Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Genetics ; 215(1): 129-141, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32156750

RESUMO

The vertebrate limb serves as an experimental paradigm to study mechanisms that regulate development of the stereotypical skeletal elements. In this study, we simultaneously inactivated Sall4 using Hoxb6Cre and Plzf in mouse embryos, and found that their combined function regulates development of the proximal-anterior skeletal elements in hindlimbs. The Sall4; Plzf double knockout exhibits severe defects in the femur, tibia, and anterior digits, distinct defects compared to other allelic series of Sall4; Plzf We found that Sall4 regulates Plzf expression prior to hindlimb outgrowth. Further expression analysis indicated that Hox10 genes and GLI3 are severely downregulated in the Sall4; Plzf double knockout hindlimb bud. In contrast, PLZF expression is reduced but detectable in Sall4; Gli3 double knockout limb buds, and SALL4 is expressed in the Plzf; Gli3 double knockout limb buds. These results indicate that Plzf, Gli3, and Hox10 genes downstream of Sall4, regulate femur and tibia development. In the autopod, we show that Sall4 negatively regulates Hedgehog signaling, which allows for development of the most anterior digit. Collectively, our study illustrates genetic systems that regulate development of the proximal-anterior skeletal elements in hindlimbs.


Assuntos
Desenvolvimento Ósseo , Proteínas de Ligação a DNA/genética , Transdução de Sinais , Fatores de Transcrição/genética , Animais , Proteínas de Ligação a DNA/metabolismo , Fêmur/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Camundongos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Proteína com Dedos de Zinco da Leucemia Promielocítica/genética , Proteína com Dedos de Zinco da Leucemia Promielocítica/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Tíbia/embriologia , Fatores de Transcrição/metabolismo , Proteína Gli3 com Dedos de Zinco/genética , Proteína Gli3 com Dedos de Zinco/metabolismo
2.
J Bone Joint Surg Am ; 100(15): e102, 2018 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-30063598

RESUMO

BACKGROUND: Since healing of anterior cruciate ligament (ACL) grafts occurs by formation of a fibrovascular scar-tissue interface rather than by reformation of the native fibrocartilage transition zone, the purpose of our study was to examine expression of various signaling molecules and transcription factors that are known to be involved in embryologic insertion-site development following ACL reconstruction. We also aimed to characterize a murine model of ACL reconstruction to allow future study of the molecular mechanisms of healing. METHODS: Seventy-nine mice underwent reconstruction of the ACL with autograft. Healing was assessed using histology in 12 mice and quantitative real-time polymerase chain reaction (qRT-PCR) gene-expression analysis in 3 mice at 1 week postoperatively (Group-1 mice) and by biomechanical analysis in 7, histological analysis in 7, immunohistochemical analysis in 5, microcomputed tomography analysis in 5, and qRT-PCR analyses in 8 at 2 weeks (Group-2 mice) and 4 weeks (Group-3 mice) postoperatively. Fifteen additional mice did not undergo surgery and were used for biomechanical (7 mice), qRT-PCR (3 mice), and immunohistochemical (5 mice) analyses to obtain baseline data for the native ACL. RESULTS: Histological analysis demonstrated healing by formation of fibrovascular tissue at the tendon-bone interface. Immunohistochemical analysis showed a positive expression of proteins in the Indian hedgehog, Wnt, and parathyroid hormone-related protein (PTHrP) pathways. There was minimal Sox-9 expression. Gene-expression analysis showed an initial increase in markers of tissue repair and turnover, followed by a subsequent decline. Mean failure force and stiffness of the native ACL were 5.60 N and 3.44 N/mm, respectively. Mean failure force and stiffness were 1.29 N and 2.28 N/mm, respectively, in Group 2 and were 1.79 N and 2.59 N/mm, respectively, in Group 3, with 12 of 14 failures in these study groups occurring by tunnel pull-out. CONCLUSIONS: The spatial and temporal pattern of expression of signaling molecules that direct embryologic insertion-site formation was not adequate to restore the structure and composition of the native insertion site. CLINICAL RELEVANCE: Development of a murine model to study ACL reconstruction will allow the use of transgenic animals to investigate the cellular, molecular, and biomechanical aspects of tendon-to-bone healing following ACL reconstruction, ultimately suggesting methods to improve healing in patients.


Assuntos
Reconstrução do Ligamento Cruzado Anterior , Ligamento Cruzado Anterior/metabolismo , Fêmur/metabolismo , Tíbia/metabolismo , Cicatrização/fisiologia , Animais , Ligamento Cruzado Anterior/diagnóstico por imagem , Ligamento Cruzado Anterior/embriologia , Ligamento Cruzado Anterior/cirurgia , Biomarcadores/metabolismo , Fenômenos Biomecânicos , Fêmur/diagnóstico por imagem , Fêmur/embriologia , Fêmur/cirurgia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais , Tíbia/diagnóstico por imagem , Tíbia/embriologia , Tíbia/cirurgia , Transplante Autólogo , Microtomografia por Raio-X
3.
Toxicol Lett ; 295: 249-255, 2018 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-29981921

RESUMO

This study investigated the mechanisms underlying the retarded development of long bone in fetus by prenatal nicotine exposure (PNE) which had been demonstrated by our previous work. Nicotine (2.0 mg/kg.d) or saline was injected subcutaneously into pregnant rats every morning from gestational day (GD) 9 to 20. Fetal femurs or tibias were harvested for analysis on GD 20. We found massive accumulation of hypertrophic chondrocytes and a delayed formation of primary ossification center (POC) in the fetal femur or tibia of rat fetus after PNE, which was accompanied by a decreased amount of osteoclasts in the POC and up-regulated expression of osteoprotegerin (OPG) but by no obvious change in the expression of receptor activator of NF-κB ligand (RANKL). In primary osteoblastic cells, both nicotine (0, 162, 1620, 16,200 ng/ml) and corticosterone (0, 50, 250, 1250 nM) promoted the mRNA expression of OPG but concentration-dependently suppressed that of RANKL. Furthermore, blocking α4ß2-nicotinic acetylcholine receptor (α4ß2-nAChR) or glucocorticoid receptor rescued the above effects of nicotine and corticosterone, respectively. In conclusion, retarded osteoclastogenesis may contribute to delayed endochondral ossification in long bone in fetal rats with PNE. The adverse effects of PNE may be mediated via the direct effect of nicotine and indirect effect of maternal corticosterone on osteoblastic cells.


Assuntos
Desenvolvimento Ósseo/efeitos dos fármacos , Fêmur/efeitos dos fármacos , Nicotina/toxicidade , Agonistas Nicotínicos/toxicidade , Osteogênese/efeitos dos fármacos , Tíbia/efeitos dos fármacos , Animais , Células Cultivadas , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Condrócitos/patologia , Corticosterona/toxicidade , Feminino , Fêmur/embriologia , Fêmur/metabolismo , Idade Gestacional , Exposição Materna/efeitos adversos , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Osteoclastos/patologia , Osteoprotegerina/metabolismo , Gravidez , Ligante RANK/metabolismo , Ratos Wistar , Receptores de Glucocorticoides/metabolismo , Receptores Nicotínicos/metabolismo , Tíbia/embriologia , Tíbia/metabolismo
4.
J Cell Biol ; 216(9): 2911-2926, 2017 09 04.
Artigo em Inglês | MEDLINE | ID: mdl-28696225

RESUMO

Autosomal-recessive omodysplasia (OMOD1) is a genetic condition characterized by short stature, shortened limbs, and facial dysmorphism. OMOD1 is caused by loss-of-function mutations of glypican 6 (GPC6). In this study, we show that GPC6-null embryos display most of the abnormalities found in OMOD1 patients and that Hedgehog (Hh) signaling is significantly reduced in the long bones of these embryos. The Hh-stimulatory activity of GPC6 was also observed in cultured cells, where this GPC increased the binding of Hh to Patched 1 (Ptc1). Consistent with this, GPC6 interacts with Hh through its core protein and with Ptc1 through its glycosaminoglycan chains. Hh signaling is triggered at the primary cilium. In the absence of Hh, we observed that GPC6 is localized outside of the cilium but moves into the cilium upon the addition of Hh. We conclude that GPC6 stimulates Hh signaling by binding to Hh and Ptc1 at the cilium and increasing the interaction of the receptor and ligand.


Assuntos
Fêmur/metabolismo , Glipicanas/metabolismo , Transtornos do Crescimento/metabolismo , Proteínas Hedgehog/metabolismo , Osteocondrodisplasias/congênito , Osteogênese , Tíbia/metabolismo , Animais , Proliferação de Células , Cílios/metabolismo , Modelos Animais de Doenças , Fêmur/embriologia , Predisposição Genética para Doença , Glicosaminoglicanos/metabolismo , Glipicanas/deficiência , Glipicanas/genética , Transtornos do Crescimento/embriologia , Transtornos do Crescimento/genética , Células HEK293 , Proteínas Hedgehog/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células NIH 3T3 , Osteocondrodisplasias/embriologia , Osteocondrodisplasias/genética , Osteocondrodisplasias/metabolismo , Receptor Patched-1/metabolismo , Fenótipo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Transdução de Sinais , Tíbia/embriologia , Fatores de Tempo , Transfecção , Proteína GLI1 em Dedos de Zinco/metabolismo
5.
Eur J Histochem ; 61(1): 2750, 2017 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-28348418

RESUMO

The neuropeptide Calcitonin Gene-Related Peptide (CGRP) is a well-characterized neurotransmitter. However, little is known about the role of CGRP in osteogenesis and vascular genesis during the developmental formation of bone. In the present study, we assessed the abundance of CGRP mRNA and the mRNA of osteogenesis and vascular genesis markers in the foetal mouse mandible and leg bone (tibia). We also analysed the expression and localization of CGRP, osteopontin (OPN) and vascular endothelial growth factor (VEGF-A) using in situ hybridization and immunohistochemical localization in the mouse mandible and tibia at embryonic days 12.5 (E12.5), E14.5, E17.5, and postnatal day 1 (P1). CGRP was clearly detected in the mandible relative to the tibia at E14.5. Hybridization using an anti-sense probe for CGRP was not detected in the mandible at P1. Hybridization with an anti-sense probe for OPN was detected at E14.5, later in the mandible and at P1 in Meckel's cartilage. However, OPN was only detected in the tibia at E17.5 and later. The abundance of CGRP mRNA differed between the mandible and tibia. The level of vasculogenesis markers, such as VEGF-A, was similar to that of CGRP in the mandible. The levels of VEGF-A, cluster of differentiation 31 (CD31) and lymphatic vessel endothelial hyaluronan receptor 1 (LIVE-1) differed from that of OPN in the mandible. In contrast, the levels of VEGF-A, CD31, matrix metalloproteinase-2 (MMP-2), collagen I (Col I), collagen II (Col II) and OPN mRNA differed from E12.5 to P1 (P<0.001) in the tibia. The abundance of mRNA of CGRP and bone matrix markers (Col I, Col II, and OPN) was low at P5 in the tibia. These differences in CGRP and other mRNAs may induce a different manner of ossification between the mandible and tibia. Therefore, a time lag of ossification occurs between the mandible and tibia during foetal development.


Assuntos
Peptídeo Relacionado com Gene de Calcitonina/biossíntese , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Mandíbula , Neovascularização Fisiológica/fisiologia , Osteogênese/fisiologia , Tíbia , Animais , Mandíbula/irrigação sanguínea , Mandíbula/embriologia , Metaloproteinase 2 da Matriz/biossíntese , Camundongos , Molécula-1 de Adesão Celular Endotelial a Plaquetas/biossíntese , RNA Mensageiro/biossíntese , Tíbia/irrigação sanguínea , Tíbia/embriologia , Fator A de Crescimento do Endotélio Vascular/biossíntese
6.
PLoS One ; 8(12): e81569, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24324705

RESUMO

Achondroplasia (ACH) is one of the most common skeletal dysplasias with short stature caused by gain-of-function mutations in FGFR3 encoding the fibroblast growth factor receptor 3. We used the drug repositioning strategy to identify an FDA-approved drug that suppresses abnormally activated FGFR3 signaling in ACH. We found that meclozine, an anti-histamine drug that has long been used for motion sickness, facilitates chondrocyte proliferation and mitigates loss of extracellular matrix in FGF2-treated rat chondrosarcoma (RCS) cells. Meclozine also ameliorated abnormally suppressed proliferation of human chondrosarcoma (HCS-2/8) cells that were infected with lentivirus expressing constitutively active mutants of FGFR3-K650E causing thanatophoric dysplasia, FGFR3-K650M causing SADDAN, and FGFR3-G380R causing ACH. Similarly, meclozine alleviated abnormally suppressed differentiation of ATDC5 chondrogenic cells expressing FGFR3-K650E and -G380R in micromass culture. We also confirmed that meclozine alleviates FGF2-mediated longitudinal growth inhibition of embryonic tibia in bone explant culture. Interestingly, meclozine enhanced growth of embryonic tibia in explant culture even in the absence of FGF2 treatment. Analyses of intracellular FGFR3 signaling disclosed that meclozine downregulates phosphorylation of ERK but not of MEK in FGF2-treated RCS cells. Similarly, meclozine enhanced proliferation of RCS cells expressing constitutively active mutants of MEK and RAF but not of ERK, which suggests that meclozine downregulates the FGFR3 signaling by possibly attenuating ERK phosphorylation. We used the C-natriuretic peptide (CNP) as a potent inhibitor of the FGFR3 signaling throughout our experiments, and found that meclozine was as efficient as CNP in attenuating the abnormal FGFR3 signaling. We propose that meclozine is a potential therapeutic agent for treating ACH and other FGFR3-related skeletal dysplasias.


Assuntos
Acondroplasia/patologia , Diferenciação Celular/efeitos dos fármacos , Condrócitos/metabolismo , Condrócitos/patologia , Meclizina/farmacologia , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Acondroplasia/metabolismo , Substituição de Aminoácidos , Animais , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Condrócitos/efeitos dos fármacos , Condrócitos/enzimologia , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Fator 2 de Crescimento de Fibroblastos/farmacologia , Humanos , Lentivirus/metabolismo , Modelos Biológicos , Mutação/genética , Fosforilação/efeitos dos fármacos , Ratos , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/antagonistas & inibidores , Tíbia/efeitos dos fármacos , Tíbia/embriologia , Tíbia/patologia
7.
Arthritis Rheum ; 65(3): 743-52, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23233348

RESUMO

OBJECTIVE: Syndecan 4, a heparan sulfate proteoglycan, has been associated with osteoarthritis. The present study was undertaken to analyze the functional role of syndecan 4 in endochondral ossification of mouse embryos and in adult fracture repair, which, like osteoarthritis, involves an inflammatory component. METHODS: Sdc4 promoter activity was analyzed in Sdc4(-/-) lacZ-knockin mice, using ß-galactosidase staining. Endochondral ossification in embryos from embryonic day 16.5 was assessed by histologic and immunohistologic staining. Bone fracture repair was analyzed in femora of adult mice on days 7 and 14 postfracture. To evaluate Sdc2 and Sdc4 gene expression with and without tumor necrosis factor α (TNFα) and Wnt-3a stimulation, quantitative real-time polymerase chain reaction was performed. RESULTS: In Sdc4(-/-) lacZ-knockin animals, syndecan 4 promoter activity was detectable at all stages of chondrocyte differentiation, and Sdc4 deficiency inhibited chondrocyte proliferation. Aggrecan turnover in the uncalcified cartilage of the epiphysis was decreased transiently in vivo, but this did not lead to a growth phenotype at birth. In contrast, among adult mice, fracture healing was markedly delayed in Sdc4(-/-) animals and was accompanied by increased callus formation. Blocking of inflammation via anti-TNFα treatment during fracture healing reduced these changes in Sdc4(-/-) mice to levels observed in wild-type controls. We analyzed the differences between the mild embryonic and the severe adult phenotype, and found a compensatory up-regulation of syndecan 2 in the developing cartilage of Sdc4(-/-) mice that was absent in adult tissue. Stimulation of chondrocytes with Wnt-3a in vitro led to increased expression of syndecan 2, while stimulation with TNFα resulted in up-regulation of syndecan 4 but decreased expression of syndecan 2. TNFα stimulation reduced syndecan 2 expression and increased syndecan 4 expression even in the presence of Wnt-3a, suggesting that inflammation has a strong effect on the regulation of syndecan expression. CONCLUSION: Our results demonstrate that syndecan 4 is functionally involved in endochondral ossification and that its loss impairs fracture healing, due to inhibition of compensatory mechanisms under inflammatory conditions.


Assuntos
Desenvolvimento Ósseo/fisiologia , Fraturas do Fêmur/fisiopatologia , Consolidação da Fratura/fisiologia , Sindecana-4/fisiologia , Animais , Diferenciação Celular/fisiologia , Condrócitos/citologia , Condrócitos/fisiologia , Feminino , Fêmur/citologia , Fêmur/embriologia , Fêmur/fisiologia , Lâmina de Crescimento/citologia , Lâmina de Crescimento/embriologia , Lâmina de Crescimento/fisiologia , Inflamação/fisiopatologia , Óperon Lac/genética , Masculino , Camundongos , Camundongos Knockout , Osteogênese/fisiologia , Gravidez , Regiões Promotoras Genéticas/fisiologia , RNA Mensageiro/metabolismo , Sindecana-2/genética , Sindecana-2/fisiologia , Sindecana-4/genética , Tíbia/citologia , Tíbia/embriologia , Tíbia/fisiologia
8.
Am J Pathol ; 180(2): 727-37, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22154935

RESUMO

Cartilage oligomeric matrix protein (COMP), a secreted glycoprotein synthesized by chondrocytes, regulates proliferation and type II collagen assembly. Mutations in the COMP gene cause pseudoachondroplasia and multiple epiphyseal dysplasia. Previously, we have shown that expression of D469del-COMP in transgenic mice causes intracellular retention of D469del-COMP, thereby recapitulating pseudoachondroplasia chondrocyte pathology. This inducible transgenic D469del-COMP mouse is the only in vivo model to replicate the critical cellular and clinical features of pseudoachondroplasia. Here, we report developmental studies of D469del-COMP-induced chondrocyte pathology from the prenatal period to adolescence. D469del-COMP retention was limited prenatally and did not negatively affect the growth plate until 3 weeks after birth. Results of immunostaining, transcriptome analysis, and qRT-PCR suggest a molecular model in which D469del-COMP triggers apoptosis during the first postnatal week. By 3 weeks (when most chondrocytes are retaining D469del-COMP), inflammation, oxidative stress, and DNA damage contribute to chondrocyte cell death by necroptosis. Importantly, by crossing the D469del-COMP mouse onto a Chop null background (Ddit3 null), thereby eliminating Chop, the unfolded protein response was disrupted, thus alleviating both D469del-COMP intracellular retention and premature chondrocyte cell death. Chop therefore plays a significant role in processes that mediate D469del-COMP retention. Taken together, these results suggest that there may be an optimal window before the induction of significant D469del-COMP retention during which endoplasmic reticulum stress could be targeted.


Assuntos
Acondroplasia/fisiopatologia , Condrócitos/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Glicoproteínas/metabolismo , Lâmina de Crescimento/patologia , Fator de Transcrição CHOP/fisiologia , Acondroplasia/embriologia , Animais , Apoptose , Proteínas Reguladoras de Apoptose/metabolismo , Desenvolvimento Ósseo/fisiologia , Cartilagem Articular/embriologia , Cartilagem Articular/crescimento & desenvolvimento , Cartilagem Articular/metabolismo , Lâmina de Crescimento/embriologia , Lâmina de Crescimento/crescimento & desenvolvimento , Membro Posterior/embriologia , Membro Posterior/crescimento & desenvolvimento , Membro Posterior/metabolismo , Proteínas Matrilinas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Tíbia/embriologia , Tíbia/crescimento & desenvolvimento , Tíbia/metabolismo , Regulação para Cima
9.
J Biomed Mater Res A ; 86(3): 597-606, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18004795

RESUMO

Previous work has reported the results of a multidisciplinary effort producing a proof-of-concept on the use of pectic polysaccharides in the surface modification of medical devices. This study was designed to learn more about the capability of engineered rhamnogalacturonan-I (RG-I) fractions of apple pectin to control bone cell and macrophage behavior. Thermanox or polystyrene Petri dishes were surface modified with two different modified hairy regions (MHRs) obtained by different enzymatic liquefaction processes of apples differing in relative amounts and lengths of their neutral side chains: (long-haired) MHR-alpha and (short-haired) MHR-B. Bone explants from 14-day-old chick embryos were cultured for 14 days on both pectic substrata. MHR-B promoted cell migration and differentiation, MHR-alpha did not. On MHR-alpha, J774.2 macrophages grew well, their percentage in G1 phase was decreased and in S phase increased, and they did not secrete either proinflammatory-cytokines or nitrites. Contrasting results were gained from macrophages on MHR-B, except for nitrite secretion. Thus, we conclude that coatings from tailored pectins show different biological activities in vitro and are potential innovative candidates for improving the biocompatibility of medical devices in various applications.


Assuntos
Enzimas Imobilizadas/metabolismo , Macrófagos/citologia , Pectinas/metabolismo , Tíbia/citologia , Animais , Ciclo Celular , Movimento Celular , Proliferação de Células , Forma Celular , Embrião de Galinha , Técnicas In Vitro , Camundongos , Poliestirenos/metabolismo , Tíbia/embriologia , Tíbia/ultraestrutura , Fator de Necrose Tumoral alfa/metabolismo
10.
Bone ; 40(2): 337-44, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17029980

RESUMO

The transcription factor nuclear factor E2 p45-related factor 2 (Nrf2) forms heterodimers with small musculoaponeurotic fibrosarcoma (Maf) proteins for the selective recognition of the antioxidant responsive element on target genes, followed by the regulation of gene expression of phase II detoxifying enzymes as well as oxidative-stress-inducible proteins in different tissues. In the present study, we investigated the role of Nrf2 in the regulation of chondrocyte differentiation as well as the expression pattern of Nrf2 in cartilage. In tibia from embryonic mice at E15.5, Nrf2 mRNA expression was restricted to both proliferating and pre-hypertrophic chondrocytes, with few signals in early and late hypertrophic chondrocytes expressing both type X collagen and osteopontin. On in situ hybridization analysis of tibia from neonatal mice at 1 day after birth, by contrast, Nrf2 was expressed in all chondrocytic layers in addition to osteoblasts attached to cancellous bone. In pre-chondrogenic cell line ATDC5 cells, furthermore, expression of Nrf2 mRNA was also confirmed together with mRNA expression of the Kelch-like ECH associating protein 1 and small Maf proteins. In ATDC5 cells stably transfected with Nrf2, significant inhibition was seen in the differentiation-dependent induction of alkaline phosphatase and increase in the Alcian blue staining intensity. Furthermore, stable overexpression of Nrf2 significantly decreased mRNA expression of several chondrocyte differentiation markers such as type II collagen, type X collagen and osteopontin. These data suggest that Nrf2 may be a negative regulator of the cellular differentiation toward maturation in chondrocytes.


Assuntos
Condrócitos/fisiologia , Fator 2 Relacionado a NF-E2/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Antígenos de Diferenciação/metabolismo , Cartilagem/citologia , Cartilagem/embriologia , Diferenciação Celular , Linhagem Celular , Condrócitos/citologia , Colágeno Tipo X/metabolismo , Proteínas do Citoesqueleto/metabolismo , Proteína 1 Associada a ECH Semelhante a Kelch , Camundongos , Osteoblastos/citologia , Osteoblastos/fisiologia , Osteopontina/metabolismo , Tíbia/citologia , Tíbia/embriologia
11.
J Bone Miner Res ; 20(6): 1011-21, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15883642

RESUMO

UNLABELLED: In situ hybridization studies allowed for the localization of three MMPs and the angiogenic factor VEGF during secondary ossification. MMPs were widely expressed during ossification of the secondary center, whereas expression of VEGF was restricted to later stages. INTRODUCTION: The spatiotemporal expression patterns of the matrix metalloproteinases gelatinase-B (MMP-9), collagenase-3 (MMP-13), and membrane-type 1 metalloproteinase (MMP-14) and the angiogenic peptide vascular endothelial growth factor (VEGF) were studied during development of the proximal epiphysis of the rat tibia. MATERIALS AND METHODS: Cell expression was analyzed by in situ hybridization. Studies on osteoclastic activity, matrix mineralization, cell proliferation, and vascular progression were also performed. RESULTS: MMP-9, MMP-13, and MMP-14 were expressed in discrete perichondrial cells that gave way to sites of intrachondral canal formation. High expression levels for the three MMPs were found at the blind ends of advancing intrachondral canals and at the expanding borders of the marrow space. Signals for MMP-9 and MMP-13 were in close proximity but did not overlap, whereas MMP-14 was expressed in both MMP-9+ and MMP-13+ cells. VEGF was not expressed during formation of intrachondral vascular canals but was observed in hypertrophic chondrocytes during formation of the bone marrow cavity. CONCLUSIONS: Expression of MMPs and VEGF are constant events during development of the secondary ossification center. We propose that MMPs are involved in targeting proteolytic activity during epiphyseal development. VEGF is not expressed during early formation of vascular canals, but it may have a role in the formation of the bone marrow cavity.


Assuntos
Osso e Ossos/fisiologia , Colagenases/biossíntese , Epífises/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Regulação Enzimológica da Expressão Gênica , Metaloproteinase 9 da Matriz/biossíntese , Metaloendopeptidases/biossíntese , Tíbia/embriologia , Fator A de Crescimento do Endotélio Vascular/biossíntese , Animais , Antimetabólitos/farmacologia , Desenvolvimento Ósseo , Células da Medula Óssea/citologia , Osso e Ossos/metabolismo , Bromodesoxiuridina/farmacologia , Proliferação de Células , Endotélio Vascular/citologia , Hibridização In Situ , Masculino , Metaloproteinase 13 da Matriz , Metaloproteinases da Matriz Associadas à Membrana , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
12.
Bone ; 36(6): 1056-64, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15869918

RESUMO

We previously reported that C-type natriuretic peptide (CNP) stimulates endochondral ossification and corrects the reduction in body length of achondroplasia model mouse with constitutive active fibroblast growth factor receptor 3 (FGFR-3). In order to examine the interaction between CNP and FGFR-3, we studied intracellular signaling by using ATDC5 cells, a mouse chondrogenic cell line, and found that FGF2 and FGF18 markedly reduced CNP-dependent intracellular cGMP production, and that these effects were attenuated by MAPK inhibitors. Western blot analysis demonstrated that the level of GC-B, a particulate guanylyl cyclase specific for CNP, was not changed by treatment with FGFs. Conversely, CNP and 8-bromo-cGMP strongly and dose-dependently inhibited the induction of ERK phosphorylation by FGF2 and FGF18 without changing the level of FGFR-3, although they did not affect the phosphorylation of STAT-1. In the organ-cultured fetal mouse tibias, CNP and FGF18 counteracted on the longitudinal bone growth, and both the size and number of hypertrophic chondrocytes. The FGF/FGFR-3 pathway is known as the negative regulator of endochondral ossification. We found that FGFs inhibited CNP-stimulated cGMP production by disrupting the signaling pathway through GC-B while CNP antagonized the activation of the MAPK cascade by FGFs. These results suggest that the CNP/GC-B pathway plays an important role in growth plate chondrocytes and constitutes the negative cross talk between FGFs and the activity of MAPK. Our results may explain one of the molecular mechanisms of the growth stimulating action of CNP and suggest that activation of the CNP/GC-B pathway may be effective as a novel therapeutic strategy for achondroplasia.


Assuntos
Condrócitos/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Peptídeo Natriurético Tipo C/farmacologia , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/fisiologia , Animais , Butadienos/farmacologia , Contagem de Células , Diferenciação Celular , Linhagem Celular Tumoral , Tamanho Celular/efeitos dos fármacos , Condrócitos/efeitos dos fármacos , Condrócitos/patologia , GMP Cíclico/metabolismo , Fator 2 de Crescimento de Fibroblastos/farmacologia , Fatores de Crescimento de Fibroblastos/farmacologia , Flavonoides/farmacologia , Expressão Gênica/efeitos dos fármacos , Expressão Gênica/genética , Lâmina de Crescimento/citologia , Lâmina de Crescimento/efeitos dos fármacos , Guanilato Ciclase/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos ICR , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Nitrilas/farmacologia , Técnicas de Cultura de Órgãos , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Ratos , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Tíbia/efeitos dos fármacos , Tíbia/embriologia , Tíbia/fisiologia
13.
Birth Defects Res A Clin Mol Teratol ; 73(6): 461-7, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15786497

RESUMO

BACKGROUND: Little is known about the developmental changes associated with tibial ray deficiencies. The aim of this study was to detect cell death, proliferation, and gene expression that result in tibial ray deficiencies. METHODS: We induced tibial ray deficiencies in rat embryos using a teratogenic agent (busulfan) and observed the developmental changes in 1126 hindlimbs. We performed Nile blue staining, whole mount in situ hybridization for fibroblast growth factor 8 (Fgf8), bone morphogenetic protein 4 (Bmp4) and Sonic hedgehog (Shh), terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end-labeling (TUNEL) and assessment of cell proliferation by 5-bromo-2'-deoxy-uridine (BrdU)/anti-BrdU immunohistochemistry. RESULTS: In situ hybridization showed reductions in Fgf8 and Bmp4 expression. Histological examination showed a delay of mesenchymal condensation, increased mesenchymal cell death, decreased mesenchymal cell proliferation, and a reduction in the number of mesenchymal cells. These abnormalities may cause hypoplasia of the limb. Bmp4 expression was markedly reduced in the anterior mesenchyme. Shh was expressed in the posterior mesenchyme. We suggest that the posterior skeletal elements may be fully formed owing to Shh expression, but the anterior skeletal elements may be underdeveloped owing to an intense reduction of Bmp4 expression in the anterior mesenchyme, causing hypoplasia of the tibial ray. CONCLUSIONS: The combined effects of increased cell death, decreased cell proliferation, reduction of Fgf8 expression, and intense reduction of Bmp4 expression in the anterior mesenchyme may play an important role in the development of tibial ray deficiency induced by busulfan.


Assuntos
Antineoplásicos Alquilantes/toxicidade , Bussulfano/toxicidade , Teratogênicos/toxicidade , Tíbia/efeitos dos fármacos , Animais , Proteína Morfogenética Óssea 4 , Proteínas Morfogenéticas Ósseas/genética , Bromodesoxiuridina , Diferenciação Celular , Divisão Celular , Fator 8 de Crescimento de Fibroblasto , Fatores de Crescimento de Fibroblastos/genética , Proteínas Hedgehog , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Ratos , Tíbia/embriologia , Transativadores/genética
14.
Anat Rec A Discov Mol Cell Evol Biol ; 279(2): 729-35, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15278943

RESUMO

Ellis-van Creveld (EvC) syndrome is an autosomal recessive chondrodysplasia characterized by short limbs, postaxial polydactyly, natal teeth, and dysplastic nails. The Ellis-van Creveld (EVC) gene, which is mutated in patients with EvC syndrome, has been identified by positional cloning. However, the physiological roles of the EVC gene have not been elucidated. Histopathological analyses of EvC syndrome have shown disturbed chondrocytic phenotypes during cartilage development. We therefore postulated that the EVC gene is a critical factor for chondrocytes during endochondral ossification. The present study focuses on the relationship between the Evc gene and chondrocytes, and examines Evc gene expression in the rat tibial growth plate at the mRNA and protein levels. Evc mRNA in tibial epiphyseal cartilage was expressed at postnatal day (P) 1, P28, and P56 by RT-PCR. Immunohistochemical analyses localized the Evc protein mainly in prehypertrophic and hypertrophic chondrocytes of the epiphyseal growth plate in the tibia during the embryonic and postnatal periods. Evc mRNA was also detected in prehypertrophic and hypertrophic chondrocytes by in situ hybridization. These results indicate that the Evc gene functions mainly in the prehypertrophic and hypertrophic chondrocytes of the epiphyseal growth plate. The data presented here are important for future studies of the underlying mechanism of chondrodysplasia in EvC syndrome.


Assuntos
Lâmina de Crescimento/embriologia , Lâmina de Crescimento/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Nucleares/metabolismo , Tíbia/embriologia , Tíbia/metabolismo , Animais , Western Blotting , Embrião de Mamíferos/metabolismo , Imuno-Histoquímica , Hibridização In Situ , Proteínas de Membrana/genética , Proteínas Nucleares/genética , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Mol Cell Biol ; 24(4): 1691-9, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14749384

RESUMO

Matrilin-3 belongs to the matrilin family of extracellular matrix (ECM) proteins and is primarily expressed in cartilage. Mutations in the gene encoding human matrilin-3 (MATN-3) lead to autosomal dominant skeletal disorders, such as multiple epiphyseal dysplasia (MED), which is characterized by short stature and early-onset osteoarthritis, and bilateral hereditary microepiphyseal dysplasia, a variant form of MED characterized by pain in the hip and knee joints. To assess the function of matrilin-3 during skeletal development, we have generated Matn-3 null mice. Homozygous mutant mice appear normal, are fertile, and show no obvious skeletal malformations. Histological and ultrastructural analyses reveal endochondral bone formation indistinguishable from that of wild-type animals. Northern blot, immunohistochemical, and biochemical analyses indicated no compensatory upregulation of any other member of the matrilin family. Altogether, our findings suggest functional redundancy among matrilins and demonstrate that the phenotypes of MED disorders are not caused by the absence of matrilin-3 in cartilage ECM.


Assuntos
Desenvolvimento Ósseo , Osso e Ossos/embriologia , Osso e Ossos/metabolismo , Proteínas da Matriz Extracelular/deficiência , Proteínas da Matriz Extracelular/metabolismo , Animais , Cartilagem/citologia , Diferenciação Celular , Proteínas da Matriz Extracelular/genética , Deleção de Genes , Imuno-Histoquímica , Proteínas Matrilinas , Camundongos , Camundongos Knockout , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Esqueleto , Tíbia/citologia , Tíbia/embriologia , Tíbia/crescimento & desenvolvimento , Tíbia/ultraestrutura
16.
J Bone Miner Res ; 18(10): 1872-81, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14584898

RESUMO

UNLABELLED: Mineralization often occurs in areas of apoptotic changes. Our findings indicate that physiological mineralization is mediated by matrix vesicles. These matrix vesicles use mechanisms to induce mineralization that are different from the mechanisms used by apoptotic bodies released from apoptotic cells. Therefore, different therapeutic approaches must be chosen to inhibit pathological mineralization depending on the mechanism of mineralization (matrix vesicles versus apoptotic bodies). INTRODUCTION: Physiological mineralization in growth plate cartilage is restricted to regions of terminally differentiated and apoptotic chondrocytes. Pathological mineralization of tissues also often occurs in areas of apoptosis. We addressed the question of whether apoptotic changes control mineralization events or whether both events are regulated independently. METHODS: To induce mineralization, we treated growth plate chondrocytes with retinoic acid (RA); apoptosis in these cells was induced by treatment with staurosporine, anti-Fas, or TNFalpha. The degrees of mineralization and apoptosis were determined, and the structure and function of matrix vesicles and apoptotic bodies were compared. RESULTS: Release of matrix vesicles and mineralization in vivo in the growth plate occurs earlier than do apoptotic changes. To determine the functional relationship between apoptotic bodies and matrix vesicles, growth plate chondrocytes were treated with RA to induce matrix vesicle release and with staurosporine to induce release of apoptotic bodies. After 3 days, approximately 90% of staurosporine-treated chondrocytes were apoptotic, whereas only 2-4% of RA-treated cells showed apoptotic changes. RA- and staurosporine-treated chondrocyte cultures were mineralized after 3 days. Matrix vesicles isolated from RA-treated cultures and apoptotic bodies isolated from staurosporine-treated cultures were associated with calcium and phosphate. However, matrix vesicles were bigger than apoptotic bodies. Furthermore, matrix vesicles but not apoptotic bodies contained alkaline phosphatase and Ca2+ channel-forming annexins II, V, and VI. Consequently, matrix vesicles but not apoptotic bodies were able to take up Ca2+ and form the first mineral phase inside their lumen. Mineralization of RA-treated cultures was inhibited by antibodies specific for annexin V but not mineralization of staurosporine-treated cultures. CONCLUSION: Physiological mineralization of growth plate chondrocytes is initiated by specialized matrix vesicles and requires alkaline phosphatase and annexins. In contrast, mineral formation mediated by apoptotic bodies occurs by a default mechanism and does not require alkaline phosphatase and annexins.


Assuntos
Apoptose , Matriz Extracelular/metabolismo , Fosfatase Alcalina/metabolismo , Animais , Anexinas/metabolismo , Antraquinonas/farmacologia , Cálcio/metabolismo , Caspase 3 , Caspases/metabolismo , Divisão Celular , Embrião de Galinha , Condrócitos/metabolismo , Corantes/farmacologia , Eletroforese em Gel de Poliacrilamida , Ativação Enzimática , Immunoblotting , Marcação In Situ das Extremidades Cortadas , Microscopia Eletrônica , Estaurosporina/metabolismo , Tíbia/embriologia , Tretinoína/metabolismo , Tretinoína/farmacologia , Receptor fas/metabolismo
17.
Gene ; 305(2): 185-94, 2003 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-12609739

RESUMO

We have previously reported the modulation, during chondrogenesis and/or inflammation, of two chicken genes laying in the same genomic locus and coding for two polypeptides of the lipocalin protein family, the extracellular fatty acid binding protein (ExFABP) and the chondrogenesis associated lipocalin beta (CALbeta). A third gene, located within the same cluster and coding for a new lipocalin, CALgamma, has been identified and is here characterized. Tissue distribution analyzed by real-time quantitative reverse transcriptase-polymerase chain reaction in chicken embryos shows a ubiquitous expression with predominant levels of mRNA transcripts in the liver and the brain. In the developing tibia, a high expression of CALgamma mRNA was evidenced by in situ hybridization within the pre-hypertrophic and the hypertrophic zones of the bone-forming cartilage. In agreement, dedifferentiated chondrocytes in vitro express the transcripts to the highest level when they re-differentiate reaching hypertrophy. Such peculiar developmental pattern of expression that is analogous to those already described for Ex-FABP and CALbeta suggests that all three proteins may act synergistically in the process of endochondral bone formation. Moreover, like Ex-FABP and CALbeta, CALgamma is also highly induced in dedifferentiated chondrocytes upon stimulation with lypopolysaccharides, indicating that the whole cluster quite possibly is transcriptionally activated not only in physiological morphogenic differentiation but also in pathological acute phase response.


Assuntos
Proteínas de Transporte/genética , Galinhas/genética , Família Multigênica/genética , Sequência de Aminoácidos , Animais , Encéfalo/embriologia , Encéfalo/metabolismo , Diferenciação Celular/genética , Divisão Celular/genética , Células Cultivadas , Embrião de Galinha , Condrócitos/citologia , Condrócitos/metabolismo , Colágeno/genética , DNA/química , DNA/genética , Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Hibridização In Situ , Fígado/embriologia , Fígado/metabolismo , Dados de Sequência Molecular , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Tíbia/embriologia , Tíbia/metabolismo , Fatores de Tempo
18.
Dev Dyn ; 225(3): 250-9, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12412007

RESUMO

We previously observed that the perichondrium (PC) and the periosteum (PO) negatively regulate endochondral cartilage growth through secreted factors. Conditioned medium from cultures of PC and PO cells when mixed (PC/PO-conditioned medium) and tested on organ cultures of embryonic chicken tibiotarsi from which the PC and PO have been removed (PC/PO-free cultures) effect negative regulation of growth. Of potential importance, this regulation compensates precisely for removal of the PC and PO, thus mimicking the regulation effected by these tissues in vivo. We have now examined whether two known negative regulators of cartilage growth (retinoic acid [RA] and transforming growth factor-beta1 [TGF-beta1]) act in a manner consistent with this PC/PO-mediated regulation. The results suggest that RA and TGF-beta1, per se, are not the regulators in the PC/PO-conditioned medium. Instead, they show that these two factors each act in regulating cartilage growth through an additional, previously undescribed, negative regulatory mechanism(s) involving the perichondrium. When cultures of perichondrial cells (but not periosteal cells) are treated with either agent, they secrete secondary regulatory factors into their conditioned medium, the action of which is to effect precise negative regulation of cartilage growth when tested on the PC/PO-free organ cultures. This negative regulation through the perichondrium is the only activity detected with TGF-beta1. Whereas, RA shows additional regulation on the cartilage itself. However, this regulation by RA is not "precise" in that it produces abnormally shortened cartilages. Overall, the precise regulation of cartilage growth effected by the action of the perichondrial-derived factor(s) elicited from the perichondrial cells by treatment with either RA or TGF-beta1, when combined with our previous results showing similar--yet clearly different--"precise" regulation by the PC/PO-conditioned medium suggests the existence of multiple mechanisms involving the perichondrium, possibly interrelated or redundant, to ensure the proper growth of endochondral skeletal elements.


Assuntos
Cartilagem/citologia , Cartilagem/embriologia , Animais , Antineoplásicos/farmacologia , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Células Cultivadas , Embrião de Galinha , Condrócitos/efeitos dos fármacos , Condrócitos/fisiologia , Meios de Cultivo Condicionados/farmacologia , Técnicas de Cultura de Órgãos , Periósteo/citologia , Periósteo/embriologia , Tarso Animal/citologia , Tarso Animal/embriologia , Tíbia/citologia , Tíbia/embriologia , Fator de Crescimento Transformador beta/farmacologia , Fator de Crescimento Transformador beta1 , Tretinoína/farmacologia
19.
J Anat ; 201(2): 153-7, 2002 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12220123

RESUMO

Vascular endothelial growth factor (VEGF) plays an important role during endochondral bone formation in hypertrophic cartilage remodelling. We examined VEGF and VEGF receptor expression in tibiae from fetuses, newborns and children immunohistochemically. Expression of mRNA for the different VEGF splice forms and for VEGF receptors KDR and FLT-1 was analysed by reverse transcription-polymerase chain reaction (RT-PCR). VEGF could be immunolocalized intracellularly in the hypertrophic chondrocytes of the growth plate and in the chondrocytes around cartilage canals of the epiphysis, respectively. The resting zone and the proliferative zone of the growth plate were VEGF-negative. In cartilage samples of all growth plates analysed, VEGF121 and VEGF165 were identified as the only VEGF splice forms expressed. RT-PCR for VEGF mRNA of normal hyaline cartilage was negative. At vessels growing into the hypertrophic cartilage FLT-1 (VEGFR-1) and KDR (VGEFR-2) could be visualized. Reverse transcription-polymerase chain reaction (RT-PCR) substantiated the results regarding FLT-1 and KDR expression. The results of our study suggest that the splice forms VEGF121 and VEGF165 and the receptors KDR and FLT-1 of the known angiogenetic peptide VEGF play a role in process of endochondral ossification.


Assuntos
Cartilagem/química , Condrócitos/química , Fatores de Crescimento Endotelial/análise , Lâmina de Crescimento/citologia , Linfocinas/análise , Adolescente , Processamento Alternativo , Cartilagem/embriologia , Fatores de Crescimento Endotelial/genética , Humanos , Imuno-Histoquímica/métodos , Lactente , Linfocinas/genética , Isoformas de Proteínas/análise , Isoformas de Proteínas/genética , Proteínas Proto-Oncogênicas/análise , RNA Mensageiro/análise , Receptores Proteína Tirosina Quinases/análise , Receptores de Fatores de Crescimento/análise , Receptores de Fatores de Crescimento do Endotélio Vascular , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tíbia/embriologia , Tíbia/crescimento & desenvolvimento , Fator A de Crescimento do Endotélio Vascular , Receptor 1 de Fatores de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA