrônica , Mutação/fisiologia , Osteoblastos/citologia , Proteína Relacionada ao Hormônio Paratireóideo , Fenótipo , Proteínas/genética , Receptor Tipo 1 de Hormônio Paratireóideo , Receptores de Hormônios Paratireóideos/genética
15.
Crit Rev Eukaryot Gene Expr ; 8(3-4): 297-320, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9807698

RESUMO

Parathyroid hormone-related peptide (PTHrP) has been identified as the factor responsible for the humoral hypercalcemia of malignancy (HHM). Since the cloning of the cDNA, it has become clear that PTHrP is a prohormone that is posttranslationally cleaved to yield a complex family of peptides. Through its homology to parathyroid hormone (PTH) in the amino-terminus region of the protein, it is able to bind to and activate a common PTH/PTHrP receptor. PTHrP has been shown to be a normal product of many adult and fetal tissues, where it appears to act in an autocrine/paracrine fashion to regulate organogenesis. PTHrP and the PTH/PTHrP receptor seem to be co-expressed in many tissues, but their role in the various systems is uncertain. The use of transgenic and knock-out animal models has contributed to a better understanding of the physiological role of this peptide and its receptor. In this review, the structure of their genes, their expression pattern, and some of their major physiological functions are discussed. Attention is focused on their interaction in the regulation of cartilage and bone development.


Assuntos
Hormônio Paratireóideo/fisiologia , Proteínas/fisiologia , Receptores de Hormônios Paratireóideos/fisiologia , Adulto , Animais , Cartilagem , Humanos , Hormônio Paratireóideo/genética , Hormônio Paratireóideo/metabolismo , Proteína Relacionada ao Hormônio Paratireóideo , Proteínas/genética , Proteínas/metabolismo , Receptor Tipo 1 de Hormônio Paratireóideo , Receptores de Hormônios Paratireóideos/genética , Receptores de Hormônios Paratireóideos/metabolismo
16.
Dev Biol ; 203(1): 75-89, 1998 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-9806774

RESUMO

Parathyroid hormone-related protein (PTHrP) was originally identified as the tumor product responsible for humoral hypercalcemia of malignancy. It is now known that PTHrP is produced by many normal tissues in which it appears to play a role as a developmental regulatory molecule. PTHrP is a normal product of mammary epithelial cells, and recent experiments in our laboratory have demonstrated that overexpression or underexpression of PTHrP in the murine mammary gland leads to severe disruptions in its development. The nature of these phenotypes suggests that PTHrP acts to modulate branching growth during mammary development by regulating mammary stromal cell function. We now demonstrate that throughout mammary development, during periods of active ductal-branching morphogenesis, PTHrP is produced by epithelial cells, whereas the PTH/PTHrP receptor is expressed on stromal cells. In addition, we show that mammary stromal cells in culture contain specific binding sites for amino terminal PTHrP and respond with an increase in intracellular cAMP. Finally, we demonstrate that the mammary mesenchyme must express the PTH/PTHrP receptor in order to support mammary epithelial cell morphogenesis. These results demonstrate that PTHrP and the PTH/PTHrP receptor represent an epithelial/mesenchymal signaling circuit that is necessary for mammary morphogenesis and that stromal cells are a critical target for PTHrP's action in the mammary gland.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/genética , Glândulas Mamárias Animais/crescimento & desenvolvimento , Morfogênese/fisiologia , Proteínas/fisiologia , Células Estromais/fisiologia , Animais , Ligação Competitiva , AMP Cíclico/farmacologia , Desenvolvimento Embrionário e Fetal/genética , Epitélio/fisiologia , Feminino , Hibridização In Situ , Mesoderma/fisiologia , Camundongos , Camundongos Endogâmicos , Proteína Relacionada ao Hormônio Paratireóideo , RNA Mensageiro/genética , Receptores de Hormônios Paratireóideos/genética
17.
Proc Natl Acad Sci U S A ; 95(22): 13030-5, 1998 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-9789035

RESUMO

During vertebrate limb development, growth plate chondrocytes undergo temporally and spatially coordinated differentiation that is necessary for proper morphogenesis. Parathyroid hormone-related peptide (PTHrP), its receptor, the PTH/PTHrP receptor, and Indian hedgehog are implicated in the regulation of chondrocyte differentiation, but the specific cellular targets of these molecules and specific cellular interactions involved have not been defined. Here we generated chimeric mice containing both wild-type and PTH/PTHrP receptor (-/-) cells, and analyzed cell-cell interactions in the growth plate in vivo. Abnormal differentiation of mutant cells shows that PTHrP directly signals to the PTH/PTHrP receptor on proliferating chondrocytes to slow their differentiation. The presence of ectopically differentiated mutant chondrocytes activates the Indian hedgehog/PTHrP axis and slows differentiation of wild-type chondrocytes. Moreover, abnormal chondrocyte differentiation affects mineralization of cartilaginous matrix in a non-cell autonomous fashion; matrix mineralization requires a critical mass of adjacent ectopic hypertrophic chondrocytes. Further, ectopic hypertrophic chondrocytes are associated with ectopic bone collars in adjacent perichondrium. Thus, the PTH/PTHrP receptor directly controls the pace and synchrony of chondrocyte differentiation and thereby coordinates development of the growth plate and adjacent bone.


Assuntos
Desenvolvimento Embrionário e Fetal , Lâmina de Crescimento/embriologia , Osteogênese , Proteínas/fisiologia , Receptores de Hormônios Paratireóideos/fisiologia , Transativadores , Animais , Blastocisto/fisiologia , Diferenciação Celular , Quimera , Cruzamentos Genéticos , Indução Embrionária , Feminino , Lâmina de Crescimento/citologia , Proteínas Hedgehog , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Camundongos Transgênicos , Proteína Relacionada ao Hormônio Paratireóideo , Proteínas/genética , Ratos , Receptor Tipo 1 de Hormônio Paratireóideo , Receptores de Hormônios Paratireóideos/deficiência , Receptores de Hormônios Paratireóideos/genética , Células-Tronco , beta-Galactosidase/genética
18.
Recent Prog Horm Res ; 53: 283-301; discussion 301-3, 1998.
Artigo em Inglês | MEDLINE | ID: mdl-9769712

RESUMO

Parathyroid hormone (PTH) and parathyroid hormone-related protein (PTHrP) are two related proteins that activate a common PTH/PTHrP receptor, yet have quite distinct physiologic missions. PTH is the major peptide regulator of blood calcium in higher vertebrates, while PTHrP predominantly acts as a paracrine regulator of differentiation and local intercellular signaling. To analyze the physiological roles of PTHrP and the PTH/PTHrP receptor, "knockout" mice missing either the PTHrP or the PTH/PTHrP receptor gene were developed. Both the PTHrP (-/-) mice and the PTH/PTHrP receptor (-/-) mice exhibit a growth plate chondrodysplasia that reflects accelerated differentiation of proliferating chondrocytes. Growth plate chondrocytes regulate the local production of PTHrP by secreting the protein, Indian hedgehog (Ihh), as they are leaving the proliferative pool. Ihh stimulates the production of PTHrP, which then slows the differentiation of chondrocytes, thereby delaying the production of Ihh. PTHrP also stimulates transport of calcium across the placenta. PTHrP (-/-) mice lack the normal elevation of fetal blood calcium (when compared to maternal levels) and have low placental transport of calcium. Fragments of PTHrP that do not bind to the PTH/PTHrP receptor can correct the defect of placental calcium transport in these mice. Thus, this action of PTHrP is not mediated by the PTH/PTHrP receptor. The "knockout" mice thus help delineate the roles of PTH. PTHrP, and the PTH/PTHrP receptor in an interacting network of ligands and receptors.


Assuntos
Hormônio Paratireóideo/fisiologia , Proteínas/fisiologia , Animais , Ligantes , Camundongos , Proteína Relacionada ao Hormônio Paratireóideo
19.
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
20.
Development ; 125(7): 1285-94, 1998 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-9477327

RESUMO

Parathyroid hormone-related protein (PTHrP) was originally discovered as a tumor product that causes humoral hypercalcemia of malignancy. PTHrP is now known to be widely expressed in normal tissues and growing evidence suggests that it is an important developmental regulatory molecule. We had previously reported that overexpression of PTHrP in the mammary glands of transgenic mice impaired branching morphogenesis during sexual maturity and early pregnancy. We now demonstrate that PTHrP plays a critical role in the epithelial-mesenchymal communications that guide the initial round of branching morphogenesis that occurs during the embryonic development of the mammary gland. We have rescued the PTHrP-knockout mice from neonatal death by transgenic expression of PTHrP targeted to chondrocytes. These rescued mice are devoid of mammary epithelial ducts. We show that disruption of the PTHrP gene leads to a failure of the initial round of branching growth that is responsible for transforming the mammary bud into the rudimentary mammary duct system. In the absence of PTHrP, the mammary epithelial cells degenerate and disappear. The ability of PTHrP to support embryonic mammary development is a function of amino-terminal PTHrP, acting via the PTH/PTHrP receptor, for ablation of the PTH/PTHrP receptor gene recapitulates the phenotype of PTHrP gene ablation. We have localized PTHrP expression to the embryonic mammary epithelial cells and PTH/PTHrP receptor expression to the mammary mesenchyme using in situ hybridization histochemistry. Finally, we have rescued mammary gland development in PTHrP-null animals by transgenic expression of PTHrP in embryonic mammary epithelial cells. We conclude that PTHrP is a critical epithelial signal received by the mammary mesenchyme and involved in supporting the initiation of branching morphogenesis.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/genética , Glândulas Mamárias Animais/crescimento & desenvolvimento , Proteínas/genética , Animais , Células Cultivadas , Deleção de Genes , Técnicas de Transferência de Genes , Imuno-Histoquímica , Hibridização In Situ , Glândulas Mamárias Animais/embriologia , Glândulas Mamárias Animais/patologia , Camundongos , Camundongos Knockout , Morfogênese/fisiologia , Proteína Relacionada ao Hormônio Paratireóideo , Fenótipo , Proteínas/fisiologia , RNA Mensageiro/análise , Receptores de Hormônios Paratireóideos/genética , Receptores de Hormônios Paratireóideos/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA