Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Stem Cells ; 30(2): 304-13, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22102178

RESUMO

Delta like-1 (Dlk1)/preadipocyte factor-1 (Pref-1)/fetal antigen-1 (FA1) is a novel surface marker for embryonic chondroprogenitor cells undergoing lineage progression from proliferation to prehypertrophic stages. However, mechanisms mediating control of its expression during chondrogenesis are not known. Thus, we examined the effect of a number of signaling molecules and their inhibitors on Dlk1 expression during in vitro chondrogenic differentiation in mouse embryonic limb bud mesenchymal micromass cultures and mouse embryonic fibroblast (MEF) pellet cultures. Dlk1/Pref-1 was initially expressed during mesenchymal condensation and chondrocyte proliferation, in parallel with expression of Sox9 and Col2a1, and was downregulated upon the expression of Col10a1 by hypertrophic chondrocytes. Among a number of molecules that affected chondrogenesis, transforming growth factor-ß1 (TGF-ß1)-induced proliferation of chondroprogenitors was associated with decreased Dlk1 expression. This effect was abolished by TGF-ß signaling inhibitor SB431542, suggesting regulation of Dlk1/FA1 by TGF-ß1 signaling in chondrogenesis. TGF-ß1-induced Smad phosphorylation and chondrogenesis were significantly increased in Dlk1(-/-) MEF, while they were blocked in Dlk1 overexpressing MEF, in comparison with wild-type MEF. Furthermore, overexpression of Dlk1 or addition of its secreted form FA1 dramatically inhibited TGF-ß1-induced Smad reporter activity. In conclusion, our data identified Dlk1/FA1 as a downstream target of TGF-ß1 signaling molecule that mediates its function in embryonic chondrogenesis. The crosstalk between TGF-ß1 and Dlk1/FA1 was shown to promote early chondrogenesis during the embryonic endochondral ossification process.


Assuntos
Condrogênese , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Proteínas de Membrana/metabolismo , Osteogênese , Fator de Crescimento Transformador beta1/fisiologia , Animais , Proteínas de Ligação ao Cálcio , Diferenciação Celular , Células Cultivadas , Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Botões de Extremidades/citologia , Botões de Extremidades/metabolismo , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fosforilação , Transdução de Sinais , Proteína Smad2/metabolismo , Fator de Crescimento Transformador beta1/farmacologia
2.
J Bone Miner Res ; 32(5): 913-926, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28019696

RESUMO

We identified the neuroprotein collapsing response mediator protein-4 (CRMP4) as a noncanonical osteogenic factor that regulates the differentiation of mouse bone marrow skeletal stem cells (bone marrow stromal stem cells [mBMSCs]) into osteoblastic cells. CRMP4 is the only member of the CRMP1-CRMP5 family to be expressed by mBMSCs and in osteoprogenitors of both adult mouse and human bones. In vitro gain-of-function and loss-of-function of CRMP4 in murine stromal cells revealed its inhibitory effect on osteoblast differentiation. In addition, Crmp4-deficient mice (Crmp4-/- ) displayed a 40% increase in bone mass, increased mineral apposition rate, and bone formation rate, compared to wild-type controls. Increased bone mass in Crmp4-/- mice was associated with enhanced BMP2 signaling and BMP2-induced osteoblast differentiation in Crmp4-/- osteoblasts (OBs). Furthermore, Crmp4-/- OBs exhibited enhanced activation of RhoA/focal adhesion kinase (FAK) signaling that led to cytoskeletal changes with increased cell spreading. In addition, Crmp4-/- OBs exhibited increased cell proliferation that was mediated via inhibiting cyclin-dependent kinase inhibitor 1B, p27Kip1 and upregulating cyclin D1 expression which are targets of RhoA signaling pathway. Our findings identify CRMP4 as a novel negative regulator of osteoblast differentiation. © 2016 American Society for Bone and Mineral Research.


Assuntos
Proteína Morfogenética Óssea 2/metabolismo , Proteínas Musculares/metabolismo , Osteoblastos/metabolismo , Osteogênese , Transdução de Sinais/fisiologia , Proteínas rho de Ligação ao GTP/metabolismo , Animais , Proteína Morfogenética Óssea 2/genética , Proliferação de Células/genética , Inibidor de Quinase Dependente de Ciclina p27/genética , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Quinase 1 de Adesão Focal/genética , Quinase 1 de Adesão Focal/metabolismo , Camundongos , Camundongos Knockout , Proteínas Musculares/genética , Osteoblastos/citologia , Células Estromais/citologia , Células Estromais/metabolismo , Proteínas rho de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP
3.
Stem Cell Res Ther ; 6: 196, 2015 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-26450135

RESUMO

INTRODUCTION: There is a clinical need for developing systemic transplantation protocols for use of human skeletal stem cells (also known bone marrow stromal stem cells) (hBMSC) in tissue regeneration. In systemic transplantation studies, only a limited number of hBMSC home to injured tissues suggesting that only a subpopulation of hBMSC possesses "homing" capacity. Thus, we tested the hypothesis that a subpopulation of hBMSC defined by ability to form heterotopic bone in vivo, is capable of homing to injured bone. METHODS: We tested ex vivo and in vivo homing capacity of a number of clonal cell populations derived from telomerized hBMSC (hBMSC-TERT) with variable ability to form heterotopic bone when implanted subcutaneously in immune deficient mice. In vitro transwell migration assay was used and the in vivo homing ability of transplanted hBMSC to bone fractures in mice was visualized by bioluminescence imaging (BLI). In order to identify the molecular phenotype associated with enhanced migration, we carried out comparative DNA microarray analysis of gene expression of hBMSC-derived high bone forming (HBF) clones versus low bone forming (LBF) clones. RESULTS: HBF clones were exhibited higher ex vivo transwell migration and following intravenous injection, better in vivo homing ability to bone fracture when compared to LBF clones. Comparative microarray analysis of HBF versus LBF clones identified enrichment of gene categories of chemo-attraction, adhesion and migration associated genes. Among these, platelet-derived growth factor receptor (PDGFR) α and ß were highly expressed in HBF clones. Follow up studies showed that the chemoattractant effects of PDGF in vitro was more enhanced in HBF compared to LBF clones and this effect was reduced in presence of a PDGFRß-specific inhibitor: SU-16 f. Also, PDGF exerted greater chemoattractant effect on PDGFRß(+) cells sorted from LBF clones compared to PDGFRß(-) cells. CONCLUSION: Our data demonstrate phenotypic and molecular association between in vivo bone forming ability and migratory capacity of hBMSC. PDGFRß can be used as a potential marker for the prospective selection of hBMSC populations with high migration and bone formation capacities suitable for clinical trials for enhancing bone regeneration.


Assuntos
Diferenciação Celular , Movimento Celular , Células-Tronco Mesenquimais/fisiologia , Adipócitos/fisiologia , Animais , Células Cultivadas , Humanos , Camundongos Endogâmicos NOD , Camundongos SCID , Osteogênese , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/metabolismo , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo
4.
PLoS One ; 6(7): e21888, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21779348

RESUMO

BACKGROUND: Acquisition of a blood supply is fundamental for extensive tumor growth. We recently described vascular heterogeneity in tumours derived from cell clones of a human mesenchymal stem cell (hMSC) strain (hMSC-TERT20) immortalized by retroviral vector mediated human telomerase (hTERT) gene expression. Histological analysis showed that cells of the most vascularized tumorigenic clone, -BD11 had a pericyte-like alpha smooth muscle actin (ASMA+) and CD146+ positive phenotype. Upon serum withdrawal in culture, -BD11 cells formed cord-like structures mimicking capillary morphogenesis. In contrast, cells of the poorly tumorigenic clone, -BC8 did not stain for ASMA, tumours were less vascularized and serum withdrawal in culture led to cell death. By exploring the heterogeneity in hMSC-TERT20 clones we aimed to understand molecular mechanisms by which mesenchymal stem cells may promote neovascularization. METHODOLOGY/PRINCIPAL FINDINGS: Quantitative qRT-PCR analysis revealed similar mRNA levels for genes encoding the angiogenic cytokines VEGF and Angiopoietin-1 in both clones. However, clone-BD11 produced a denser extracellular matrix that supported stable ex vivo capillary morphogenesis of human endothelial cells and promoted in vivo neovascularization. Proteomic characterization of the -BD11 decellularized matrix identified 50 extracellular angiogenic proteins, including galectin-1. siRNA knock down of galectin-1 expression abrogated the ex vivo interaction between decellularized -BD11 matrix and endothelial cells. More stable shRNA knock down of galectin-1 expression did not prevent -BD11 tumorigenesis, but greatly reduced endothelial migration into -BD11 cell xenografts. CONCLUSIONS: Decellularized hMSC matrix had significant angiogenic potential with at least 50 angiogenic cell surface and extracellular proteins, implicated in attracting endothelial cells, their adhesion and activation to form tubular structures. hMSC -BD11 surface galectin-1 expression was required to bring about matrix-endothelial interactions and for xenografted hMSC -BD11 cells to optimally recruit host vasculature.


Assuntos
Galectina 1/metabolismo , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Angiopoietina-1/genética , Angiopoietina-1/metabolismo , Animais , Western Blotting , Linhagem Celular , Feminino , Galectina 1/genética , Humanos , Imuno-Histoquímica , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Microscopia Confocal , Proteômica , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Ugeskr Laeger ; 172(38): 2616-9, 2010 Sep 20.
Artigo em Dinamarquês | MEDLINE | ID: mdl-20920407

RESUMO

Mesenchymal stem cells (MSC) are capable of multilineage differentiation into cells like osteoblasts, chondrocytes or adipocytes. MSCs can be isolated from bone marrow and expanded ex vivo for up to 25-40 population doublings while maintaining genetic stability and differentiation potential. MSCs have great potential in the field of tissue engineering and regenerative medicine where cartilage and bone conditions which are non-treatable or show very slow improvement can be effectively handled. Several clinical trials have been performed using MSC and show very promising results.


Assuntos
Doenças Ósseas/terapia , Doenças das Cartilagens/terapia , Transplante de Células-Tronco Mesenquimais , Animais , Doenças Ósseas/cirurgia , Transplante de Medula Óssea , Doenças das Cartilagens/cirurgia , Técnicas de Cultura de Células , Diferenciação Celular/fisiologia , Fraturas Ósseas/cirurgia , Fraturas Ósseas/terapia , Humanos , Osteoartrite/cirurgia , Osteoartrite/terapia , Resultado do Tratamento
6.
J Bone Miner Res ; 25(4): 796-808, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19821776

RESUMO

The ability of bone marrow stromal cells (BMSCs) to differentiate into osteoblasts is being exploited in cell-based therapy for repair of bone defects. However, the phenotype of ex vivo cultured BMSCs predicting their bone-forming capacity is not known. Thus we employed DNA microarrays comparing two human bone marrow stromal cell (hBMSC) populations: One is capable of in vivo heterotopic bone formation (hBMSC-TERT(+Bone)), and the other is not (hBMSC-TERT(-Bone)). Compared with hBMSC-TERT(-Bone), the hBMSC-TERT(+Bone) cells had an increased overrepresentation of extracellular matrix genes (17% versus 5%) and a larger percentage of genes with predicted SP3 transcription factor-binding sites in their promoter region (21% versus 8%). On the other hand, hBMSC-TERT(-Bone) cells expressed a larger number of immune-response-related genes (26% versus 8%). In order to test for the predictive value of these markers, we studied the correlation between their expression levels in six different hBMSC-derived clones and the ability to form bone in vivo. We found a significant correlation for decorin, lysyl oxidase-like 4, natriuretic peptide receptor C, and tetranectin. No significant positive correlation was found for canonical osteoblastic markers Runx2, alkaline phosphatase, collagen type I, osteopontin, and bone sialoprotein. Prospective isolation of four additional hBMSC clones based on their expression levels of the molecular markers correlated with their in vivo bone-formation ability. In conclusion, our data suggest an in vitro molecular signature predictive for hBMSCs' in vivo bone-formation ability. Identifying more of these predictive markers would be very useful in the quality control of osteoblastic cells before use in therapy.


Assuntos
Células da Medula Óssea/metabolismo , Osteoblastos/metabolismo , Osteogênese/genética , Aminoácido Oxirredutases/genética , Aminoácido Oxirredutases/metabolismo , Diferenciação Celular/genética , Células Cultivadas , Decorina , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Genes MHC da Classe II , Humanos , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Fenótipo , Proteína-Lisina 6-Oxidase , Proteoglicanas/genética , Proteoglicanas/metabolismo , Receptores do Fator Natriurético Atrial/genética , Receptores do Fator Natriurético Atrial/metabolismo , Fator de Transcrição Sp3/genética , Fator de Transcrição Sp3/metabolismo , Células Estromais/metabolismo
7.
Tissue Eng Part A ; 16(7): 2331-42, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20196644

RESUMO

Osteoblastic differentiation of human mesenchymal stem cells (hMSC) in monolayer culture is artefactual, lacking an organized bone-like matrix. We present a highly reproducible microwell protocol generating three-dimensional ex vivo multicellular aggregates of telomerized hMSC (hMSC-telomerase reverse transcriptase (TERT)) with improved mimicry of in vivo tissue-engineered bone. In osteogenic induction medium the hMSC were transitioned with time-dependent specification toward the osteoblastic lineage characterized by production of alkaline phosphatase, type I collagen, osteonectin, and osteocalcin. Introducing a 1-2 mm(3) crystalline hydroxyapatite/beta-tricalcium phosphate scaffold generated osteospheroids with upregulated gene expression of transcription factors RUNX2/CBFA1, Msx-2, and Dlx-5. An organized lamellar bone-like collagen matrix, evident by birefringence of polarized light, was deposited in the scaffold concavities. Here, mature osteoblasts stained positively for differentiated osteoblast markers TAZ, biglycan, osteocalcin, and phospho-AKT. Quantification of collagen birefringence and relatively high expression of genes for matrix proteins, including type I collagen, biglycan, decorin, lumican, elastin, microfibrillar-associated proteins (MFAP2 and MFAP5), periostin, and tetranectin, in vitro correlated predictively with in vivo bone formation. The three-dimensional hMSC-TERT/hydroxyapatite-tricalcium phosphate osteospheroid cultures in osteogenic induction medium recapitulated many characteristics of in vivo bone formation, providing a highly reproducible and resourceful platform for improved in vitro modeling of osteogenesis and refinement of bone tissue engineering.


Assuntos
Regeneração Óssea/fisiologia , Técnicas de Cultura de Células/métodos , Células-Tronco Mesenquimais/citologia , Osteogênese/fisiologia , Esferoides Celulares/citologia , Telômero/metabolismo , Alicerces Teciduais/química , Adulto , Animais , Biomarcadores/metabolismo , Birrefringência , Regeneração Óssea/efeitos dos fármacos , Fosfatos de Cálcio/farmacologia , Agregação Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Durapatita/farmacologia , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/genética , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos SCID , Osteoblastos/citologia , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteogênese/efeitos dos fármacos , Esferoides Celulares/efeitos dos fármacos , Esferoides Celulares/metabolismo , Células Estromais/citologia , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA