Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Cell Physiol ; 227(7): 2936-46, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21959563

RESUMO

Osteoblast differentiation is regulated by the presence of collagen type I (COL I) extracellular matrix (ECM). We have recently demonstrated that Factor XIIIA (FXIIIA) transglutaminase (TG) is required by osteoblasts for COL I secretion and extracellular deposition, and thus also for osteoblast differentiation. In this study we have further investigated the link between COL I and FXIIIA, and demonstrate that COL I matrix increases FXIIIA levels in osteoblast cultures and that FXIIIA is found as cellular (cFXIIIA) and extacellular matrix (ecmFXIIIA) forms. FXIIIA mRNA, protein expression, cellular localization and secretion were enhanced by ascorbic acid (AA) treatment and blocked by dihydroxyproline (DHP) which inhibits COL I externalization. FXIIIA mRNA was regulated by the MAP kinase pathway. Secretion of ecmFXIIIA, and its enzymatic activity in conditioned medium, were also decreased in osteoblasts treated with the lysyl oxidase inhibitor ß-aminopropionitrile, which resulted in a loosely packed COL I matrix. Osteoblasts secrete a latent, inactive dimeric ecmFXIIIA form which is activated upon binding to the matrix. Monodansyl cadaverine labeling of TG substrates in the cultures revealed that incorporation of the label occurred at sites where fibronectin co-localized with COL I, indicating that ecmFXIIIA secretion could function to stabilize newly deposited matrix. Our results suggest that FXIIIA is an integral part of the COL I deposition machinery, and also that it is part of the ECM-feedback loop, both of which regulate matrix deposition and osteoblast differentiation.


Assuntos
Colágeno Tipo I/metabolismo , Fator XIIIa/metabolismo , Sistema de Sinalização das MAP Quinases , Osteoblastos/metabolismo , Transglutaminases/biossíntese , Aminopropionitrilo/farmacologia , Animais , Ácido Ascórbico/metabolismo , Diferenciação Celular/fisiologia , Células Cultivadas , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Fator XIIIa/genética , Fibronectinas/metabolismo , Camundongos , Osteoblastos/enzimologia , Proteína-Lisina 6-Oxidase/antagonistas & inibidores , RNA Mensageiro/genética , Transglutaminases/genética , Transglutaminases/metabolismo
2.
J Histochem Cytochem ; 55(7): 675-85, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17341477

RESUMO

Transglutaminases (TGs) are protein crosslinking enzymes involved in cell adhesion and signaling and matrix stabilization and maturation, in many cell types and tissues. We previously described that in addition to transglutaminase 2 (TG2), cultured MC3T3-E1 osteoblasts also express the plasma TG Factor XIIIA (FXIIIA). Here we report on the expression and localization of FXIIIA in bone in vivo and provide confirmatory in vitro data. Immunohistochemistry and in situ hybridization demonstrated that FXIIIA is expressed by osteoblasts and osteocytes in long bones formed by endochondral ossification (femur) and flat bones formed primarily by intramembranous ossification (calvaria and mandible). FXIIIA immunoreactivity was localized to osteoblasts, osteocytes, and the osteoid. RT-PCR analysis revealed FXIIIA expression by both primary osteoblasts and by the MC3T3-E1 osteoblast cell line. Western blot analysis of bone and MC3T3-E1 culture extracts demonstrated that FXIIIA is produced mainly as a small, 37-kDa form. Sequential RT-PCR analysis using overlapping PCR primers spanning the full FXIIIA gene showed that the entire FXIIIA gene is expressed, thus indicating that the 37-kDa FXIIIA is not a splice variant but a product of posttranslational proteolytic processing. Forskolin inhibition of osteoblast differentiation revealed that FXIIIA processing is regulated by the protein kinase A pathway.


Assuntos
Osso e Ossos/enzimologia , Fator XIIIa/metabolismo , Animais , Diferenciação Celular , Linhagem Celular , Proteínas Quinases Dependentes de AMP Cíclico/fisiologia , Fator XIIIa/biossíntese , Fator XIIIa/genética , Fêmur/enzimologia , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Osteoblastos/citologia , Osteoblastos/enzimologia , Osteócitos/enzimologia , RNA Mensageiro/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Tíbia/enzimologia , Extratos de Tecidos/metabolismo
3.
Matrix Biol ; 25(3): 135-48, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16469487

RESUMO

Transglutaminase (TG) enzymes and protein crosslinking have long been implicated in the formation of mineralized tissues. The aim of this study was to analyze the expression, activity and function of TGs in differentiating osteoblasts to gain further insight into the role of extracellular matrix protein crosslinking in bone formation. MC3T3-E1 (subclone 14) pre-osteoblast cultures were treated with ascorbic acid and beta-glycerophosphate to induce cell differentiation and matrix mineralization. Expression of TG isoforms was analyzed by RT-PCR. TG activity was assessed during osteoblast differentiation by in vitro biochemical assays and by in situ labeling of live cell cultures. We demonstrate that MC3T3-E1/C14 osteoblasts express two TG isoforms--TG2 and FXIIIA. Abundant TG activity was observed during cell differentiation which increased significantly after thrombin treatment, a result confirming the presence of FXIIIA in the cultures. Ascorbic acid treatment, which stimulated collagen secretion and assembly, also stimulated externalization of TG activity, likely from FXIIIA which was externalized upon this treatment as analyzed by immunofluoresence microscopy. Inhibition of TG activity in the cultures by cystamine resulted in complete abrogation of mineralization, attributable to decreased matrix accumulation and an arrested state of osteoblast differentiation as measured by decreased levels of bone sialoprotein, osteocalcin and alkaline phosphatase. Additional functional studies and substrate characterization showed that TG activity was required for the formation of a fibronectin-collagen network during the early stages of matrix formation and assembly. This network, in turn, appeared to be essential for further matrix production and progression of the osteoblast differentiation program, and ultimately for mineralization.


Assuntos
Diferenciação Celular/fisiologia , Matriz Extracelular/metabolismo , Fator XIIIa/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Isoenzimas/metabolismo , Osteoblastos/enzimologia , Osteoblastos/fisiologia , Transglutaminases/metabolismo , Células 3T3 , Animais , Calcificação Fisiológica/fisiologia , Linhagem Celular , Colágeno Tipo I/metabolismo , Meios de Cultivo Condicionados/química , Matriz Extracelular/química , Fator XIIIa/genética , Fibronectinas/metabolismo , Proteínas de Ligação ao GTP/genética , Isoenzimas/genética , Camundongos , Osteoblastos/citologia , Proteína 2 Glutamina gama-Glutamiltransferase , Transglutaminases/genética
4.
PLoS One ; 6(1): e15893, 2011 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-21283799

RESUMO

Transglutaminase activity, arising potentially from transglutaminase 2 (TG2) and Factor XIIIA (FXIIIA), has been linked to osteoblast differentiation where it is required for type I collagen and fibronectin matrix deposition. In this study we have used an irreversible TG-inhibitor to 'block -and-track' enzyme(s) targeted during osteoblast differentiation. We show that the irreversible TG-inhibitor is highly potent in inhibiting osteoblast differentiation and mineralization and reduces secretion of both fibronectin and type I collagen and their release from the cell surface. Tracking of the dansyl probe by Western blotting and immunofluorescence microscopy demonstrated that the inhibitor targets plasma membrane-associated FXIIIA. TG2 appears not to contribute to crosslinking activity on the osteoblast surface. Inhibition of FXIIIA with NC9 resulted in defective secretory vesicle delivery to the plasma membrane which was attributable to a disorganized microtubule network and decreased microtubule association with the plasma membrane. NC9 inhibition of FXIIIA resulted in destabilization of microtubules as assessed by cellular Glu-tubulin levels. Furthermore, NC9 blocked modification of Glu-tubulin into 150 kDa high-molecular weight Glu-tubulin form which was specifically localized to the plasma membrane. FXIIIA enzyme and its crosslinking activity were colocalized with plasma membrane-associated tubulin, and thus, it appears that FXIIIA crosslinking activity is directed towards stabilizing the interaction of microtubules with the plasma membrane. Our work provides the first mechanistic cues as to how transglutaminase activity could affect protein secretion and matrix deposition in osteoblasts and suggests a novel function for plasma membrane FXIIIA in microtubule dynamics.


Assuntos
Matriz Extracelular/metabolismo , Fator XIII/metabolismo , Microtúbulos/fisiologia , Osteoblastos/citologia , Osteogênese , Células 3T3 , Animais , Diferenciação Celular , Membrana Celular/enzimologia , Membrana Celular/metabolismo , Fator XIII/fisiologia , Fator XIIIa/metabolismo , Proteínas de Ligação ao GTP , Camundongos , Proteína 2 Glutamina gama-Glutamiltransferase , Transglutaminases
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA