Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
1.
Gene Ther ; 23(6): 500-9, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26934099

RESUMO

Our previous work showed that a Sca-1(+) cell-based FGF2 therapy was capable of promoting robust increases in trabecular bone formation and connectivity on the endosteum of long bones. Past work reported that administration of FGF2 protein promoted bone formation in red marrow but not in yellow marrow. The issue as to whether the Sca-1(+) cell-based FGF2 therapy is effective in yellow marrow is highly relevant to its clinical potential for osteoporosis, as most red marrows in a person of an advanced age are converted to yellow marrows. Accordingly, this study sought to compare the osteogenic effects of this stem cell-based FGF2 therapy on red marrow-filled lumbar vertebrae with those on yellow marrow-filled caudal vertebrae of young adult W(41)/W(41) mice. The Sca-1(+) cell-based FGF2 therapy drastically increased trabecular bone formation in lumbar vertebrae, but the therapy not only did not promote bone formation but instead caused substantial loss of trabecular bone in caudal vertebrae. The lack of an osteogenic response was not due to insufficient engraftment of FGF2-expressing Sca-1(+) cells or inadequate FGF2 expression in caudal vertebrae. Previous studies have demonstrated that recipient mice of this stem cell-based FGF2 therapy developed secondary hyperparathyroidism and increased bone resorption. Thus, the loss of bone mass in caudal vertebrae might in part be due to an increase in resorption without a corresponding increase in bone formation. In conclusion, the Sca-1(+) cell-based FGF2 therapy is osteogenic in red marrow but not in yellow marrow.


Assuntos
Antígenos Ly/genética , Antígenos Ly/metabolismo , Fator 2 de Crescimento de Fibroblastos/genética , Terapia Genética/métodos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Animais , Células da Medula Óssea/metabolismo , Transplante de Medula Óssea/métodos , Osso Esponjoso/citologia , Osso Esponjoso/transplante , Caspase 3/genética , Feminino , Fator 2 de Crescimento de Fibroblastos/administração & dosagem , Fator 2 de Crescimento de Fibroblastos/biossíntese , Fator 2 de Crescimento de Fibroblastos/sangue , Humanos , Vértebras Lombares , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Osteogênese/genética , Osteomalacia/etiologia , Osteomalacia/genética , Transplante de Células-Tronco/métodos
2.
Gene Ther ; 22(9): 721-8, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25965395

RESUMO

We have previously obtained compelling proof-of-principle evidence for COX2 gene therapy for fracture repair using integrating retroviral vectors. For this therapy to be suitable for patient uses, a suitable vector with high safety profile must be used. Accordingly, this study sought to evaluate the feasibility of AAV as the vector for this COX2 gene therapy, because AAV raises less safety issues than the retroviral vectors used previously. However, an appropriate AAV serotype is required to provide early increase in and adequate level of COX2 expression that is needed for fracture repair. Herein, we reported that AAV-DJ, an artificial AAV pseudoserotype, is highly effective in delivering COX2 gene to fracture sites in a mouse femoral fracture model. Compared with AAV-2, the use of AAV-DJ led to ~5-fold increase in infectivity in mesenchymal stem cells (MSCs) and provided an earlier and significantly higher level of transgene expression at the fracture site. Injection of this vector at a dose of 7.5 × 10(11) genomic copies led to high COX2 level at the fracture site on day 3 after injections and significantly promoted fracture union at 21 days, as analyzed by radiography and µ-CT. The therapeutic effect appears to involve enhanced osteoblastic differentiation of MSCs and remodeling of callus tissues to laminar bone. This interpretation is supported by the enhanced expression of several key genes participating in the fracture repair process. In conclusion, AAV-DJ is a promising serotype for the AAV-based COX2 gene therapy of fracture repair in humans.


Assuntos
Ciclo-Oxigenase 2/metabolismo , Dependovirus/metabolismo , Consolidação da Fratura , Tíbia/lesões , Transgenes , Animais , Modelos Animais de Doenças , Terapia Genética , Vetores Genéticos/administração & dosagem , Masculino , Camundongos Endogâmicos C57BL
3.
J Bone Miner Metab ; 32(2): 124-35, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23700285

RESUMO

Urokinase plasminogen activator (uPA) regulates a proteolytic cascade of extracellular matrix degradation that functions in tissue development and tissue repair. The development and remodeling of the skeletal extracellular matrix during wound healing suggests that uPA might regulate bone development and repair. To determine whether uPA functions regulate bone development and repair, we examined the basal skeletal phenotype and endochondral bone fracture repair in uPA-deficient mice. The skeletal phenotype of uPA knockout mice was compared with that of control mice under basal conditions by dual-energy X-ray absorptiometry and micro-CT analysis, and during femur fracture repair by micro-CT and histological examination of the fracture callus. No effects of uPA gene deficiency were observed in the basal skeletal phenotype of the whole body or the femur. However, uPA gene deficiency resulted in increased fracture callus cartilage abundance during femur fracture repair at 14 days healing. The increase in cartilage corresponded to reduced tartrate-resistant acid phosphatase (TRAP) staining for osteoclasts in the uPA knockout fracture callus at this time, consistent with impaired osteoclast-mediated remodeling of the fracture cartilage. CD31 staining was reduced in the knockout fracture tissues at this time, suggesting that angiogenesis was also reduced. Osteoclasts also colocalized with CD31 expression in the endothelial cells of the fracture tissues during callus remodeling. These results indicate that uPA promotes remodeling of the fracture cartilage by osteoclasts that are associated with angiogenesis and suggest that uPA promotes angiogenesis and remodeling of the fracture cartilage at this time of bone fracture repair.


Assuntos
Cartilagem/metabolismo , Consolidação da Fratura/genética , Ativador de Plasminogênio Tipo Uroquinase/genética , Animais , Cartilagem/patologia , Regulação da Expressão Gênica , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase
4.
Am J Physiol Endocrinol Metab ; 305(2): E271-81, 2013 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-23715728

RESUMO

This study sought to determine whether deficient Igf1 expression in osteocytes would affect loading-induced osteogenic response. Tibias of osteocyte Igf1 conditional knockout (KO) mice (generated by cross-breeding Igf1 floxed mice with Dmp1-Cre transgenic mice) and wild-type (WT) littermates were subjected to four-point bending for 2 wk. Microcomputed tomography confirmed that the size of tibias of conditional mutants was smaller. Loading with an equivalent loading strain increased periosteal woven bone and endosteal lamellar bone formation in WT mice but not in conditional KO mice. Consistent with the lack of an osteogenic response, the loading failed to upregulate expression of early mechanoresponsive genes (Igf1, Cox-2, c-fos) or osteogenic genes (Cbfa-1, and osteocalcin) in conditional KO bones. The lack of osteogenic response was not due to reduced osteocyte density or insufficient loading strain. Deficient osteocyte Igf1 expression reduced the loading-induced upregulation of expression of canonical Wnt signaling genes (Wnt10b, Lrp5, Dkk1, sFrp2). The loading also reduced (by 40%) Sost expression in WT mice, but the loading not only did not reduce but upregulated (~1.5-fold) Sost expression in conditional KO mice. Conditional disruption of Igf1 in osteocytes also abolished the loading-induced increase in the bone ß-catenin protein level. These findings suggest an impaired response in the loading-induced upregulation of the Wnt signaling in conditional KO mice. In summary, conditional disruption of Igf1 in osteocytes abolished the loading-induced activation of the Wnt signaling and the corresponding osteogenic response. In conclusion, osteocyte-derived IGF-I plays a key determining role in bone mechanosensitivity.


Assuntos
Osso e Ossos/fisiologia , Fator de Crescimento Insulin-Like I/fisiologia , Mecanotransdução Celular/fisiologia , Osteócitos/fisiologia , Animais , Fenômenos Biomecânicos , Western Blotting , Desenvolvimento Ósseo/genética , Desenvolvimento Ósseo/fisiologia , Osso e Ossos/anatomia & histologia , Osso e Ossos/diagnóstico por imagem , DNA/genética , MAP Quinases Reguladas por Sinal Extracelular/fisiologia , Genótipo , Fator de Crescimento Insulin-Like I/genética , Mecanotransdução Celular/genética , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Fosfatidilinositóis/genética , Reação em Cadeia da Polimerase em Tempo Real , Tíbia/fisiologia , Tomografia Computadorizada por Raios X , Via de Sinalização Wnt/fisiologia , beta Catenina/metabolismo
5.
Mol Ther ; 20(2): 408-16, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22108860

RESUMO

The reprogramming of cord blood (CB) cells into induced pluripotent stem cells (iPSCs) has potential applications in regenerative medicine by converting CB banks into iPSC banks for allogeneic cell replacement therapy. Therefore, further investigation into novel approaches for efficient reprogramming is necessary. Here, we show that the lentiviral expression of OCT4 together with SOX2 (OS) driven by a strong spleen focus-forming virus (SFFV) promoter in a single vector can convert 2% of CB CD34(+) cells into iPSCs without additional reprogramming factors. Reprogramming efficiency was found to be critically dependent upon expression levels of OS. To generate transgene-free iPSCs, we developed an improved episomal vector with a woodchuck post-transcriptional regulatory element (Wpre) that increases transgene expression by 50%. With this vector, we successfully generated transgene-free iPSCs using OS alone. In conclusion, high-level expression of OS alone is sufficient for efficient reprogramming of CB CD34(+) cells into iPSCs. This report is the first to describe the generation of transgene-free iPSCs with the use of OCT4 and SOX2 alone. These findings have important implications for the clinical applications of iPSCs.


Assuntos
Diferenciação Celular/genética , Células-Tronco Hematopoéticas/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Fator 3 de Transcrição de Octâmero/genética , Fatores de Transcrição SOXB1/genética , Antígenos CD34/metabolismo , Sangue Fetal/citologia , Expressão Gênica , Vetores Genéticos/genética , Células-Tronco Hematopoéticas/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/metabolismo , Lentivirus/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo
6.
Am J Physiol Cell Physiol ; 302(11): C1676-86, 2012 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-22460711

RESUMO

This study utilized the glutathione transferase (GST) pull-down assay to identify novel substrates of an osteoclastic protein-tyrosine phosphatase, PTP-oc. Consistent with the previous findings that the phosphorylated tyr-527 (pY527) of Src is a substrate of PTP-oc, the major protein pulled down with the phosphatase-deficient (PD)-PTP-oc-GST trapping mutant in RAW264.7 cells was Src. The GST-PD-PTP-oc also pulled down pY-Syk and pY-ß(3)-integrin, but not after PP2 pretreatment. However, PTP-oc transgenic osteoclasts or PTP-oc-overexpressing RAW264.7 cells had elevated, and not reduced, levels of pY525/526-Syk and pY759-ß(3) integrin, and the PTP-oc siRNA treatment drastically reduced levels of pY525/526 Syk and pY759-ß(3)-integrin in RAW264.7 cells. These findings are incompatible with the premise that they are substrates of PTP-oc. The PTP-oc-dependent increases in pY525/526-Syk and pY759-ß(3)-integrin levels were completely blocked by PP2, indicating that these effects are secondary to PTP-oc-mediated activation of the Src protein-tyrosine kinase (PTK). Overexpression of PTP-oc increased, and siRNA-mediated suppression of PTP-oc reduced, pY160-Vav1, pY173-Vav3, and pY783-PLCγ levels, and Rac1 activation, which are downstream mediators of the ITAM/Syk signaling. Overexpression of PTP-oc also increased, and PTP-oc siRNA treatment decreased, the pY-Shp1 levels, which were blocked by PP2. Since Shp1 is a negative regulator of osteoclast activity and is a key mediator of the ITIM signaling, these findings suggest that PTP-oc is an upstream suppressor of the ITIM/Shp1 signaling through PTP-oc-induced Src-dependent Shp1 phosphorylation. In summary, PTP-oc plays a central regulatory role in the concerted regulation of the ß(3)-integrin, the ITAM/Syk, and the ITIM/Shp1 signaling indirectly through activation of Src PTK.


Assuntos
Integrina beta3/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Osteoclastos/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Quinases da Família src/metabolismo , Animais , Reabsorção Óssea , Linhagem Celular , Integrina beta3/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Neuropeptídeos/biossíntese , Neuropeptídeos/metabolismo , Osteoclastos/fisiologia , Fosfolipase C gama/biossíntese , Fosforilação , Proteínas Tirosina Fosfatases/genética , Proteínas Tirosina Quinases/genética , Proteínas Proto-Oncogênicas c-vav/biossíntese , Interferência de RNA , RNA Interferente Pequeno , Proteínas Recombinantes de Fusão/metabolismo , Transdução de Sinais , Quinase Syk , Proteínas rac de Ligação ao GTP/biossíntese , Proteínas rac de Ligação ao GTP/metabolismo , Proteínas rac1 de Ligação ao GTP
7.
J Biol Chem ; 285(48): 37607-18, 2010 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-20851886

RESUMO

This study investigated the role of leptin receptor (Lepr) signaling in determining the bone mechanosensitivity and also evaluated whether differences in the Lepr signaling may contribute to the differential osteogenic response of the C57BL/6J (B6) and C3H/HeJ (C3H) pair of mouse strains to mechanical stimuli. This study shows that a loading strain of ∼2,500 µÎµ, which was insufficient to produce a bone formation response in B6 mice, significantly increased bone formation parameters in leptin-deficient ob(-)/ob(-) mice and that a loading strain of ∼3,000 µÎµ also yielded greater osteogenic responses in Lepr-deficient db(-)/db(-) mice than in wild-type littermates. In vitro, a 30-min steady shear stress increased [(3)H]thymidine incorporation and Erk1/2 phosphorylation in ob(-)/ob(-) osteoblasts and db(-)/db(-) osteoblasts much greater than those in corresponding wild-type osteoblasts. The siRNA-mediated suppression of Lepr expression in B6 osteoblasts enhanced (but in osteoblasts of C3H (the mouse strain with poor bone mechanosensitivity) restored) their anabolic responses to shear stress. The Lepr signaling (leptin-induced Jak2/Stat3 phosphorylation) in C3H osteoblasts was higher than that in B6 osteoblasts. One of the three single nucleotide polymorphisms in the C3H Lepr coding region yielded an I359V substitution near the leptin binding region, suggesting that genetic variation of Lepr may contribute to a dysfunctional Lepr signaling in C3H osteoblasts. In conclusion, Lepr signaling is a negative modulator of bone mechanosensitivity. Genetic variations in Lepr, which result in a dysfunctional Lepr signaling in C3H mice, may contribute to the poor osteogenic response to loading in C3H mice.


Assuntos
Osso e Ossos/metabolismo , Regulação para Baixo , Variação Genética , Mecanotransdução Celular , Camundongos/metabolismo , Osteoblastos/metabolismo , Receptores para Leptina/genética , Receptores para Leptina/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Osso e Ossos/química , Células Cultivadas , Masculino , Camundongos/genética , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Osteogênese , Polimorfismo de Nucleotídeo Único , Resistência ao Cisalhamento , Estresse Mecânico
8.
Am J Physiol Endocrinol Metab ; 301(6): E1191-7, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21878662

RESUMO

To establish a causal role for locally produced IGF-I in the mechanical strain response in the bone, we have generated mice with conditional disruption of the insulin-like growth factor (IGF) I gene in type 1α(2) collagen-expressing cells using the Cre-loxP approach. At 10 wk of age, loads adjusted to account for bone size difference were applied via four-point bending or axial loading (AL) in mice. Two wk of bending and AL produced significant increases in bone mineral density and bone size at the middiaphysis of wild-type (WT), but not knockout (KO), mice. In addition, AL produced an 8-25% increase in trabecular parameters (bone volume-tissue volume ratio, trabecular thickness, and trabecular bone mineral density) at the secondary spongiosa of WT, but not KO, mice. Histomorphometric analysis at the trabecular site revealed that AL increased osteoid width by 60% and decreased tartrate-resistance acidic phosphatase-labeled surface by 50% in the WT, but not KO, mice. Consistent with the in vivo data, blockade of IGF-I action with inhibitory IGF-binding protein (IGFBP4) in vitro completely abolished the fluid flow stress-induced MC3T3-E1 cell proliferation. One-way ANOVA revealed that expression levels of EFNB1, EFNB2, EFNA2, EphB2, and NR4a3 were different in the loaded bones of WT vs. KO mice and may, in part, be responsible for the increase in bone response to loading in the WT mice. In conclusion, IGF-I expressed in type 1 collagen-producing bone cells is critical for converting mechanical signal to anabolic signal in bone, and other growth factors cannot compensate for the loss of local IGF-I.


Assuntos
Osso e Ossos/metabolismo , Colágeno Tipo I/metabolismo , Deleção de Genes , Fator de Crescimento Insulin-Like I/genética , Fator de Crescimento Insulin-Like I/fisiologia , Suporte de Carga/fisiologia , Animais , Densidade Óssea , Osso e Ossos/citologia , Osso e Ossos/ultraestrutura , Colágeno Tipo I/genética , Cadeia alfa 1 do Colágeno Tipo I , Feminino , Perfilação da Expressão Gênica , Técnicas de Transferência de Genes , Metabolismo/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Especificidade de Órgãos/genética , Osteoblastos/citologia , Osteoblastos/metabolismo , Estresse Mecânico
9.
J Gene Med ; 13(2): 77-88, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21322098

RESUMO

BACKGROUND: The present study assesses the effect of the stem cell antigen-1 positive (Sca-1(+) ) cell-based human growth hormone (hGH) ex vivo gene transfer strategy on endosteal bone mass in the mouse. METHODS: Sublethally irradiated recipient mice were transplanted with Sca-1(+) cells transduced with lentiviral vectors expressing hGH or ß-galactosidase control genes. Bone parameters were assessed by micro-computed tomography and histomorphometry. RESULTS: This hGH strategy drastically increased hGH mRNA levels in bone marrow cells and serum insulin-like growth factor-I (IGF-I) (by nearly 50%, p < 0.002) in hGH recipient mice. Femoral trabecular bone volume of the hGH mice was significantly reduced by 35% (p < 0.002). The hGH mice also had decreased trabecular number (by 26%; p < 0.0001), increased trabecular separation (by 38%; p < 0.0002) and reduced trabecular connectivity density (by 64%; p < 0.001), as well as significantly more osteoclasts (2.5-fold; p < 0.05) and greater osteoclastic surface per bone surface (2.6-fold; p < 0.01). CONCLUSIONS: Targeted expression of hGH in cells of marrow cavity through the Sca-1(+) cell-based gene transfer strategy increased circulating IGF-I and decreased endosteal bone mass through an increase in resorption in recipient mice. These results indicate that high local levels of hGH or IGF-I in the bone marrow microenvironment enhanced resorption, which is consistent with previous findings in transgenic mice with targeted bone IGF-I expression showing that high local IGF-I expression increased bone remodeling, favoring a net bone loss. Thus, GH and/or IGF-I would not be an appropriate transgene for use in this Sca-1(+) cell-based gene transfer strategy to promote endosteal bone formation. Published 2011 John Wiley & Sons, Ltd.


Assuntos
Antígenos Ly/metabolismo , Reabsorção Óssea , Técnicas de Transferência de Genes , Hormônio do Crescimento Humano , Proteínas de Membrana/metabolismo , Animais , Antígenos Ly/genética , Células da Medula Óssea/metabolismo , Transplante de Medula Óssea , Reabsorção Óssea/diagnóstico por imagem , Reabsorção Óssea/genética , Reabsorção Óssea/metabolismo , Reabsorção Óssea/patologia , Osso e Ossos/anatomia & histologia , Osso e Ossos/citologia , Osso e Ossos/patologia , Fêmur/anatomia & histologia , Fêmur/citologia , Dosagem de Genes , Regulação da Expressão Gênica , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Células HEK293 , Hormônio do Crescimento Humano/genética , Hormônio do Crescimento Humano/metabolismo , Humanos , Lentivirus/genética , Lentivirus/metabolismo , Masculino , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , RNA Mensageiro/metabolismo , Radiografia , Transdução Genética , Irradiação Corporal Total
10.
Cell Mol Life Sci ; 66(11-12): 1946-61, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19189046

RESUMO

Osteoclasts, the primary cell type mediating bone resorption, are multinucleated, giant cells derived from hematopoietic cells of monocyte-macrophage lineage. Osteoclast activity is, in a large part, regulated by protein-tyrosine phosphorylation. While information about functional roles of several protein-tyrosine kinases (PTK), including c-Src, in osteoclastic resorption has been accumulated, little is known about the roles of protein-tyrosine phosphatases (PTPs) in regulation of osteoclast activity. Recent evidence implicates important regulatory roles for four PTPs (SHP-1, cyt-PTP-epsilon, PTP-PEST, and PTPoc) in osteoclasts. Cyt-PTP-epsilon, PTP-PEST, and PTP-oc are positive regulators of osteoclast activity, while SHP-1 is a negative regulator. Of these PTPs in osteoclasts, only PTP-oc is a positive regulator of c-Src PTK through dephosphorylation of the inhibitory phosphotyrosine-527 residue. Although some information about mechanisms of action of these PTPs to regulate osteoclast activity is reviewed in this article, much additional work is required to provide more comprehensive details about their functions in osteoclasts.


Assuntos
Reabsorção Óssea/metabolismo , Osteoclastos/fisiologia , Proteínas Tirosina Fosfatases/fisiologia , Animais , Proteína Tirosina Quinase CSK , Humanos , Fosforilação , Isoformas de Proteínas/fisiologia , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , Proteínas Tirosina Quinases/fisiologia , Transdução de Sinais/fisiologia , Tirosina/metabolismo , Quinases da Família src
11.
J Gene Med ; 11(10): 877-88, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19629966

RESUMO

BACKGROUND: This study assessed whether a Sca-1+ cell-based ex vivo gene transfer strategy, which has been shown to promote robust endosteal bone formation with a modified fibroblast growth factor-2 (FGF2) gene, can be extended to use with bone morphogenetic protein (BMP)2/4 hybrid gene. METHODS: Sublethally irradiated recipient mice were transplanted with lentiviral (LV)-BMP2/4-transduced Sca-1+ cells. Bone parameters were monitored by pQCT and microCT. Gene expression was assessed by the real-time reverse transcriptase-polymerase chain reaction. RESULTS: Recipient mice of LV-BMP2/4-transduced Sca-1+ cells yielded high engraftment and increased BMP4 mRNA levels in marrow cells; but exhibited only insignificant increases in serum and bone alkaline phosphatase activity compared to control mice. pQCT and microCT analyses of femurs showed that, with the exception of small changes in trabecular bone mineral density and cortical bone mineral content in LV-BMP2/4 mice, there were no differences in measured bone parameters between mice of the LV-BMP2/4 group and controls. The lack of large endosteal bone formation effects with the BMP4 strategy could not be attributed to ineffective engraftment or expansion of BMP4-expressing Sca-1+ cells, an inability of the transduced cells to secrete active BMP4 proteins, or to use of the LV-based vector. CONCLUSIONS: Sca-1+ cell-based BMP4 ex vivo strategy did not promote robust endosteal bone formation, raising the possibility of intrinsic differences between FGF2- and BMP4-based strategies in their ability to promote endosteal bone formation. It emphasizes the importance of choosing an appropriate bone growth factor gene for delivery by this Sca-1+ cell-based ex vivo systemic gene transfer strategy to promote bone formation.


Assuntos
Antígenos Ly/biossíntese , Proteína Morfogenética Óssea 4/biossíntese , Terapia Genética , Proteínas de Membrana/biossíntese , Osteogênese/genética , Osteoporose/terapia , Animais , Proteína Morfogenética Óssea 2/biossíntese , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 4/genética , Fator 2 de Crescimento de Fibroblastos/biossíntese , Fator 2 de Crescimento de Fibroblastos/genética , Expressão Gênica , Vetores Genéticos , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/patologia , Humanos , Camundongos , Osteoporose/genética , Osteoporose/metabolismo , Proteínas Proto-Oncogênicas c-kit/deficiência , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/genética , Transdução Genética
12.
Calcif Tissue Int ; 85(4): 356-67, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19763374

RESUMO

This study evaluated whether the murine leukemia virus (MLV)-based cyclooxygenase-2 (Cox-2) ex vivo gene-transfer strategy promotes healing of calvarial defects and/or synergistically enhances bone morphogenetic protein (BMP) 4-mediated bone regeneration. Gelatin scaffolds impregnated with mouse marrow stromal cells (MSCs) transduced with MLV-expressing BMP4, Cox-2, or a control gene were implanted into mouse calvarial defects. Bone regeneration was assessed by X-ray, dual-energy X-ray absorptiometry, and histology. In vitro, Cox-2 or prostanglandin E(2) enhanced synergistically the osteoblastic differentiation action of BMP4 in mouse MSCs. In vivo, implantation of BMP4-expressing MSCs yielded massive bone regeneration in calvarial defects after 2 weeks, but the Cox-2 strategy surprisingly did not promote bone regeneration even after 4 weeks. Staining for alkaline phosphatase (ALP)-expressing osteoblasts was strong throughout the defect of animals receiving BMP2/4-expressing cells, but defects receiving Cox-2-expressing cells displayed weak ALP staining along the edge of original intact bone, indicating that the Cox-2 strategy lacked bone-regeneration effects. The Cox-2 strategy not only lacked bone-regeneration effects but also suppressed the BMP4-induced bone regeneration. In vitro coculture of Cox-2-expressing MSCs with BMP4-expressing MSCs in gelatin scaffolds reduced BMP4 mRNA transcript levels, suggesting that Cox-2 may promote BMP4 gene silencing in BMP4-expressing cells, which may play a role in the suppressive action of Cox-2 on BMP4-mediated bone formation. In summary, the Cox-2 ex vivo gene-transfer strategy not only lacks bone-regeneration effects but also suppresses the bone-regeneration action of BMP4 in healing of calvarial defects.


Assuntos
Células da Medula Óssea/metabolismo , Proteína Morfogenética Óssea 4/metabolismo , Regeneração Óssea/fisiologia , Ciclo-Oxigenase 2/metabolismo , Crânio/metabolismo , Células Estromais/metabolismo , Animais , Células da Medula Óssea/citologia , Proteína Morfogenética Óssea 2/genética , Proteína Morfogenética Óssea 2/metabolismo , Proteína Morfogenética Óssea 4/genética , Células Cultivadas , Ciclo-Oxigenase 2/genética , Dinoprostona/metabolismo , Técnicas de Transferência de Genes , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais , RNA Mensageiro/metabolismo , Crânio/lesões , Células Estromais/citologia
13.
FEBS Lett ; 582(10): 1451-8, 2008 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-18381073

RESUMO

This study presents gene expression, protein expression, and in situ immunohistochemical evidence that osteoclasts express high levels of osteoactivin (OA), which had previously been reported to be an osteoblast-specific protein in bone. OA expression in osteoclasts was up-regulated upon receptor activator of NFkappaB ligand-induced differentiation. Suppression of functional activity of OA with neutralizing antibody reduced cell size, number of nuclei, fusion, and bone resorption activity of osteoclasts. OA was co-immunoprecipitated with integrin beta3 and beta1, indicating that OA co-localizes with integrin beta3 and/or beta1 in a hetero-polymeric complex in osteoclasts. These findings indicate that OA is a novel osteoclastic protein and plays a role in osteoclast differentiation and/or activity.


Assuntos
Diferenciação Celular , Proteínas do Olho/metabolismo , Glicoproteínas de Membrana/metabolismo , Osteoclastos/citologia , Fosfatase Ácida/genética , Fosfatase Ácida/metabolismo , Animais , Anticorpos/farmacologia , Diferenciação Celular/genética , Proteínas do Olho/genética , Expressão Gênica , Imunoprecipitação , Integrina beta1/metabolismo , Integrina beta3/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Glicoproteínas de Membrana/genética , Camundongos , NF-kappa B/metabolismo , Osteoclastos/metabolismo , Ligante RANK/genética , Ligante RANK/metabolismo , Fosfatase Ácida Resistente a Tartarato
14.
Bone ; 43(5): 880-8, 2008 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18708175

RESUMO

This study sought to determine the role of the pro-apoptotic gene, Bax, in fracture healing by comparing femoral fracture healing in Bax knockout (KO) and wild-type C57BL/6J (background strain) mice. Bax KO fractures were larger, had more bone mineral content, had approximately 2-fold larger cartilage area per callus area in the first and second weeks of fracture healing, and showed an increased osteoclast surface area in the third and fourth weeks of fracture healing compared to C57BL/6J fractures. The increased cartilage area in the Bax KO fracture callus was due to increases in number of both pre-hypertropic and hypertropic chondrocytes. TUNEL analysis showed no significant differences in the number of either chondrocyte or non-chondrocyte apoptotic cells between Bax KO and C57BL/6J fractures at 7 or 14 days post-fracture, indicating that the increased number of chondrocytes in Bax KO fractures was not due to reduced apoptosis. Analysis of expression of apoptotic genes revealed that although the expression levels of Bcl-2 and Bcl-xL were not different between the Bax KO and C57BL/6J mice at 7 or 14 days post-fracture, the expression of BH3-domain only Bak and "Bik-like" pro-apoptotic gene increased approximately 1.5-fold and approximately 2-fold, respectively, in Bax KO fractures at 7 and 14 days post-fracture, compared to C57BL/6J fractures, suggesting that up-regulation of the Bak and Bik-like pro-apoptotic genes in Bax KO mice might compensate for the lack of Bax functions in the context of apoptosis. Analysis by in vivo incorporation of bromodeoxyuridine into chondrocytes within the fracture tissues indicated a highly significant increase in chondrocyte proliferation in Bax KO fractures compared to C57BL/6J fractures at day 7. The increased expression of collagen 2alpha1 and 9alpha1 gene in Bax KO fractures during early healing was consistent with an increased chondrocyte proliferation. In conclusion, this study demonstrates for the first time that Bax has an important role in the early stage of fracture healing, and that the increased callus size and cartilage area in Bax KO fractures was due to increased chondrocyte proliferation and not to reduced apoptosis or increased chondrocyte hypertrophy. The unexpected effect of Bax deficiency on chondrocyte proliferation implicates a novel regulatory function for Bax on chondrocyte proliferation during fracture repair.


Assuntos
Apoptose/fisiologia , Cartilagem/fisiologia , Condrócitos/fisiologia , Consolidação da Fratura/fisiologia , Proteína X Associada a bcl-2/metabolismo , Animais , Calo Ósseo/citologia , Calo Ósseo/metabolismo , Cartilagem/citologia , Proliferação de Células , Feminino , Fêmur/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína X Associada a bcl-2/genética
15.
Calcif Tissue Int ; 83(4): 276-84, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18820962

RESUMO

To evaluate the role of plasminogen activator inhibitor (PAI)-1, a key negative regulator of the plasmin system of extracellular matrix proteases in developmental bone growth and fracture repair, the bone phenotype of male adult PAI-1-deficient mice was determined and femoral fracture healing was compared with that of age- and sex-matched wild-type C57BL/6J control mice. Regarding bone phenotype, the length and size (but not cortical thickness) of the femur of male PAI-1-deficient mice were smaller than those of wild-type controls. Although the total bone mineral content of PAI-1-deficient mice was not significantly different from that of wild-type mice, the total bone area in PAI-1-deficient mice was smaller, leading to an increase in total bone mineral density. With respect to fracture healing, PAI-1-deficient mice developed fracture calluses that were larger and more mineralized than those of wild-type mice but only at 14 days postfracture. These changes were even greater given the smaller size of the normal femur in PAI-1-deficient mice. Surprisingly, the larger fracture callus remodeled rapidly to normal size and mineral content by 21 days postfracture. Examination of fracture histology revealed that these changes were associated with a dramatic increase followed by a rapid remodeling of the fracture callus cartilage. The remodeling of fracture callus cartilage in PAI-1-deficient mice also displayed an abnormal pattern. These findings demonstrate for the first time that PAI-1 (and potentially the plasminogen extracellular matrix protease system) is an important regulator of bone size during developmental growth and plays a regulatory role in the determination of fracture callus size, cartilage formation, and resorption during bone fracture repair.


Assuntos
Consolidação da Fratura/fisiologia , Inibidor 1 de Ativador de Plasminogênio/fisiologia , Animais , Densidade Óssea/fisiologia , Modelos Animais de Doenças , Fêmur/citologia , Fêmur/fisiologia , Consolidação da Fratura/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Inibidor 1 de Ativador de Plasminogênio/genética , Ativadores de Plasminogênio/metabolismo , Tomografia Computadorizada por Raios X , Ativador de Plasminogênio Tipo Uroquinase/metabolismo
16.
FEBS Lett ; 581(13): 2503-8, 2007 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-17498702

RESUMO

An osteoclastic protein-tyrosine phosphatase, PTP-oc, shares the same gene with a renal PTP, Glepp1. This study demonstrated that targeted deletion of PTP-oc promoter by homologous recombination in RAW264.7 cells completely abolished PTP-oc expression without affecting Glepp1 expression. This strategy to inhibit PTP-oc function has three advantages over commonly used gene knock down strategies (e.g., small interference RNA). This strategy: (1) yielded cells completely devoid of PTP-oc, (2) had no off-target gene silencing effects, and (3) did not affect Glepp1 expression. The inability of PTP-oc-deficient RAW264.7 cells to undergo RANKL-mediated osteoclastic differentiation confirmed a regulatory role for PTP-oc in RANKL-mediated osteoclast differentiation.


Assuntos
Proteínas de Membrana/genética , Osteoclastos/citologia , Proteínas Tirosina Fosfatases/genética , Animais , Diferenciação Celular , Linhagem Celular , Deleção de Genes , Camundongos , Plasmídeos , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores , Recombinação Genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Deleção de Sequência
17.
Bone ; 38(4): 521-9, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16321582

RESUMO

Microarray analysis of gene expression was performed in the healing femur fractures of 13-week-old male rats during the inflammatory stage of repair, at 3 days post-fracture, and the endochondral bone formation stage of repair, at 11 days post-fracture. Multiple replicate pairs of fracture tissues paired with unfractured tissues, and unfractured control bones that had the stabilizing K-wire were introduced. This approach normalized the marrow contributions to the RNA repertoire. We identified 6555 genes with significant changes in expression in fracture tissues at 3 days and 11 days healing. The repertoire of growth factor genes expressed was also surprisingly restricted at both post-fracture intervals. The large number of Expressed Sequence Tags (ESTs) expressed at both post-fracture times indicates that several molecular pathways yet to be identified regulate fracture repair. The number of genes expressed during immune responses and inflammatory processes was restricted with higher expression largely during the early post-fracture analysis. Several of the genes identified in this study have been associated with regulation of cell and extracellular matrix interactions during scarless healing of fetal skin wounds. These observations suggest that these genes might also regulate the scarless healing characteristic of bone regeneration by similar mechanisms.


Assuntos
Fraturas do Fêmur/genética , Consolidação da Fratura , Perfilação da Expressão Gênica , Inflamação/genética , Análise de Sequência com Séries de Oligonucleotídeos , Animais , Sequência de Bases , Primers do DNA , Etiquetas de Sequências Expressas , Fraturas do Fêmur/patologia , Inflamação/patologia , Masculino , Ratos , Ratos Sprague-Dawley
18.
Curr Stem Cell Res Ther ; 11(2): 114-21, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26027679

RESUMO

Mesenchymal stem cells (MSCs) have been successfully used to treat multiple diseases in animal studies and clinical trials. Currently, the commonly used MSCs are derived from bone marrow and adipose tissue. Alternative approaches include differentiation of induced pluripotent stem cells (iPSCs) into MSCs, or direct reprogramming of blood cells into MSCs. This review summarizes recent progresses concerning how to generate MSCs by blood cell reprogramming and how studies in cellular reprogramming may help identify new factors to expand or even rejuvenate adult MSCs.


Assuntos
Células Sanguíneas/transplante , Reprogramação Celular/genética , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/citologia , Tecido Adiposo/citologia , Células Sanguíneas/citologia , Diferenciação Celular/genética , Proliferação de Células/genética , Humanos , Células-Tronco Pluripotentes Induzidas
19.
J Bone Miner Res ; 18(10): 1897-900, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14584902

RESUMO

Although type 5 TRACP is recognized as a histochemical and biochemical marker of osteoclasts, there is evidence that bone forming cells, osteoblasts, and osteocytes also express a type 5 TRACP. Accordingly, an osteoblastic type 5 TRACP has been purified from human osteoblasts and from bovine cortical bone matrices. Comparison of biochemical properties of osteoblastic type 5 TRACP with those of osteoclastic type 5 TRACP suggests that osteoblastic type 5 TRACP is a different isoenzyme from osteoclastic type 5 TRACP. Two properties of osteoblastic type 5 TRACP may be relevant to its physiological functions: (1) it acts as a protein-tyrosine phosphatase (protein tyrosine phosphorylation) under physiologically relevant conditions, and (2) it is sensitive to inhibition by clinically relevant concentrations of fluoride. Because fluoride is a stimulator of osteoblastic proliferation and differentiation and a potent osteogenic agent and because protein tyrosine phosphorylation plays an important regulatory role in cell proliferation and differentiation, these unique properties and other evidence summarized in this review led to the proposal that the osteogenic action of fluoride is mediated, at least in part, by the fluoride-mediated inhibition of osteoblastic type 5 TRACP/protein tyrosine phosphorylation, which leads to a stimulation of osteoblast proliferation and differentiation, and subsequently, an increase in bone formation.


Assuntos
Fosfatase Ácida/fisiologia , Isoenzimas/fisiologia , Osteoblastos/enzimologia , Animais , Osso e Ossos/metabolismo , Bovinos , Diferenciação Celular , Divisão Celular , Humanos , Osteoblastos/metabolismo , Osteoclastos/enzimologia , Fosforilação , Isoformas de Proteínas , Proteínas Tirosina Fosfatases/metabolismo , Fosfatase Ácida Resistente a Tartarato
20.
Crit Rev Eukaryot Gene Expr ; 14(1-2): 89-136, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15104530

RESUMO

Skeletal gene therapy is an attractive new approach to the treatment of bone disorders. Impressive advances in our knowledge of the molecular genetic basis of skeletal disorders and fracture healing have led to the development of novel therapeutics based on ectopic expression of one or more genes in patient cells that can influence repair or regenerative processes in bone. Although still a relatively immature field, proof-of-principle for enhanced bone formation through skeletal gene therapy has already been established. The challenge now is to more precisely define optimal cellular targets and therapeutic genes, and to develop safe and efficient ways to deliver therapeutic genes to target cells. In this review, we will highlight some of the exciting advances that have been made in skeletal gene therapy in recent years, with a focus on treatment of localized skeletal lesions. Strengths and weaknesses of current approaches will be discussed, as will strategies for improved safety and therapeutic outcome in the future. Skeletal gene therapy can have an enormous impact on patient care. The next 5 years will present us with unparalleled opportunities to develop more effective therapeutic strategies and overcome obstacles presented by current gene transfer technologies.


Assuntos
Doenças Ósseas/terapia , Fraturas Ósseas/terapia , Terapia Genética/métodos , Terapia Genética/tendências , Consolidação da Fratura/genética , Vetores Genéticos/genética , Vetores Genéticos/uso terapêutico , Substâncias de Crescimento/uso terapêutico , Humanos , Osteogênese/genética , Vírus/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa