Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Nat Genet ; 32(4): 639-44, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12434156

RESUMO

Runx2 (runt-related transcription factor 2, also known as Cbfa1, Osf2 and AML3) is essential for bone development in mice, and mutations in RUNX2 are found in 65-80% of individuals with cleidocranial dysplasia. Although all Runx family members can interact with Cbfbeta (core-binding factor b, encoded by Cbfb), a role for Cbfbeta in bone development has not been demonstrated owing to lethality in Cbfb(-/-) mouse embryos at 12.5 days post coitum (d.p.c.) from hemorrhages and lack of definitive hematopoiesis. Using a 'knock-in' strategy, we generated mouse embryonic stem (ES) cells that express Cbfb fused in-frame to a cDNA encoding green fluorescent protein (GFP). Cbfb(+/GFP) mice had normal life spans and appeared normal, but Cbfb(GFP/GFP) pups died within the first day after birth. The Cbfb(GFP/GFP) mice exhibited a delay in endochondral and intramembranous ossification as well as in chondrocyte differentiation, similar to but less severe than delays observed in Runx2(-/-) mice. We demonstrate that Cbfbeta is expressed in developing bone and forms a functional interaction with Runx2, and that Cbfb(GFP) is a hypomorphic allele. The fusion allele maintains sufficient function in hematopoietic cells to bypass the early embryonic lethality, and identifies a new role for Cbfb in bone development. Our findings raise the possibility that mutations in CBFB may be responsible for some cases of cleidocranial dysplasia that are not linked to mutations in RUNX2.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Ligação a DNA/fisiologia , Proteínas de Neoplasias , Osteogênese , Fatores de Transcrição/metabolismo , Fatores de Transcrição/fisiologia , Alelos , Animais , Diferenciação Celular , Células Cultivadas , Condrócitos/citologia , Condrócitos/fisiologia , Subunidade alfa 1 de Fator de Ligação ao Core , Subunidades alfa de Fatores de Ligação ao Core , Subunidade beta de Fator de Ligação ao Core , Fatores de Ligação ao Core , Proteínas de Ligação a DNA/genética , Desenvolvimento Embrionário e Fetal/genética , Regulação da Expressão Gênica no Desenvolvimento , Genes Letais , Proteínas de Fluorescência Verde , Células HeLa , Homozigoto , Humanos , Proteínas Luminescentes/metabolismo , Camundongos , Camundongos Transgênicos , Fenótipo , Células-Tronco Pluripotentes/fisiologia , Proteínas Recombinantes de Fusão/fisiologia , Fator de Transcrição AP-2 , Fatores de Transcrição/genética , Células Tumorais Cultivadas
2.
Trends Mol Med ; 24(11): 919-930, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30213702

RESUMO

The promise of tissue engineering and regenerative medicine to reduce the burden of disease and improve quality of life are widely acknowledged. Traditional tissue engineering and regenerative medicine approaches rely on generation of tissue constructs in vitro for subsequent transplantation or injection of exogenously manipulated cells into a host. While promising, few such therapies have succeeded in clinical practice. Here, we propose that recent advances in stem cell and developmental biology, immunology, bioengineering, and material sciences, position us to develop a new generation of in vivo regenerative medicine therapies, which we term autotherapies. Autotherapies are strategies based on optimizing endogenous tissue responses and capitalizing on manipulation of stem cell niches and endogenous tissue microenvironments to enhance tissue healing and regeneration.


Assuntos
Regeneração , Cicatrização , Animais , Linhagem da Célula/genética , Microambiente Celular , Reprogramação Celular/genética , Epigênese Genética , Matriz Extracelular/metabolismo , Humanos , Nicho de Células-Tronco , Células-Tronco/citologia , Células-Tronco/metabolismo
4.
J Bone Miner Res ; 22(10): 1479-91, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17663640

RESUMO

UNLABELLED: ONJ has been increasingly suspected to be a potential complication of bisphosphonate therapy in recent years. Thus, the ASBMR leadership appointed a multidisciplinary task force to address key questions related to case definition, epidemiology, risk factors, diagnostic imaging, clinical management, and future areas for research related to the disorder. This report summarizes the findings and recommendations of the task force. INTRODUCTION: The increasing recognition that use of bisphosphonates may be associated with osteonecrosis of the jaw (ONJ) led the leadership of the American Society for Bone and Mineral Research (ASBMR) to appoint a task force to address a number of key questions related to this disorder. MATERIALS AND METHODS: A multidisciplinary expert group reviewed all pertinent published data on bisphosphonate-associated ONJ. Food and Drug Administration drug adverse event reports were also reviewed. RESULTS AND CONCLUSIONS: A case definition was developed so that subsequent studies could report on the same condition. The task force defined ONJ as the presence of exposed bone in the maxillofacial region that did not heal within 8 wk after identification by a health care provider. Based on review of both published and unpublished data, the risk of ONJ associated with oral bisphosphonate therapy for osteoporosis seems to be low, estimated between 1 in 10,000 and <1 in 100,000 patient-treatment years. However, the task force recognized that information on incidence of ONJ is rapidly evolving and that the true incidence may be higher. The risk of ONJ in patients with cancer treated with high doses of intravenous bisphosphonates is clearly higher, in the range of 1-10 per 100 patients (depending on duration of therapy). In the future, improved diagnostic imaging modalities, such as optical coherence tomography or MRI combined with contrast agents and the manipulation of image planes, may identify patients at preclinical or early stages of the disease. Management is largely supportive. A research agenda aimed at filling the considerable gaps in knowledge regarding this disorder was also outlined.


Assuntos
Difosfonatos/efeitos adversos , Doenças Maxilomandibulares/induzido quimicamente , Doenças Maxilomandibulares/diagnóstico , Osteonecrose/induzido quimicamente , Osteonecrose/diagnóstico , Sociedades Médicas , Adulto , Idoso , Idoso de 80 Anos ou mais , América , Animais , Diagnóstico Diferencial , Difosfonatos/farmacologia , Feminino , Humanos , Doenças Maxilomandibulares/metabolismo , Masculino , Pessoa de Meia-Idade , Minerais/metabolismo , Osteonecrose/metabolismo , Fatores de Risco
6.
Int J Dev Biol ; 47(6): 423-31, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-14598792

RESUMO

Defects in the growth and development of the endochondral bones that comprise the cranial base contribute to several craniofacial dysmorphic syndromes. Since Bone Morphogenetic Protein (BMP) signaling regulates chondrocyte differentiation and endochondral ossification in developing long bones, we have tested the hypothesis that BMP signaling also participates in regulating development of the cranial base. During in vivo developmental progression of the cranial base in mice, a burst of skeletal growth and chondrocyte maturation was identified in the perinatal period. Using a novel serum-free organ culture system, cranial base structures were cultured as explants in the presence of BMP4 or noggin, and analyzed for morphological and molecular changes. Growth of perinatal cranial base explants was inhibited by treatment with noggin, a BMP inhibitor. Exogenous BMP4 promoted cartilage growth, matrix deposition and chondrocyte proliferation in a dose dependent manner. Correspondingly, expression level of the cartilage markers Sox9 and collagen type II were also increased. Alkaline phosphatase and collagen type X expression were up-regulated and expressed in ectopic hypertrophic chondrocytes after treatment of the cultures with 100 ng/ml BMP4 for seven days. This increase in chondrocyte hypertrophy was accompanied by increased indian hedgehog (Ihh) and parathyroid hormone/parathyroid hormone related peptide (PTH/PTHrP) receptor (PPR) expression, but not increased PTHrP expression. We conclude that endogenous BMPs are required to maintain cartilage growth, and exogenous BMP4 can enhance cartilage maturation and induce ectopic chondrocyte hypertrophy in the cranial base. Therefore, appropriate levels of BMP signaling are important for normal cranial base development.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Condrócitos/metabolismo , Base do Crânio/crescimento & desenvolvimento , Animais , Proteína Morfogenética Óssea 4 , Proteínas de Transporte , Cartilagem/citologia , Cartilagem/crescimento & desenvolvimento , Cartilagem/metabolismo , Divisão Celular/fisiologia , Condrócitos/citologia , Camundongos , Osteogênese/fisiologia , Proteínas/genética , Proteínas/metabolismo
7.
J Bone Joint Surg Am ; 85-A Suppl 3: 13-8, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-12925604

RESUMO

BACKGROUND: Bone morphogenetic proteins (BMPs) are pleiotropic differentiation factors that regulate cell fate determination by orchestrating the activities of downstream signal transducers. Although BMP ligands can elicit signal transduction from heterodimeric combinations of several type-I and type-II receptors, cytoplasmic transducers of the BMP signal include only three known BMP-specific regulatory Smad proteins: Smad1, 5, and 8. In order to determine the combination of signals that regulate chondrogenesis by BMPs, we analyzed the functions of BMP Smad subtypes. METHODS: Multipotential mesenchymal C3H10T1/2 cells and monopotential chondroprogenitor MC615 cells were placed in micromass culture in the presence or absence of BMP4. Chondrogenic differentiation was assayed by measuring Sox9 and type-II collagen gene expression and by alcian blue staining. Transactivation of type-II collagen by regulatory Smads singly, or in combination with Smad4, which partners with regulatory Smads, was assayed by luciferase activity. RESULTS: In the absence of BMP4, mesenchymal cells did not exhibit chondrogenic differentiation, whereas chondroprogenitor cells showed increased cartilage marker expression. In the presence of BMP4, the rate and extent of chondrogenesis increased in a dose-dependent manner for both cell types. We further determined that Smad1 or Smad5, but not Smad8, synergized with Smad4 in the transactivation of the type-II collagen promoter in chondroprogenitor cells. In contrast, Smad8 and Smad4 presented modest synergy in mesenchymal cells. CONCLUSIONS: Taken together, our data suggest that uncommitted mesenchymal cells do not have the cellular competence to respond to the rate-limiting chondroinductive factor BMP. However, in chondroprogenitor cells, BMP stimulates differentiation through mechanisms mediated by Smad1 or Smad5 in combination with Smad4.


Assuntos
Proteínas Morfogenéticas Ósseas/fisiologia , Diferenciação Celular/fisiologia , Condrócitos/citologia , Proteínas de Ligação a DNA/fisiologia , Mesoderma/citologia , Transdução de Sinais/fisiologia , Células-Tronco/citologia , Transativadores/fisiologia , Animais , Linhagem Celular , Humanos , Camundongos , Camundongos Endogâmicos C3H , Fosfoproteínas/fisiologia , Proteínas Smad , Proteína Smad1 , Proteína Smad4 , Proteína Smad5 , Engenharia Tecidual
9.
Bone ; 48(4): 847-56, 2011 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-21129456

RESUMO

Activation of osteoblastic bone anabolism in the calvarial sutures is considered to be the essential pathologic condition underlying mutant FGFR2-related craniofacial dysostosis. However, early clinical investigations indicated that abnormal cartilage development in the cranial base was rather a primary site of abnormal feature in Apert Syndrome (AS). To examine the significance of cartilaginous growth of the cranial base in AS, we generated a transgenic mouse bearing AS-type mutant Fgfr2IIIc under the control of the Col2a1 promoter-enhancer (Fgfr2IIIc(P253R) mouse). Despite the lacking expression of Fgfr2IIIc(P253R) in osteoblasts, exclusive disruption of chondrocytic differentiation and growth reproduced AS-like acrocephaly accompanied by short anterior cranial base with fusion of the cranial base synchondroses, maxillary hypoplasia and synostosis of the calvarial sutures with no significant abnormalities in the trunk and extremities. Gene expression analyses demonstrated upregulation of p21, Ihh and Mmp-13 accompanied by modest increase in expression of Sox9 and Runx2, indicating acceleration of chondrocytic maturation and hypertrophy in the cranial base of the Fgfr2IIIc(P253R) mice. Furthermore, an acquired affinity and specificity of mutant FGFR2IIIc(P253R) receptor with FGF2 and FGF10 is suggested as a mechanism of activation of FGFR2 signaling selectively in the cranial base. In this report, we strongly suggest that the acrocephalic feature of AS is not alone a result of the coronal suture synostosis, but is a result of the primary disturbance in growth of the cranial base with precocious endochondral ossification.


Assuntos
Acrocefalossindactilia/patologia , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais , Acrocefalossindactilia/metabolismo , Animais , Sequência de Bases , Primers do DNA , Hibridização In Situ , Masculino , Camundongos , Camundongos Transgênicos , Mutagênese Sítio-Dirigida , Reação em Cadeia da Polimerase , Transgenes
10.
Eur J Oral Sci ; 114 Suppl 1: 169-77; discussion 201-2, 381, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16674681

RESUMO

Amelogenin proteins are essential in the control of enamel biomineralization and the amelogenin gene therefore is spatiotemporally regulated to ensure proper amelogenin protein expression. In this study, we examined the role of sumoylation to alter CCAAT/enhancer-binding protein alpha (C/EBPalpha) activity, and performed a search using a protein/DNA array system for other proteins that act co-operatively with C/EBPalpha to alter amelogenin expression. We observed that C/EBPalpha was modified by sumoylation, and that this modification played an indirect inhibitory role on the regulation of C/EBPalpha activity which appeared to act through other transcription factors. The protein/DNA array allowed us to single out the transcription factor, YY1, which acts in the absence of direct DNA binding to repress both the basal amelogenin promoter activity and C/EBPalpha-mediated transactivation. Taken together, these pathways may account for part of the physiological modulation of the amelogenin gene expression in accordance with tooth developmental and enamel biomineralization requirements.


Assuntos
Proteínas do Esmalte Dentário/genética , Proteína SUMO-1/genética , Fatores de Transcrição/genética , Fator de Transcrição YY1/genética , Amelogênese/genética , Amelogenina , Animais , Western Blotting , Proteína alfa Estimuladora de Ligação a CCAAT/genética , Esmalte Dentário/metabolismo , Regulação da Expressão Gênica/genética , Camundongos , Microscopia de Fluorescência , Odontogênese/genética , Análise de Sequência com Séries de Oligonucleotídeos , Regiões Promotoras Genéticas/genética , Ativação Transcricional/genética
11.
Birth Defects Res A Clin Mol Teratol ; 73(12): 956-65, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16323168

RESUMO

BACKGROUND: Cleft lip with or without cleft palate is the most common congenital anomaly in the craniofacial region. Knowledge of the molecular mechanisms behind normal lip fusion can contribute to better intervention and improved functional clinical outcome. Transforming growth factor-beta3 (TGF-beta3) has been implicated in lip morphogenesis. Therefore, we hypothesized that TGF-beta3 functions during lip fusion through regulation of angiogenesis and mesenchymal cell cycle progression during early developmental stages. METHODS: To test this hypothesis we used the CL/Fraser mouse model, which has a high incidence of cleft lip. Lips isolated from embryonic day (ED) 11.5 mouse embryos were allowed to develop in serum-free organ cultures in the presence or absence of TGF-beta3. The lips that developed in these cultures fused in 2 days. RESULTS: During normal development, we detected positive immunoreactions for TGF-beta3 at the site of fusion. We also detected mesenchymal cells that were immunopositive for Flk-1 and CD31, which are markers for endothelial cell precursors. Exogenous TGF-beta3 accelerated lip fusion in culture. This enhancement was associated with an increase in the number of capillary blood vessels in the lips cultured in the presence of TGF-beta3, in comparison with controls. In tandem, TGF-beta3 increased the level of expression of both Flk-1 and CD31. Our data suggest that an elevated level of TGF-beta3 may promote angiogenesis in developing lips that is mediated by increased Flk-1 and CD31 expression. We also detected increased cyclin D1 expression (a marker for cell proliferation) in the presence of TGF-beta3, which suggests that TGF-beta3 promoted cell proliferation. CONCLUSIONS: TGF-beta3 promoted cell proliferation and angiogenesis in lip mesenchymal tissues. These events led to enhanced lip fusion in the presence of TGF-beta3.


Assuntos
Fenda Labial , Ciclina D1/genética , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Neovascularização Fisiológica/efeitos dos fármacos , Molécula-1 de Adesão Celular Endotelial a Plaquetas/genética , Fator de Crescimento Transformador beta/farmacologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Animais , Proliferação de Células/efeitos dos fármacos , Fenda Labial/embriologia , Fenda Labial/genética , Fenda Labial/metabolismo , Ciclina D1/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Mesoderma/citologia , Mesoderma/efeitos dos fármacos , Camundongos , Camundongos Mutantes , Neovascularização Fisiológica/genética , Técnicas de Cultura de Órgãos , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Crescimento Transformador beta3 , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
12.
Differentiation ; 73(5): 233-9, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-16026545

RESUMO

The molecular mechanisms by which the primordia of the midface grow and fuse to form the primary palate are not well characterized. This is in spite of the fact that failure of growth and/or fusion of these facial primordia leads to the common human craniofacial birth defects, clefts of the lip with or without clefts of the palate. Members of the transforming growth factor beta (Tgfbeta) superfamily have been shown to play critical roles during craniofacial development. Specifically, the role of Tgfbeta-3 in mediating the fusion of the embryonic secondary palatal shelves is well documented. In a screen for genes expressed during fusion of the murine midfacial processes, Tgfbeta2 was identified as a gene differentially expressed during fusion of the lateral and medial nasal processes. The objective of our study was to analyze the spatial and temporal expression of Tgfbeta2 during critical stages of midfacial morphogenesis at both the transcript and protein levels. We also compared the pattern of expression of Tgfbeta2 with that of Bmp4, a gene shown previously to be involved in mediating the fusion process in the midface. Our results showed Tgfbeta2 expression in a very restrictive area of the epithelial layer along the borders of the midfacial primordia, in a pattern very similar to that of Bmp4. The highly restrictive and spatial and temporal pattern of expression of Tgfbeta2 implicates its role in mediating the fusion of the midfacial processes, possibly through interacting with Bmp4 in the regulation of apoptosis and/or epithelial-mesenchymal transformation. A greater understanding of the role of this gene will clarify how the normal midface grows and the mechanisms behind cleft development.


Assuntos
Palato/embriologia , Fator de Crescimento Transformador beta/biossíntese , Fator de Crescimento Transformador beta/genética , Animais , Proteína Morfogenética Óssea 4 , Proteínas Morfogenéticas Ósseas/biossíntese , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/fisiologia , Feminino , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/metabolismo , Fator de Crescimento Transformador beta/fisiologia , Fator de Crescimento Transformador beta2
13.
Arthritis Res ; 4(2): 94-106, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-11879545

RESUMO

Cartilage serves multiple functions in the developing embryo and in postnatal life. Genetic mutations affecting cartilage development are relatively common and lead to skeletal malformations, dysfunction or increased susceptibility to disease or injury. Characterization of these mutations and investigation of the molecular pathways in which these genes function have contributed to an understanding of the mechanisms regulating skeletal patterning, chondrogenesis, endochondral ossification and joint formation. Extracellular growth and differentiation factors including bone morphogenetic proteins, fibroblast growth factors, parathyroid hormone-related peptide, extracellular matrix components, and members of the hedgehog and Wnt families provide important signals for the regulation of cell proliferation, differentiation and apoptosis. Transduction of these signals within the developing mesenchymal cells and chondrocytes results in changes in gene expression mediated by transcription factors including Smads, Msx2, Sox9, signal transducer and activator of transcription (STAT), and core-binding factor alpha 1. Further investigation of the interactions of these signaling pathways will contribute to an understanding of cartilage growth and development, and will allow for the development of strategies for the early detection, prevention and treatment of diseases and disorders affecting the skeleton.


Assuntos
Desenvolvimento Ósseo/genética , Osso e Ossos/fisiologia , Cartilagem/embriologia , Cartilagem/crescimento & desenvolvimento , Condrócitos/fisiologia , Doenças do Desenvolvimento Ósseo/genética , Osso e Ossos/embriologia , Cartilagem/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Substâncias de Crescimento/metabolismo , Humanos , Mutação , Transdução de Sinais , Fatores de Transcrição/genética
14.
J Cell Biochem ; 91(6): 1204-17, 2004 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15048875

RESUMO

Bone morphogenetic protein 4 (BMP4) and growth/differentiation factor 5 (GDF5) are closely related protein family members and regulate early cartilage patterning and differentiation. In this study, we compared the functional outcome of their actions systematically at various stages of chondrogenesis in mouse embryonic limb bud mesenchyme grown in micromass cultures. Overall, both growth factors enhanced cartilage growth and differentiation in these cultures. Uniquely, BMP4 not only accelerated the formation and maturation of cartilaginous nodules, but also induced internodular mesenchymal cells to express cartilage differentiation markers. On the other hand, GDF5 increased the number of prechondrogenic mesenchymal cell condensation and cartilaginous nodules, without altering the overall pattern of differentiation. In addition, GDF5 caused a more sustained elevated expression level of Sox9 relative to that associated with BMP4. BMP4 accelerated chondrocyte maturation throughout the cultures and sustained an elevated level of Col10 expression, whereas GDF5 caused a transient increase in Col10 expression. Taken together, we conclude that BMP4 is instructive to chondrogenesis and induces mesenchymal cells toward the chondrogenic lineage. Furthermore, BMP4 accelerates the progression of cartilage differentiation to maturation. GDF5 enhances cartilage formation by promoting chondroprogenitor cell aggregation, and amplifying the responses of cartilage differentiation markers. These differences may serve to fine-tune the normal cartilage differentiation program, and can be exploited for the molecular manipulation in biomimetics.


Assuntos
Proteínas Morfogenéticas Ósseas/farmacologia , Condrogênese/efeitos dos fármacos , Animais , Proteína Morfogenética Óssea 4 , Proteínas Morfogenéticas Ósseas/metabolismo , Cartilagem/citologia , Cartilagem/embriologia , Cartilagem/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Condrócitos/citologia , Condrócitos/metabolismo , Condrogênese/fisiologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Fator 5 de Diferenciação de Crescimento , Proteínas de Grupo de Alta Mobilidade/metabolismo , Membro Posterior/embriologia , Camundongos , Gravidez , Fatores de Transcrição SOX9 , Fatores de Transcrição/metabolismo
15.
Birth Defects Res C Embryo Today ; 69(2): 102-22, 2003 May.
Artigo em Inglês | MEDLINE | ID: mdl-12955856

RESUMO

Cartilage patterning and differentiation are prerequisites for skeletal development through endochondral ossification (EO). Multipotential mesenchymal cells undergo a complex process of cell fate determination to become chondroprogenitors and eventually differentiate into chondrocytes. These developmental processes require the orchestration of cell-cell and cell-matrix interactions. In this review, we present limb bud development as a model for cartilage patterning and differentiation. We summarize the molecular and cellular events and signaling pathways for axis patterning, cell condensation, cell fate determination, digit formation, interdigital apoptosis, EO, and joint formation. The interconnected nature of these pathways underscores the effects of genetic and teratogenic perturbations that result in skeletal birth defects. The topics reviewed also include limb dysmorphogenesis as a result of genetic disorders and environmental factors, including FGFR, GLI3, GDF5/CDMP1, Sox9, and Cbfa1 mutations, as well as thalidomide- and alcohol-induced malformations. Understanding the complex interactions involved in cartilage development and EO provides insight into mechanisms underlying the biology of normal cartilage, congenital disorders, and pathologic adult cartilage.


Assuntos
Padronização Corporal/fisiologia , Desenvolvimento Ósseo/fisiologia , Osso e Ossos/anormalidades , Extremidades/embriologia , Animais , Condrogênese/fisiologia , Humanos , Osteogênese
16.
Osteoarthritis Cartilage ; 12(12): 963-73, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15564063

RESUMO

OBJECTIVE: To understand the molecular mechanisms that lead to increased MMP-13 expression and cartilage degeneration during the progression of osteoarthritis (OA), we have investigated the expression of the transcription factor RUNX2 in OA cartilage and the regulation of MMP-13 expression by RUNX2 and FGF2 in articular chondrocytes. DESIGN: RUNX2 and MMP-13 expression in human OA and control cartilage was analyzed by immunohistochemistry. The effects of RUNX2 over-expression, with or without FGF2 treatment, on MMP-13 promoter activity and enzyme accumulation were measured in articular chondrocytes. Inhibitors of MEK/ERK were assayed for their ability to block FGF2 and RUNX2 up-regulation of the MMP-13 promoter. We analyzed RUNX2 phosphorylation in response to FGF2. RESULTS: Fibrillated OA cartilage exhibited increased RUNX2 immunoreactivity when compared to control cartilage. RUNX2 co-localized with MMP-13 in clusters of chondrocytes in fibrillated OA cartilage. RUNX2 over-expression in cultured chondrocytes increased their responsiveness to FGF2 treatment, which led to increased MMP-13 expression. Inhibitors of MEK/ERK signaling blocked up-regulation of the MMP-13 promoter by RUNX2 and FGF2, and also blocked the activation of RUNX2 by FGF2. FGF2 treatment of articular chondrocytes increased RUNX2 phosphorylation approximately 2-fold. CONCLUSIONS: Increased expression of RUNX2 in OA cartilage may contribute to increased expression of MMP-13. FGF2, which is present in OA synovial fluid, activated RUNX2 via the MEK/ERK pathway and increased MMP-13 expression. However, it is unlikely that RUNX2 is a substrate of ERK1/2. RUNX2 expression and activation may be a significant step in the progression of OA by promoting changes in gene expression and chondrocyte differentiation.


Assuntos
Cartilagem Articular/metabolismo , Colagenases/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Proteínas de Neoplasias/metabolismo , Osteoartrite do Joelho/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Animais , Western Blotting/métodos , Butadienos/farmacologia , Cartilagem Articular/química , Cartilagem Articular/efeitos dos fármacos , Estudos de Casos e Controles , Bovinos , Células Cultivadas , Condrócitos/química , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Colagenases/análise , Subunidade alfa 1 de Fator de Ligação ao Core , Flavonoides/farmacologia , Humanos , Imunoprecipitação/métodos , Metaloproteinase 13 da Matriz , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Naftalenos/farmacologia , Proteínas de Neoplasias/análise , Nitrilas/farmacologia , Fosforilação , Proteína Quinase C/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição/análise
17.
J Biol Chem ; 279(48): 50382-90, 2004 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-15342645

RESUMO

The family of UDP-GalNAc:polypeptide N-acetylgalactosaminyltransferases (ppGaNTases) is responsible for initiating mucin-type O-linked glycosylation in higher eukaryotes. To begin to examine the biological role of O-linked glycosylation, mammalian cells were treated with a small molecule inhibitor (designated 1-68A, Ref. 15) of ppGaNTase activity. NIH3T3 cells exposed to the inhibitor were shown to undergo a significant reduction in cell surface O-glycosylation as detected by staining with jacalin and peanut agglutinin lectins after 30 min of treatment; no reduction in staining using antibodies to O-linked N-acetylglucosamine or the lectin concanavalin A was detected. Apoptosis was also observed in treated cells after 45 min of exposure, ostensibly following the O-glycosylation reduction. Overexpression of several different ppGaNTase isoforms restored cell surface O-glycosylation and rescued inhibitor-induced apoptosis. Additionally, mouse embryonic mandibular organ cultures exposed to 1-68A developed abnormally, presumably because of epithelial and mesenchymal apoptosis that followed a reduction in jacalin and peanut agglutinin staining. Our studies suggest that mucin-type O-linked glycosylation may be required for normal development and that ppGaNTases may play a role in the regulation of apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Mandíbula/embriologia , N-Acetilgalactosaminiltransferases/antagonistas & inibidores , Pirogalol/análogos & derivados , Pirogalol/farmacologia , Uridina/análogos & derivados , Uridina/farmacologia , Animais , Glicosilação/efeitos dos fármacos , Lectinas/metabolismo , Mandíbula/efeitos dos fármacos , Mandíbula/metabolismo , Camundongos , Células NIH 3T3
18.
J Biol Chem ; 278(37): 35743-8, 2003 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-12851394

RESUMO

Amelogenins, major components of developing enamel, are predominantly involved in the formation of tooth enamel. Although amelogenins are also implicated in cementogenesis, their precise spatial expression pattern and molecular role are not clearly understood. Here, we report for the first time the expression of two alternate splice forms of amelogenins, M180 and the leucine-rich amelogenin peptide (LRAP), in the periodontal region of mouse tooth roots. Lack of M180 and LRAP mRNA expression correlated with cementum defects observed in the amelogenin-null mice. The cementum defects were characterized by an increased presence of multinucleated cells, osteoclasts, and cementicles. These defects were associated with an increased expression of the receptor activator of the nuclear factor-kappa B ligand (RANKL), a critical regulator of osteoclastogenesis. These findings indicate that the amelogenin splice variants, M180 and LRAP, are critical in preventing abnormal resorption of cementum.


Assuntos
Processamento Alternativo , Proteínas de Transporte/fisiologia , Cemento Dentário/patologia , Proteínas do Esmalte Dentário/deficiência , Proteínas do Esmalte Dentário/genética , Glicoproteínas de Membrana/fisiologia , Amelogenina , Animais , Primers do DNA , Camundongos , Camundongos Knockout , Dente Molar/patologia , Osteoclastos/patologia , Ligante RANK , Receptor Ativador de Fator Nuclear kappa-B , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA