Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Calcif Tissue Int ; 78(3): 162-70, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16525748

RESUMO

Approximately 50% of neurofibromatosis type 1 (NF1) patients exhibit skeletal pathology, such as premature osteoporosis or pseudoarthroses. Loss of neurofibromin deregulates Ras signal transduction to affect generation of mitogen-activated protein kinase and Akt, both of which have been implicated in parathyroid hormone (PTH) anabolic mechanisms. Our aim was to determine if loss of neurofibromin impaired the anabolic effect of PTH on bone mass. Nf1 heterozygote (Nf1(+/-)) and wild type (Nf1(+/+)) mice were treated with recombinant human PTH(1-34) or vehicle once daily for 3-28 days. PTH enhanced mRNA expression of c-fos, junB, and fra2 in the distal femur metaphyses of both genotypes; expression of these transcripts was consistently lower in PTH-treated Nf1(+/-) mice. Despite lowered c-fos expression in Nf1(+/-) mice, PTH increased bone mass equivalently in both genotypes by 28 days. Ex vivo, Nf1 heterozygosity was associated with increased inducible osteoclasts in PTH-treated bone marrow cells and impairment of the actin stress fiber and cyclic adenosine monophosphate response to PTH in osteoprogenitors. Lower c-fos expression was previously thought to abrogate PTH responsiveness. Our results suggest crosstalk might occur between Ras signal transduction and the protein kinase A pathway in Nf1(+/-) mice. Ras signal transduction does not appear to be essential for the anabolic actions of PTH on bone. Because PTH was effective in the absence of Nf1, it may offer a useful approach to treat osteoporosis in NF1 patients.


Assuntos
Expressão Gênica , Genes da Neurofibromatose 1 , Hormônio Paratireóideo/metabolismo , Hormônio Paratireóideo/farmacologia , Fator de Transcrição AP-1/genética , Fosfatase Ácida/metabolismo , Animais , Densidade Óssea/efeitos dos fármacos , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Células Cultivadas , AMP Cíclico/análise , Fêmur/metabolismo , Fêmur/cirurgia , Humanos , Isoenzimas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Hormônio Paratireóideo/genética , RNA Mensageiro/metabolismo , Proteínas Recombinantes/farmacologia , Ribonucleases/biossíntese , Fosfatase Ácida Resistente a Tartarato , Tíbia/metabolismo , Tomografia Computadorizada por Raios X
2.
Bone ; 36(5): 793-802, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15804420

RESUMO

Skeletal problems and osteoporosis occur in up to 50% affected neurofibromatosis type 1 (NF1) humans. Inactivation of neurofibromin results in deregulation of Ras signal transduction. Little is known of bone biology in humans with NF1. The goal of our work was to determine if loss-of-function of Nf1 gene was associated with altered bone homeostasis and Ras signal transduction. Because homozygous Nf1 mice are embryonically lethal, heterozygote Nf1 (Nf1+/-) male mice were used to investigate skeletal phenotypes and osteoprogenitor functions, using standard in vivo and in vitro assays. We found that bone mass and geometry of Nf1+/- mice did not differ from wild type controls, despite a trend to less bone formation. Nf1+/- committed osteoprogenitors from femur metaphysis exhibited premature apoptosis and higher proliferation. Ras signaling was activated in primary Nf1+/- bone marrow-inducible osteoprogenitors. Inducible osteoprogenitors exhibited lower induction of osteoblast differentiation, assessed as alkaline phosphatase positive CFU-f. A screen of osteoblast marker genes showed a selective increase in osteopontin (OPN) mRNA and protein expression in these cells. OPN protein was increased in Nf1+/- bone, especially in cortical bone matrix. Because bone cell abnormalities in Nf1 haploinsufficiency were detected in vitro, redundant pathways must compensate for the deregulation of Ras signaling in vivo to maintain normal bone mass and function in vivo. Our in vitro data revealed that neurofibromin and its control of Ras signaling are required for osteoprogenitor homeostasis.


Assuntos
Neurofibromina 1/fisiologia , Proteína Oncogênica p21(ras)/antagonistas & inibidores , Osteoblastos/citologia , Animais , Western Blotting , Divisão Celular , Humanos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Knockout , Neurofibromina 1/genética , Osteopontina , Fenótipo , Sialoglicoproteínas/genética , Sialoglicoproteínas/fisiologia
3.
J Pharmacol Exp Ther ; 302(1): 304-13, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12065731

RESUMO

We report the consequences of prolonged treatment with recombinant human parathyroid hormone (1-34) (PTH) in male and ovariectomized female rats with mature skeletons. Intact male and osteopenic, ovariectomized, female F-344 rats were evaluated after 1 year of treatment with 0, 8, or 40 microg/kg/day s.c. PTH. Males and females were about 6 months of age at study initiation; females were ovariectomized (Ovx) for 5 weeks before initiation of PTH treatment. PTH did not affect the survival of either intact males or ovariectomized females. Qualitative histopathology showed expected changes associated with aging in kidneys and proximal tibiae, with no treatment-related anomalies after 1 year of PTH administration. PTH slightly increased the femoral length of ovariectomized females but not that of males. No significant differences in femoral length were observed between sham and Ovx controls. Proximal femora of the males and ovariectomized females given the high dose of 40 microg/kg showed 211 and 186% greater trabecular bone area, 118 and 94% greater cortical thickness, 170 and 189% greater trabecular number, and 321 and 404% greater connectivity (node-to-node struts) compared with respective vehicle controls. Increased trabecular and endocortical surface apposition coincided with a 78 and 70% loss of marrow space for males and females treated with PTH, respectively. Biomechanical strength (ultimate load) of the femoral neck increased by 73 and 76%, respectively, in males and ovariectomized females. Cortical bone analyses of the femoral midshaft showed 105 and 72% increases in bone mineral content, 67 and 55% increases in bone mineral density, and 22 and 10% increases in cross-sectional area for males and ovariectomized females, respectively, with altered shape of femora. Biomechanical analyses of the midshaft showed substantial increases in strength and stiffness but a reduction in ultimate strain, which was likely due to the altered geometry of the midshaft for PTH groups. Aging effects on strength of vertebra and femoral midshaft were reversed by PTH treatment. In summary, the 1-year treatment duration, which represents about 50% of lifetime, did not affect survival and was not associated with any treatment-related anomalies in the kidney or skeleton. PTH reversed the aging process in bones but not kidneys and substantially increased bone mass and strength to well beyond normally attained levels. However, compared with short-term studies reported previously, there seemed to be no advantages to extending PTH treatment to 12 months in rat bones.


Assuntos
Osso e Ossos/efeitos dos fármacos , Hormônio Paratireóideo/farmacologia , Absorciometria de Fóton , Animais , Fenômenos Biomecânicos , Peso Corporal/efeitos dos fármacos , Densidade Óssea/efeitos dos fármacos , Osso e Ossos/patologia , Feminino , Fêmur/efeitos dos fármacos , Fêmur/patologia , Rim/efeitos dos fármacos , Rim/patologia , Masculino , Ovariectomia , Hormônio Paratireóideo/sangue , Ratos , Ratos Endogâmicos F344 , Tíbia/patologia
4.
J Cell Biochem ; 85(2): 229-42, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-11948679

RESUMO

Parathyroid hormone (PTH) stimulates bone formation in both animals and humans, and the expression of a number of genes has been implicated in the mediation of this effect. To discover new bone factors that initiate and support this phenomenon, we used differential display reverse transcription polymerase chain reaction (DDRT-PCR) and screened for genes, which are differentially expressed in osteoblast-enriched femoral metaphyseal primary spongiosa of young male rats after a single subcutaneous (s.c.) injection of hPTH (1-38) (8 microg/100 g). We found and cloned one full-length cDNA, which encodes a putative 348 amino acid protein. Sequence analysis of this protein demonstrates a 98, 93.7, and 82.5% identity with mouse, human, and chicken ubiquitin-specific protease UBP41, respectively. Northern blot analysis confirmed that a 3.8-4 kb UBP41 mRNA transcript was rapidly increased 1 h after acute hPTH (1-38) exposure in both metaphyseal (6- to 8-fold) and diaphyseal (3-fold) bone, but returned to control levels by 24 h after exposure. In contrast, continuous exposure to hPTH (1-38), resulted in a rapid and sustained elevation of UBP41 mRNA. PTH (1-31), which stimulates intracellular cAMP, and PTHrP (1-34) both induced UBP41 mRNA expression; whereas PTH analogs (3-34) and (7-34), that do not stimulate cAMP, had no effect on UBP41 expression. UBP41 mRNA expression was also rapidly induced 1 h after injection of PGE2, but returned to the control level by 6 to 24 h. In vitro, UBP41 mRNA is expressed in primary osteoblasts (metaphyseal and diaphyseal derived) and in the osteoblast-like cell lines UMR106, ROS17/2.8, and BALC. PTH (1-38) treatment induced UPB41 expression (3.6- to 13-fold) in both primary cultures of osteoblasts and in UMR106 cells. Further analysis in UMR 106 cells demonstrated that PGE2, forskolin and dibutyryl cAMP increased UBP41 mRNA expression 4-, 4.5-, and 2.4-fold, respectively. Tissue distribution analysis of UBP41 mRNA detected transcripts in brain, heart, skeletal muscle, kidney, liver, and testis. Together, these results demonstrate that UBP41, an ubiquitin-specific protease, is selectively upregulated in bone by the osteotropic agents PTH, PTHrP, and PGE2, possibly via the PKA/cAMP pathway. We speculate that the rapid induction of UBP41 in response to these physiological regulators contributes to the mechanism by which either the structure, activity, half-life or localization of essential proteins are modified to maintain bone homeostasis.


Assuntos
Osso e Ossos/efeitos dos fármacos , Endopeptidases/biossíntese , Hormônio Paratireóideo/farmacologia , Fragmentos de Peptídeos/farmacologia , Ubiquitina/metabolismo , Animais , Northern Blotting , Osso e Ossos/metabolismo , Células Cultivadas , Primers do DNA/química , Endopeptidases/genética , Fêmur/metabolismo , Perfilação da Expressão Gênica , Biblioteca Gênica , Masculino , Osteoblastos/metabolismo , Reação em Cadeia da Polimerase , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Ubiquitina Tiolesterase , Regulação para Cima
5.
J Musculoskelet Neuronal Interact ; 2(6): 546-8, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15758390
6.
J Musculoskelet Neuronal Interact ; 2(4): 321-6, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15758423

RESUMO

Human parathyroid hormone (hPTH 1-34) stimulates an anabolic response in human and animal skeletons; however, it is unclear if the effect is strain dependent. To determine if the anabolic response to hPTH (1-34) was dependent upon strain in rats we used 2 outbred strains (Sprague Dawley, Wistar), 2 inbred strains (Fischer 344, Wistar spontaneously hypertensive:SHR), and 2 mutant strains (Zucker obese, Zucker lean) of rats. Male rats, 5 weeks of age, from each strain were treated subcutaneously with 80 microg/kg body weight hPTH (1-34) or vehicle for 12 days. The response to PTH was similar in all strains whereby PTH exerted an anabolic effect on femoral bone mass and cancellous bone histology that was independent of strain differences. Histomorphometric indices of bone volume, mineralized surface and bone formation in lumbar vertebrae increased in all PTH-treated rats. Additionally, femur bone mineral content and bone mineral density measured by dual energy X-ray absorptiometry (DEXA), and ash weight increased in all PTH-treated rats. These increases occurred regardless of strain. In summary, PTH exerted comparable anabolic effects on bone mass, bone mineral density and bone formation in all rat models tested demonstrating that the skeletal responsiveness to PTH was not dependent upon strain.

7.
J Bone Miner Res ; 16(6): 975-84, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11393794

RESUMO

With the discoveries of different death mechanisms, an emerging definition of apoptosis is the process of cell death associated with caspase activation or caspase-mediated cell death. This definition accepts that caspases represent the final common mechanistic pathway in apoptosis. Apoptosis may be triggered either by activation events that target mitochondria or endoplasmic reticulum or by activation of cell surface "death receptors," for example, those in the tumor necrosis factor (TNF) superfamily. In the postnatal and adult skeleton, apoptosis is integral to physiological bone turnover, repair, and regeneration. The balance of osteoblast proliferation, differentiation, and apoptosis determines the size of the osteoblast population at any given time. Although apoptosis has been recorded in many studies of bone, the selective mechanisms invoked in the different models studied rarely have been identified. This review offers a broad overview of the current general concepts and controversies in apoptosis research and then considers specific examples of osteoblast apoptosis pertinent to skeletal development and to the regulation of bone turnover. In reviewing selected work on interdigital apoptosis in the developing skeleton, we discuss the putative roles of the bone morphogenetic proteins (BMPs), Msx2, RAR-gamma, and death inducer obliterator 1 (DIO-1). In reviewing factors regulating apoptosis in the postnatal skeleton, we discuss roles of cytokines, growth factors, members of the TNF pathway, and the extracellular matrix (ECM). Finally, the paradoxical effects of parathyroid hormone (PTH) on osteoblast apoptosis in vivo are considered in the perspective of a recent hypothesis speculating that this may be a key mechanism to explain the anabolic effects of the hormone. An improved understanding of the apoptotic pathways and their functional outcomes in bone turnover and fracture healing may facilitate development of more targeted therapeutics to control bone balance in patients with osteoporosis and other skeletal diseases.


Assuntos
Apoptose/fisiologia , Remodelação Óssea/fisiologia , Osteoblastos/patologia , Animais , Caspases/metabolismo , Matriz Extracelular/fisiologia , Humanos , Osteoblastos/metabolismo , Hormônio Paratireóideo/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
8.
Bone ; 28(5): 538-47, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11344054

RESUMO

We have previously shown that parathyroid hormone (PTH) increases cortical bone mass and mechanical strength of female rabbits after 140 days of treatment. However, cortical porosity was also shown to increase. If cortical porosity increases prior to the change in geometry, there may be a transient decrease in cortical bone strength that could make the bone more susceptible to fracture in the early phase of treatment. The purpose of this study is to examine the effects of PTH on the remodeling dynamics and mechanical properties of cortical bone in rabbits, which exhibit haversian remodeling, during the first remodeling cycle after the initiation of treatment. Fifty 9-month-old intact female New Zealand white rabbits were randomized into five groups. A baseline control group was killed at the start of the experiment. The two PTH-treated groups were given human PTH(1-34) at 10 microg/kg daily subcutaneously for 35 (P35) or 70 (P70) days. Two respective age-matched control groups (V35, V70) were injected with vehicle. Histomorphometry of the cortical bone in the tibial midshaft showed that, although intracortical activation frequency was significantly increased by PTH at 35 days, there was no significant increase of cortical porosity in the first remodeling cycle (70 days). Moreover, stimulation of cortical surface bone formation in the treated animals led to significantly greater cortical area and greater bone strength in both P35 and P70. We conclude that, although intracortical remodeling increases within the first remodeling period (70 days) in animals treated with 10 microg/kg PTH, the greater cortical area due to acceleration of bone formation on cortical surfaces increases cortical bone strength. There is no mechanical risk during the first remodeling cycle associated with intermittent PTH treatment in animals with normal bone mass.


Assuntos
Remodelação Óssea/efeitos dos fármacos , Osso e Ossos/efeitos dos fármacos , Tamanho do Órgão/efeitos dos fármacos , Teriparatida/farmacologia , Fosfatase Alcalina/sangue , Animais , Fenômenos Biomecânicos , Nitrogênio da Ureia Sanguínea , Densidade Óssea/efeitos dos fármacos , Densidade Óssea/fisiologia , Remodelação Óssea/fisiologia , Osso e Ossos/citologia , Osso e Ossos/metabolismo , Cálcio/sangue , Feminino , Tamanho do Órgão/fisiologia , Osteoporose/tratamento farmacológico , Osteoporose/metabolismo , Osteoporose/fisiopatologia , Fosfatos/sangue , Coelhos , Teriparatida/metabolismo
9.
Bone ; 28(2): 150-9, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11182372

RESUMO

A key feature of postmenopausal osteoporosis is the loss of trabecular bone mass and connectivity. The current study focuses on these parameters in the assessment of long-term (12 and 18 months) parathyroid hormone (PTH) therapy and its withdrawal (6 months) in the ovariectomized cynomolgus monkey (Macaca fascicularis), a well-characterized model for bone changes associated with postmenopausal osteoporosis. We used static and dynamic histomorphometric parameters to assess the amount and architecture of cancellous bone in four clinically important sites for osteoporotic fractures, including the lumbar vertebra, femoral neck, distal radius, and iliac crest. Recombinant human PTH(1-34) was administered daily to two groups for 18 months at 1.0 microg/kg per day (n = 19) and 5.0 microg/kg per day (n = 21). To study the effects of PTH withdrawal, two groups were administered PTH(1-34) daily for 12 months at 1.0 microg/kg per day (n = 20) and 5.0 microg/kg per day (n = 20), followed by daily administration of vehicle for 6 months. Sham-ovariectomized and ovariectomized (ovx) groups each received daily injections of vehicle for 18 months. Treatment with PTH had minimal effects on bone formation rates at the timepoints studied, but markedly increased cancellous bone volume relative to ovx monkeys in iliac crest biopsies at 6 and 15 months, as well as in terminal specimens of lumbar vertebrae, femoral neck, and distal radius after 18 months. At all sites, PTH significantly improved trabecular architecture, as evidenced by increased trabecular number (Tb.N) and decreased trabecular separation (Tb.Sp), with no significant change in trabecular thickness (Tb.Th). The mechanism of these structural changes is suggested by qualitative observations of trabecular tunneling observed in the iliac crest and vertebra. Longitudinal tunneling of thickened individual trabeculae is hypothesized to convert them into multiple trabeculae, resulting in a normalization of Tb.Th, but an increase in Tb.N. A significant positive effect on cancellous bone volume was still apparent after a 3-6 month withdrawal period following 12 months of PTH treatment in the iliac crest, vertebra, and femoral neck. Corresponding increases in Tb.N and decreases in Tb.Sp also remained significant after PTH withdrawal at these three sites. The distal radius was relatively insensitive to PTH treatment or its withdrawal, compared with the other bones. In summary, PTH therapy dramatically improved cancellous bone mass and architecture in both axial and appendicular sites.


Assuntos
Osteoporose/tratamento farmacológico , Ovariectomia , Hormônio Paratireóideo/farmacologia , Fragmentos de Peptídeos/farmacologia , Absorciometria de Fóton , Animais , Biópsia , Remodelação Óssea/efeitos dos fármacos , Remodelação Óssea/fisiologia , Feminino , Colo do Fêmur/patologia , Ílio/patologia , Vértebras Lombares/patologia , Macaca fascicularis , Osteoporose/diagnóstico por imagem , Rádio (Anatomia)/patologia
10.
J Bone Miner Res ; 16(1): 157-65, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11149480

RESUMO

Cortical porosity in patients with hyperparathyroidism has raised the concern that intermittent parathyroid hormone (PTH) given to treat osteoporotic patients may weaken cortical bone by increasing its porosity. We hypothesized that treatment of ovariectomized (OVX) cynomolgus monkeys for up to 18 months with recombinant human PTH(1-34) [hPTH(1-34)] LY333334 would significantly increase porosity in the midshaft of the humerus but would not have a significant effect on the strength or stiffness of the humerus. We also hypothesized that withdrawal of PTH for 6 months after a 12-month treatment period would return porosity to control OVX values. OVX female cynomolgus monkeys were given once daily subcutaneous (sc) injections of recombinant hPTH(1-34) LY333334 at 1.0 microg/kg (PTH1), 5.0 microg/kg (PTH5), or 0.1 ml/kg per day of phosphate-buffered saline (OVX). Sham OVX animals (sham) were also given vehicle. After 12 months, PTH treatment was withdrawn from half of the monkeys in each treatment group (PTH1-W and PTH5-W), and they were treated for the remaining 6 months with vehicle. Double calcein labels were given before death at 18 months. After death, static and dynamic histomorphometric measurements were made intracortically and on periosteal and endocortical surfaces of sections from the middiaphysis of the left humerus. Bone mechanical properties were measured in the right humeral middiaphysis. PTH dose dependently increased intracortical porosity. However, the increased porosity did not have a significant detrimental effect on the mechanical properties of the bone. Most porosity was concentrated near the endocortical surface where its mechanical effect is small. In PTH5 monkeys, cortical area (Ct.Ar) and cortical thickness (Ct.Th) increased because of a significantly increased endocortical mineralizing surface. After withdrawal of treatment, porosity in PTH1-W animals declined to sham values, but porosity in PTH5-W animals remained significantly elevated compared with OVX and sham. We conclude that intermittently administered PTH(1-34) increases intracortical porosity in a dose-dependent manner but does not reduce the strength or stiffness of cortical bone.


Assuntos
Densidade Óssea/efeitos dos fármacos , Remodelação Óssea/efeitos dos fármacos , Úmero/efeitos dos fármacos , Macaca fascicularis/fisiologia , Ovariectomia , Teriparatida/administração & dosagem , Teriparatida/farmacologia , Animais , Peso Corporal/efeitos dos fármacos , Feminino , Úmero/metabolismo , Úmero/fisiologia , Injeções Subcutâneas , Macaca fascicularis/metabolismo , Porosidade/efeitos dos fármacos , Resistência à Tração/efeitos dos fármacos
11.
J Musculoskelet Neuronal Interact ; 2(1): 33-47, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15758475

RESUMO

A brief historical perspective reviews studies that tested the hypotheses that PTH induces an anabolic effect in bone, and that the gain in trabecular bone was not at the expense of cortical bone. As PTH reduces the risk of fracture in humans with osteoporosis, the myths that postulated cortical bone porosity and increased bone turnover might increase fracture risk, are examined in the light of data from animals with osteonal bone. These show that PTH "braces" the bone by immediately stimulating bone formation at modeling and remodeling sites. Increased porosity is a late event, occurring close to the neutral axis of bone where detrimental effects on biomechanical strength are unlikely. PTH increases bone mass by stimulating modeling in favor of bone formation, and restructures bone geometry via more extensive remodeling. Cell and genetic events induced in bone by PTH have been studied in rats and are time- and regimen-dependent. In addition to the stimulation of gene expression for matrix proteins, early genes upregulated by once daily PTH are those associated with matrix degradation and induction of osteoclastic resorption, indicative of possible mechanisms by which PTH may increase bone turnover. Boneforming surfaces are increased due to increased numbers of newly differentiated osteoblasts and retention of older osteoblasts by inhibition of apoptosis. After stopping treatment, the number of osteoblasts is quickly reduced and bone turnover returns to that of controls, slowing both bone formation and resorption. The increased proportion of bone undergoing PTH-induced remodeling requires maturation and completion of mineralization. These responses may explain the delay in reversal of gains in bone mass and biomechanical properties for at least two turnover cycles following withdrawal in large animal models. Thus, the skeletal benefits of PTH extend beyond the active treatment phase.

12.
Crit Rev Eukaryot Gene Expr ; 11(4): 279-97, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-12067068

RESUMO

The functional role of the osteoblast nuclear matrix has been a matter of supposition. Its presumed function as an architectural agent of transcription derives primarily from the low solubility of nuclear matrix proteins and their typical localization into discrete subnuclear domains. In addressing how the nuclear matrix regulates skeletal genes, the authors compare Nmp4, Cbfal, and YY1 for the purpose of profiling osteoblast nuclear matrix transcription factors. All three proteins contribute to the transcription of ECM genes and partition into the osteoblast nuclear matrix via a nuclear matrix targeting domain. The authors propose that osteoblast nuclear matrix transcription factors involved in ECM regulation generally have the capacity to alter DNA geometry and reciprocally respond to DNA as an allosteric ligand. This may allow these proteins to adapt to the local nuclear architecture and generate the pattern of regulation specified by that architecture via unmasking of the appropriate transactivation domains. Osteoblast nuclear matrix transcription factors may also act as transcriptional adaptor molecules by supporting the formation of higher order protein complexes along target gene promoters. The genes encoding all three proteins considered here have trinucleotide repeat domains, although the significance of this is unclear. There is no canonical nuclear matrix binding motif, but finger-like structures may be suited for anchoring proteins to discrete subnuclear domains. Finally, the ability to leave the osteoblast nuclear matrix may be as important to the function of some nuclear matrix transcription factors as their association with this subcompartment.


Assuntos
Osso e Ossos/fisiologia , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica , Proteínas Associadas à Matriz Nuclear , Matriz Nuclear/fisiologia , Proteínas Nucleares/genética , Transativadores/genética , Fatores de Transcrição/genética , Animais , Antígenos Nucleares , Sequência de Bases , Sequência Consenso , Fatores de Ligação de DNA Eritroide Específicos , Humanos , Camundongos , Dados de Sequência Molecular , Isoformas de Proteínas/genética , Ratos , Fator de Transcrição YY1
14.
Bone ; 27(6): 819-26, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11113393

RESUMO

Intermittent parathyroid hormone (PTH) treatment increases bone mass in humans and animals. Although intact human PTH has 84 amino acids, the N-terminal 31 to 38 amino acids are sufficient for bone anabolic activity in vivo. Prior studies have evaluated hPTH(1-34) and hPTH(1-84) with respect to bone mass increase and quality, but there have been no in vivo comparisons of dose-dependent molecular responses. After confirming that young male BALB/c mice respond to daily PTH with increased bone mass, we profiled the steady-state mRNA levels of activating protein-1 (AP-1) genes regulated by hPTH(1-34) and hPTH(1-84) at doses ranging from 0 to 19.4 nmol/kg in the distal femur metaphyses. We selected AP-1 genes, which include jun and fos, as they play a fundamental role mediating signals for proliferation, differentiation, and apoptosis in cells of different origins, including bone, and are known to be regulated by PTH. Human PTH(1-34) and hPTH(1-84) increased steady-state mRNA expression of c-jun, junB, c-fos, and fra-2 in an equivalent dose- and time-dependent manner. Expression of fosB or fra-1 was not detected with either peptide. When averaged across dose and time, responses to hPTH(1-34) and hPTH(1-84) were not significantly different from each other. Expression of c-jun, junB, and c-fos peaked 30 minutes after the injection while fra-2 expression peaked 30 minutes later. All AP-1 genes stimulated by PTH returned to the levels of vehicle treated controls by 3 h after injection. The expression level of junD, which was abundant in the distal metaphysis, was not altered by either peptide. No change in magnitude was observed after 1, 3, or 7 days of once-daily subcutaneous treatment of either peptide. When individual comparisons for each dose between peptides were made, the minimum effective dose necessary to stimulate a significant increase in c-fos and junB expression was equivalent for both peptides. The minimum effective dose for hPTH(1-34) was at least tenfold lower than hPTH(1-84) in stimulating c-jun and fra-2 expression. Area under the curve for the highest dose (19.4 nmol/kg) of either peptide showed no significant differences in the expression of any of the genes. In conclusion, in young mice given once-daily subcutaneous injections up to 7 days, hPTH(1-34) and hPTH(1-84) induced equivalent responses by time and dose in the selected AP-1 genes. These data on molecular regulation in mouse bone confirm and extend prior data from rat studies showing equivalence on bone mass at equimolar doses.


Assuntos
Fêmur/efeitos dos fármacos , Hormônio Paratireóideo/farmacologia , Fragmentos de Peptídeos/farmacologia , Fator de Transcrição AP-1/genética , Fatores Etários , Animais , Relação Dose-Resposta a Droga , Fêmur/citologia , Fêmur/metabolismo , Expressão Gênica/efeitos dos fármacos , Genes fos/fisiologia , Genes jun/fisiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Tamanho do Órgão , Osteoblastos/efeitos dos fármacos , Osteoblastos/fisiologia , Proteínas Proto-Oncogênicas c-fos/genética , RNA Mensageiro/análise , Fatores de Tempo
15.
Endocrinology ; 141(12): 4533-42, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11108265

RESUMO

PTH stimulates bone formation in animals and humans, and the expressions of a number of genes have been implicated in the mediation of this effect. To discover new bone factors that initiate and support this phenomenon we used differential display RT-PCR and screened for genes that are selectively expressed in osteoblast-enriched femoral metaphyseal primary spongiosa of young male rats after a single s.c. injection of human PTH-(1-38) (8 microg/100 g). We show that one of the messenger RNAs that is up-regulated in bone is ADAMTS-1, a new member of the ADAM (A disintegrin and metalloprotease) gene family containing thrombospondin type I motifs. ADAMTS-1 consists of multiple domains common to ADAM family of proteins, including pro-, metalloprotease-like, and disintegrin-like domains. However, unlike other ADAMs, ADAMTS-1 does not possess a transmembrane or cytoplasmic domain and is a secreted protein. Northern blot analysis confirmed that ADAMTS-1 was up-regulated in both metaphyseal (14- to 35-fold) and diaphyseal (4.2-fold) bone 1 h after PTH-(1-38) injection and returned to control levels by 24 h. We also analyzed the regulation of ADAMTS-1 in response to various PTH/PTH-related peptide (PTHrP) analogs and found that PTH-(1-31) and PTHrP-(1-34), which activate the protein kinase A (PKA) pathway, induce ADAMTS-1 expression 1 h after injection, whereas PTH-(3-34) and PTH-(7-34), which do not activate the PKA pathway, did not regulate expression. To investigate the effect of other osteotropic agents, we analyzed ADAMTS-1 expression after a single dose of PGE2 (6 mg/kg) and found that it was up-regulated 1 h after injection and returned to control levels by 6 h. In vitro ADAMTS-1 is expressed in primary osteoblasts and osteoblastic cell lines, but was not detectable in osteoclasts generated from macrophage colony-stimulating factor/receptor activator of NF-kappaB ligand/transforming growth factor-beta1-treated bone marrow cells. Treatment of UMR 106 osteosarcoma cells with PTH, PGE2, forskolin, or (Bu)2cAMP increased ADAMTS-1 expression 7-, 4-, 5-, and 5-fold, respectively. Also, in vitro treatment with 1alpha,25-dihydroxyvitamin D3 increased ADAMTS-1 expression 3-fold. Tissue distribution analysis showed that ADAMTS-1 is expressed at high levels in many tissues, including the heart, lung, liver, skeletal muscle, and kidney. Taken together, these results demonstrate that ADAMTS-1 is specifically up-regulated in bone and osteoblasts by the osteotropic agents PTH, PTHrP, and PGE2 possibly via the cAMP/PKA pathway. We speculate that the rapid and transient increase in ADAMTS-1 expression may contribute to some of the effects of PTH on bone turnover.


Assuntos
Osso e Ossos/enzimologia , Desintegrinas/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Metaloendopeptidases/genética , Hormônio Paratireóideo/farmacologia , Proteínas ADAM , Proteína ADAMTS1 , Animais , Calcitriol/farmacologia , Bovinos , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Dinoprostona/farmacologia , Desintegrinas/metabolismo , Ativação Enzimática/efeitos dos fármacos , Fêmur , Humanos , Cinética , Masculino , Metaloendopeptidases/metabolismo , Especificidade de Órgãos , Osteoblastos/enzimologia , Osteoclastos/enzimologia , Osteossarcoma/enzimologia , Proteína Relacionada ao Hormônio Paratireóideo , Fragmentos de Peptídeos/farmacologia , Proteínas/farmacologia , RNA Mensageiro/análise , Ratos , Ratos Endogâmicos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas
17.
J Bone Miner Res ; 15(8): 1517-25, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10934650

RESUMO

Daily administration of parathyroid hormone (PTH) and PTH-related protein (PTHrP) peptides has been shown to increase bone mass and strength in animals and, for PTH, to increase bone mass in humans. Long-term direct comparison of multiple members of the PTH/PTHrP family in vivo has not been reported. We therefore selected three PTH/PTHrP molecules for direct comparison in vivo in an adult rat model of postmenopausal osteoporosis: PTH(1-34), PTHrP(1-36), and the PTH analog, SDZ-PTH 893 ¿Leu8, Asp10, Lys11, Ala16, Gln18, Thr33, Ala34 human PTH 1-34 [hPTH(1-34)]¿. A 6-month study was performed in which adult (6-month-old) vehicle-treated ovariectomized (OVX) and sham OVX rats were compared with OVX rats receiving 40 micrograms/kg per day of either PTH(1-34), PTHrP(1-36), or PTH-SDZ-893. Bone mass, as assessed by ash weight and densitometry, bone histomorphometry, biomechanical properties at trabecular and cortical sites, and indices of bone formation markedly increased in all three PTH/PTHrP peptide-treated groups as compared with controls. In general, this improvement followed a rank order of SDZ-PTH-893 > PTH > PTHrP. The adverse effect profile also was greatest with SDZ-PTH-893; these rats developed moderate hypercalcemia, marked renal calcium accumulation, and displayed a 13% mortality. These studies show that PTH(1-34), PTHrP(1-36), and PTH-SDZ-893 significantly and progressively increase bone mass and bone strength in this rat model of postmenopausal osteoporosis. The adverse effect profile correlates in general terms with efficacy. All three peptides show promise as skeletal anabolic agents. Further studies in humans will be required to define optimal efficacy-to-adverse effect ratios and relative efficacy for each peptide in human osteoporosis.


Assuntos
Ovariectomia/efeitos adversos , Proteína Relacionada ao Hormônio Paratireóideo , Hormônio Paratireóideo/farmacologia , Fragmentos de Peptídeos/farmacologia , Proteínas/farmacologia , Teriparatida/análogos & derivados , Animais , Peso Corporal/efeitos dos fármacos , Densidade Óssea , Cálcio/sangue , Cálcio/urina , Feminino , Fêmur/efeitos dos fármacos , Fêmur/patologia , Humanos , Vértebras Lombares/efeitos dos fármacos , Vértebras Lombares/patologia , Hormônio Paratireóideo/administração & dosagem , Fragmentos de Peptídeos/administração & dosagem , Proteínas/administração & dosagem , Ratos , Ratos Sprague-Dawley , Teriparatida/administração & dosagem , Teriparatida/farmacologia , Fatores de Tempo
18.
Bone ; 27(2): 209-18, 2000 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-10913913

RESUMO

Osteoblast differentiation and function can be studied in situ in the metaphysis of growing long bones. Proliferation and apoptosis dominate in the primary spongiosa subjacent to the growth plate, and differentiation and function dominate in the proximal metaphysis. Apoptosis of osteocytes dominates at the termination of the trabeculae in diaphyseal marrow. As parathyroid hormone regulates all phases of osteoblast development, we studied the in vivo regulation by human parathyroid hormone (1-34) (PTH) of apoptosis in bone cells of the distal metaphysis of young male rats. Rats were given PTH at 80 microg/kg per day, once daily, for 1-28 days. Bone cells were defined for flow cytometry as PTH1-receptor-positive (PTH1R(+)) and growth factor-receptor-positive (GFR(+)) cells. Apoptotic cells stained positive for either TdT-mediated dUTP-X nick end labeling (TUNEL) or annexin V (annV(+)) were detected by either flow cytometry or immunohistochemistry. Apoptosis was also assessed at the tissue level by RNAse protection and caspase enzyme activity assays. PTH increased apoptotic osteoblasts in the proliferating zone and apoptotic osteocytes in the terminal trabecular zone, by 40%-60% within 2-6 days of PTH treatment, but values became equivalent to controls after 21-28 days of treatment. This transient increase was confirmed in PTH1R(+), GFR(+) bone cells isolated by flow cytometry. There was no detectable change in the steady-state mRNA levels of selected apoptotic genes. Starting at 3 days, at the tissue level, PTH inhibited activity of caspases, which recognize the DEVD peptide substrate (caspases 2, 3, and/or 7), but not those caspases recognizing LEHD or YVAD peptide sequences. We speculate that the localized and tissue level effects of PTH on apoptosis can be explained on the basis of its anabolic effect on bone. The transient increase in apoptosis in the proliferating zone and terminal trabecular zone may be the result of the increased activation frequency and bone turnover seen with daily PTH treatment. As once-daily PTH increases the number of differentiated osteoblasts, and as these and hematopoietic marrow cells dominate metaphyseal tissue, inhibition of caspase activity may contribute to their prolonged survival, enabling extension of trabecular bone into the diaphyseal marrow to increase bone mass.


Assuntos
Apoptose/efeitos dos fármacos , Fêmur/citologia , Osteócitos/citologia , Hormônio Paratireóideo/farmacologia , Fragmentos de Peptídeos/farmacologia , Fatores Etários , Animais , Anexina A5/análise , Caspases/metabolismo , Divisão Celular/efeitos dos fármacos , Diáfises/citologia , Citometria de Fluxo , Expressão Gênica/fisiologia , Humanos , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Masculino , Osteócitos/química , Osteócitos/enzimologia , Proteínas Proto-Oncogênicas c-bcl-2/genética , RNA Mensageiro/análise , Ratos , Ratos Endogâmicos F344 , Ratos Sprague-Dawley , Receptores Proteína Tirosina Quinases/análise , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos , Receptor IGF Tipo 1/análise , Receptores de Superfície Celular/análise , Receptores de Fatores de Crescimento de Fibroblastos/análise , Receptores de Hormônios Paratireóideos/análise , Receptores do Fator de Crescimento Derivado de Plaquetas/análise , Fator de Crescimento Transformador beta/análise , Receptor fas/genética
19.
Calcif Tissue Int ; 66(6): 456-60, 2000 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-10821883

RESUMO

The purpose of this study was to determine if the increased cortical bone porosity induced by intermittently administered parathyroid hormone (PTH) reduces bone strength significantly. Mature ovary-intact New Zealand white rabbits were treated with once daily injections of vehicle, or PTH(1-34), LY333334, at 10 or 40 microg/kg/day for 140 days. Geometry of the femoral midshaft was measured to evaluate changes in the cross-sectional moment of inertia (CSMI). Cortical porosity was measured in the midshaft of the tibia by dividing cortical area into three zones based on equal divisions of cortical diameter: near endocortical (Zone I), near intermediate (Zone II), and near periosteal (Zone III) regions. Total cortical porosity significantly increased after PTH treatment from 1.4% in the controls to 6.3% in the higher dose group, but the location of the new porosities was not randomly distributed. In the controls, porosity of Zones I and II (both 1.7%) was almost twice as much as that of Zone III (0.9%). In the lower dose group, cortical porosity of Zone I (5.5%) and II (1.8%) was greater than in Zone III (0.9%), but these differences were not statistically significant. In the higher dose group, cortical porosity of Zone I (11.5%) and II (6. 1%) significantly increased compared with Zone III (1.4%) (P < 0. 0005). Histomorphometric measurements showed that bone formation rate on both periosteal and endocortical surfaces increased, resulting in increased bone area and cortical area in the higher dose group. A model was developed to evaluate the effect of the changes in geometry and porosity on CSMI in the different zones. This simulation model indicated that CSMI in the higher dose group was significantly greater than in the other two groups, despite the increased porosity. We speculate the reason to be that porosity increased near the endocortical surface, where its mechanical effect is small. This increase was more than offset by apposition of new bone on the periosteal surface. These data suggest that (1) PTH increases cortical porosity in a dose-dependent manner, primarily near endocortical surfaces; (2) because of this nonhomogeneous distribution, the mechanical effect of increased porosity is small; (3) the increased cortical porosity associated with PTH treatment is more than offset by periosteal apposition of new bone, causing an overall increase in the bending rigidity of cortical bone; and (4) these changes cannot be accurately evaluated using noninvasive methods of bone densitometry, which cannot account for the location of bone gain and bone loss.


Assuntos
Osso e Ossos/efeitos dos fármacos , Teriparatida/farmacologia , Animais , Fenômenos Biomecânicos , Osso e Ossos/anatomia & histologia , Osso e Ossos/fisiologia , Relação Dose-Resposta a Droga , Feminino , Fêmur/efeitos dos fármacos , Humanos , Modelos Teóricos , Porosidade/efeitos dos fármacos , Coelhos , Teriparatida/administração & dosagem , Tíbia/efeitos dos fármacos
20.
Bone ; 26(3): 227-34, 2000 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-10709994

RESUMO

Bone cells undergo changes in cell structure during phenotypic development. Parathyroid hormone (PTH) induces a change in osteoblast shape, a determinant of collagen expression. We hypothesize that alterations in bone cell shape reflect and direct gene expression as governed, in part, by nuclear organization. In this study, we determined whether the expression of nuclear matrix proteins that mediate nuclear architecture, NuMA, topoisomerase II (topo II)-alpha, and -beta, were altered during osteoblast development and response to PTH in vivo. NuMA forms an interphase nuclear scaffold in some cells, the absence of which may accommodate alterations in nuclear organization necessary for specific functions. Topo II enzymes are expressed in bone cells; the alpha-isoform is specific to proliferating cells. We used immunohistochemistry and flow cytometry to determine whether NuMA is expressed in the primary spongiosa of the rat metaphyseal femur and whether expression of NuMA, topo II-alpha, and II-beta changes during osteoblast development or with PTH treatment. NuMA and topo II-beta were expressed in marrow cells, osteoblasts, osteocytes, and chondrocytes. These proteins were not detected in osteoclasts in vivo, but were observed in cultured cells. Bone marrow cells expressed topo II-alpha. All three proteins were expressed in cultures of rat osteoblast-like UMR-106 cells. PTH treatment downregulated the number of topo II-alpha-immunopositive cells, correlated with a decrease in S-phase cells, in both bone tissue and cell culture. We conclude that, in vivo, nuclear matrix composition is altered during bone cell development and that anabolic doses of PTH attenuate the proliferative capacity of osteogenic cells, in part, by targeting topo II-alpha expression.


Assuntos
Osso e Ossos/efeitos dos fármacos , DNA Topoisomerases Tipo II/metabolismo , Isoenzimas/metabolismo , Proteínas Nucleares/metabolismo , Hormônio Paratireóideo/farmacologia , Animais , Antígenos de Neoplasias , Antígenos Nucleares , Osso e Ossos/enzimologia , Osso e Ossos/metabolismo , Ciclo Celular , Proteínas de Ciclo Celular , Células Cultivadas , Proteínas de Ligação a DNA , Masculino , Microscopia de Fluorescência , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...