Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Immunol ; 203(1): 105-116, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31109956

RESUMO

We found that protease-activated receptor 1 (PAR1) was transiently induced in cultured osteoclast precursor cells. Therefore, we examined the bone phenotype and response to resorptive stimuli of PAR1-deficient (knockout [KO]) mice. Bones and bone marrow-derived cells from PAR1 KO and wild-type (WT) mice were assessed using microcomputed tomography, histomorphometry, in vitro cultures, and RT-PCR. Osteoclastic responses to TNF-α (TNF) challenge in calvaria were analyzed with and without a specific neutralizing Ab to the Notch2-negative regulatory region (N2-NRR Ab). In vivo under homeostatic conditions, there were minimal differences in bone mass or bone cells between PAR1 KO and WT mice. However, PAR1 KO myeloid cells demonstrated enhanced osteoclastogenesis in response to receptor activator of NF-κB ligand (RANKL) or the combination of RANKL and TNF. Strikingly, in vivo osteoclastogenic responses of PAR1 KO mice to TNF were markedly enhanced. We found that N2-NRR Ab reduced TNF-induced osteoclastogenesis in PAR1 KO mice to WT levels without affecting WT responses. Similarly, in vitro N2-NRR Ab reduced RANKL-induced osteoclastogenesis in PAR1 KO cells to WT levels without altering WT responses. We conclude that PAR1 functions to limit Notch2 signaling in responses to RANKL and TNF and moderates osteoclastogenic response to these cytokines. This effect appears, at least in part, to be cell autonomous because enhanced osteoclastogenesis was seen in highly purified PAR1 KO osteoclast precursor cells. It is likely that this pathway is involved in regulating the response of bone to diseases associated with inflammatory signals.


Assuntos
Doenças Ósseas/imunologia , Inflamação/imunologia , Osteoclastos/fisiologia , Receptor Notch2/metabolismo , Receptor PAR-1/metabolismo , Animais , Anticorpos Neutralizantes/metabolismo , Células Cultivadas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteogênese/genética , Ligante RANK/metabolismo , Receptor Notch2/imunologia , Receptor PAR-1/genética , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
2.
Bone ; 67: 1-9, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24994594

RESUMO

Cbl and Cbl-b are E3 ubiquitin ligases and adaptor proteins, which perform regulatory roles in bone remodeling. Cbl-/- mice have delayed bone development due to decreased osteoclast migration. Cbl-b-/- mice are osteopenic due to increased bone resorbing activity of osteoclasts. Unique to Cbl, but not present in Cbl-b, is tyrosine 737 in the YEAM motif, which upon phosphorylation provides a binding site for the regulatory p85 subunit of PI3K. Substitution of tyrosine 737 with phenylalanine (Y737F, CblYF/YF mice) prevents Y737 phosphorylation and abrogates the Cbl-PI3K interaction. We have previously reported that CblYF/YF mice had increased bone volume due to defective bone resorption and increased bone formation. Here we show that the lumbar vertebra from CblYF/YF mice did not have significant bone loss following ovariectomy. Our data also suggests that abrogation of Cbl-PI3K interaction in mice results in the loss of coupling between bone resorption and formation, since ovariectomized CblYF/YF mice did not show significant changes in serum levels of c-terminal telopeptide (CTX), whereas the serum levels of pro-collagen type-1 amino-terminal pro-peptide (P1NP) were decreased. In contrast, following ovariectomy, Cbl-/- and Cbl-b-/- mice showed significant bone loss in the tibiae and L2 vertebrae, concomitant with increased serum CTX and P1NP levels. These data indicate that while lack of Cbl or Cbl-b distinctly affects bone remodeling, only the loss of Cbl-PI3K interaction protects mice from significant bone loss following ovariectomy.


Assuntos
Reabsorção Óssea/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-cbl/metabolismo , Animais , Reabsorção Óssea/sangue , Reabsorção Óssea/genética , Colágeno Tipo I/sangue , Feminino , Camundongos , Camundongos Knockout , Ovariectomia , Peptídeos/sangue , Fosfatidilinositol 3-Quinases/genética , Ligação Proteica/genética , Ligação Proteica/fisiologia , Proteínas Proto-Oncogênicas c-cbl/genética
3.
J Cell Biochem ; 115(8): 1449-57, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24590570

RESUMO

Cathepsin K (CatK) is a lysosomal cysteine protease necessary for bone resorption by osteoclasts (OCs), which originate from myeloid hematopoietic precursors. CatK-deficient (CatK(-/-) ) mice show osteopetrosis due to defective resorption by OCs, which are increased in number in these mice. We investigated whether genetic ablation of CatK altered the number of hematopoietic stem cells (HSCs) and OC precursor cells (OCPs) using two mouse models: CatK(-/-) mice and a knock-in mouse model in which the CatK gene (ctsk) is replaced by cre recombinase. We found that CatK deletion in mice significantly increased the number of HSCs in the spleen and decreased their number in bone marrow. In contrast, the number of early OCPs was unchanged in the bone marrow. However, the number of committed CD11b(+) OCPs was increased in the bone marrow of CatK(-/-) compared to wild-type (WT) mice. In addition, the percentage but not the number of OCPs was decreased in the spleen of CatK(-/-) mice relative to WT. To understand whether increased commitment to OC lineage in CatK(-/-) mice is influenced by the bone marrow microenvironment, CatK(Cre/+) or CatK(Cre/Cre) red fluorescently labeled OCPs were injected into WT mice, which were also subjected to a mid-diaphyseal femoral fracture. The number of OCs derived from the intravenously injected CatK(Cre/Cre) OCPs was lower in the fracture callus compared to mice injected with CatK(+/Cre) OCPs. Hence, in addition to its other effects, the absence of CatK in OCP limits their ability to engraft in a repairing fracture callus compared to WT OCP.


Assuntos
Reabsorção Óssea/genética , Catepsina K/genética , Células-Tronco Hematopoéticas/metabolismo , Osteogênese , Animais , Reabsorção Óssea/patologia , Catepsina K/metabolismo , Consolidação da Fratura/genética , Células-Tronco Hematopoéticas/patologia , Camundongos , Camundongos Knockout , Osteoclastos/metabolismo , Osteoclastos/patologia , Osteopetrose/genética , Osteopetrose/patologia
4.
J Cell Biochem ; 115(7): 1277-89, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24470255

RESUMO

Cbl family proteins, Cbl and Cbl-b, are E3 ubiquitin ligases and adaptor proteins, which play important roles in bone-resorbing osteoclasts. Loss of Cbl in mice decreases osteoclast migration, resulting in delayed bone development where as absence of Cbl-b decreases bone volume due to hyper-resorptive osteoclasts. A major structural difference between Cbl and Cbl-b is tyrosine 737 (in YEAM motif) only on Cbl, which upon phosphorylation interacts with the p85 subunit of phosphatidylinositol-3 Kinase (PI3K). In contrast to Cbl(-/-) and Cbl-b(-/-) , mice lacking Cbl-PI3K interaction due to a Y737F (tyrosine to phenylalanine, YF) mutation showed enhanced osteoclast survival, but defective bone resorption. To investigate whether Cbl-PI3K interaction contributes to distinct roles of Cbl and Cbl-b in osteoclasts, mice bearing CblY737F mutation in the Cbl-b(-/-) background (YF/YF;Cbl-b(-/-) ) were generated. The differentiation and survival were augmented similarly in YF/YF and YF/YF;Cbl-b(-/-) osteoclasts, associated with enhanced PI3K signaling suggesting an exclusive role of Cbl-PI3K interaction, independent of Cbl-b. In addition to PI3K, the small GTPase Ras also regulates osteoclast survival. In the absence of Cbl-PI3K interaction, increased Ras GTPase activity and Ras-PI3K binding were observed and inhibition of Ras activation attenuated PI3K mediated osteoclast survival. In contrast to differentiation and survival, increased osteoclast activity observed in Cbl-b(-/-) mice persisted even after introduction of the resorption-defective YF mutation in YF/YF;Cbl-b(-/-) mice. Hence, Cbl and Cbl-b play mutually exclusive roles in osteoclasts. Whereas Cbl-PI3K interaction regulates differentiation and survival, bone resorption is predominantly regulated by Cbl-b in osteoclasts.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Classe Ia de Fosfatidilinositol 3-Quinase/metabolismo , Osteoclastos/citologia , Proteínas Proto-Oncogênicas c-cbl/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Animais , Densidade Óssea/genética , Remodelação Óssea/genética , Reabsorção Óssea/genética , Diferenciação Celular/genética , Movimento Celular/genética , Sobrevivência Celular/genética , Células Cultivadas , Cromonas/farmacologia , Classe Ia de Fosfatidilinositol 3-Quinase/biossíntese , Classe Ia de Fosfatidilinositol 3-Quinase/genética , Quinase 3 da Glicogênio Sintase/metabolismo , Glicogênio Sintase Quinase 3 beta , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Morfolinas/farmacologia , Mutação , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação/efeitos dos fármacos , Ligação Proteica , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Proto-Oncogênicas c-cbl/biossíntese , Proteínas Proto-Oncogênicas c-cbl/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/antagonistas & inibidores , Proteínas Proto-Oncogênicas p21(ras)/biossíntese , Ligante RANK/farmacologia , Transdução de Sinais/genética
5.
J Bone Miner Res ; 28(5): 1203-13, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23165930

RESUMO

Osteoclasts are specialized bone-resorbing cells that derive from monocyte precursors. We have identified three populations of cells with high osteoclastogenic potential in murine bone marrow, which expressed the phenotype B220(-) CD3(-) CD11b(-/low) CD115(+) and either CD117(hi), CD117(intermediate), or CD117(low). We have evaluated these populations for their ability to also generate macrophages and dendritic cells. At a single-cell level, the population expressing higher CD117 levels was able to generate bone-resorbing osteoclasts, phagocytic macrophages, and antigen-presenting dendritic cells in vitro with efficiencies of more than 90%, indicating that there exists a common developmental pathway for these cell types. Cells with osteoclastogenic potential also exist in blood and peripheral hematopoietic organs. Their functional meaning and/or their relationship with bone marrow progenitors is not well established. Hence, we characterized murine peripheral cell populations for their ability to form osteoclasts, macrophages, and dendritic cells in vitro. The spleen and peripheral blood monocyte progenitors share phenotypic markers with bone marrow progenitors but differ in their expression of CD11b, which was low in bone marrow but high in periphery. We propose that circulating monocyte progenitors are derived from a common bone marrow osteoclasts/macrophage/dendritic cell progenitor (OcMDC), which we have now characterized at a clonal level. However, the lineage relationship between the bone marrow and peripheral monocyte progenitors has yet to be defined.


Assuntos
Células da Medula Óssea/citologia , Células Dendríticas/citologia , Macrófagos/citologia , Osteoclastos/citologia , Animais , Masculino , Camundongos , Camundongos Endogâmicos C57BL
6.
J Bone Miner Res ; 28(1): 135-49, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22893614

RESUMO

Negative regulation of osteoclastogenesis is important for bone homeostasis and prevention of excessive bone resorption in inflammatory and other diseases. Mechanisms that directly suppress osteoclastogenesis are not well understood. In this study we investigated regulation of osteoclast differentiation by the ß2 integrin CD11b/CD18 that is expressed on myeloid lineage osteoclast precursors. CD11b-deficient mice exhibited decreased bone mass that was associated with increased osteoclast numbers and decreased bone formation. Accordingly, CD11b and ß2 integrin signaling suppressed osteoclast differentiation by preventing receptor activator of NF-κB ligand (RANKL)-induced induction of the master regulator of osteoclastogenesis nuclear factor of activated T cells, cytoplasmic 1 (NFATc1) and of downstream osteoclast-related NFATc1 target genes. CD11b suppressed induction of NFATc1 by the complementary mechanisms of downregulation of RANK expression and induction of recruitment of the transcriptional repressor B-cell lymphoma 6 (BCL6) to the NFATC1 gene. These findings identify CD11b as a negative regulator of the earliest stages of osteoclast differentiation, and provide an inducible mechanism by which environmental cues suppress osteoclastogenesis by activating a transcriptional repressor that makes genes refractory to osteoclastogenic signaling.


Assuntos
Antígeno CD11b/metabolismo , Antígenos CD18/metabolismo , Diferenciação Celular , Proteínas de Ligação a DNA/metabolismo , Osteoclastos/citologia , Transdução de Sinais , Células-Tronco/citologia , Animais , Contagem de Células , Diferenciação Celular/efeitos dos fármacos , Fêmur/diagnóstico por imagem , Fêmur/efeitos dos fármacos , Fêmur/metabolismo , Fibrinogênio/metabolismo , Fibrinogênio/farmacologia , Humanos , Integrina beta3/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Transcrição NFATC/genética , Fatores de Transcrição NFATC/metabolismo , Tamanho do Órgão/efeitos dos fármacos , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Osteogênese/efeitos dos fármacos , Fenótipo , Proteínas Proto-Oncogênicas c-bcl-6 , Ligante RANK/farmacologia , Transdução de Sinais/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Transcrição Gênica/efeitos dos fármacos , Microtomografia por Raio-X
7.
J Bone Miner Res ; 26(6): 1207-16, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21611963

RESUMO

Parathyroid hormone (PTH) increases both the number of osteoclast in bone and the number of early hematopoietic stem cells (HSCs) in bone marrow. We previously characterized the phenotype of multiple populations of bone marrow cells with in vitro osteoclastogenic potential in mice. Here we examined whether intermittent administration of PTH influences these osteoclast progenitor (OCP) populations. C57BL/6 mice were treated with daily injections of bPTH(1-34) (80 µg/kg/day) for 7 or 14 days. We found that PTH caused a significant increase in the percentage of TN/CD115(+) CD117(high) and TN/CD115(+) CD117(int) cells (p < .05) in bone marrow on day 7. In contrast, PTH decreased the absolute number of TN/CD115(+) CD117(low) cells by 39% on day 7 (p < .05). On day 14, there was no effect of PTH on osteoclast progenitor distribution in vivo. However, PTH treatment for 7 and 14 days did increase receptor activator of NF-κB ligand (RANKL)- and macrophage colony-stimulating factor (M-CSF)-stimulated in vitro osteoclastogenesis and bone resorption in TN/CD115(+) cells. In the periphery, 14 days of treatment increased the percentage and absolute numbers of HSCs (Lin(-) CD117(+) Sca-1(+) ) in the spleen (p < .05). These data correlated with an increase in the percent and absolute numbers of HSCs in bone marrow on day 14 (p < .05). Interestingly, the effects on hematopoietic progenitors do not depend on osteoclast resorption activity. These results suggest that in vivo PTH treatment increased in vitro osteoclastogenesis and resorption without altering the number of osteoclast precursors. This implies that in vivo PTH induces sustained changes, possibly through an epigenetic mechanism, in the in vitro responsiveness of the cells to M-CSF and RANKL.


Assuntos
Células da Medula Óssea/citologia , Diferenciação Celular/efeitos dos fármacos , Células-Tronco Hematopoéticas/citologia , Osteoclastos/citologia , Osteoclastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Hormônio Paratireóideo/farmacologia , Fosfatase Ácida/metabolismo , Animais , Bioensaio , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Reabsorção Óssea/metabolismo , Reabsorção Óssea/patologia , Bovinos , Tamanho Celular/efeitos dos fármacos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Células-Tronco Hematopoéticas/metabolismo , Isoenzimas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Osteoclastos/enzimologia , Osteoclastos/patologia , Hormônio Paratireóideo/administração & dosagem , Baço/efeitos dos fármacos , Fosfatase Ácida Resistente a Tartarato
8.
Bone ; 47(3): 472-9, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20601290

RESUMO

It is now well established that important regulatory interactions occur between the cells in the hematopoietic, immune and skeletal systems (osteoimmunology). B lymphocytes (B cells) are responsible for the generation and production of antibodies or immunoglobulins in the body. Together with T cells these lymphocytes comprise the adaptive immune system, which allows an individual to develop specific responses to an infection and retain memory of that infection, allowing for a faster and more robust response if that same infection occurs again. In addition to this immune function, B cells have a close and multifaceted relationship with bone cells. B cells differentiate from hematopoietic stem cells (HSCs) in supportive niches found on endosteal bone surfaces. Cells in the osteoblast lineage support HSC and B cell differentiation in these niches. B cell differentiation is regulated, at least in part, by a series of transcription factors that function in a temporal manner. While these transcription factors are required for B cell differentiation, their loss causes profound changes in the bone phenotype. This is due, in part, to the close relationship between macrophage/osteoclast and B cell differentiation. Cross talk between B cells and bone cells is reciprocal with defects in the RANKL-RANK, OPG signaling axis resulting in altered bone phenotypes. While the role of B cells during normal bone remodeling appears minimal, activated B cells play an important role in many inflammatory diseases with associated bony changes. This review examines the relationship between B cells and bone cells and how that relationship affects the skeleton and hematopoiesis during health and disease.


Assuntos
Linfócitos B/fisiologia , Doenças Ósseas Metabólicas , Osso e Ossos/citologia , Osso e Ossos/metabolismo , Animais , Linfócitos B/citologia , Doenças Ósseas Metabólicas/metabolismo , Doenças Ósseas Metabólicas/patologia , Doenças Ósseas Metabólicas/fisiopatologia , Osso e Ossos/patologia , Osso e Ossos/fisiopatologia , Diferenciação Celular , Estrogênios/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Humanos , Sistema Imunitário/fisiologia , Osteoprotegerina/metabolismo , Ligante RANK/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Transdução de Sinais/fisiologia , Fatores de Transcrição/metabolismo
9.
Am J Pathol ; 176(5): 2405-13, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20348238

RESUMO

This study addressed the role of impairment of osteoblastic differentiation as a mechanism underlying pathophysiology of the osteogenesis imperfecta (OI). We hypothesized that combination of impaired osteogenic differentiation with increased bone resorption leads to diminished bone mass. By introducing visual markers of distinct stages of osteoblast differentiation, pOBCol3.6GFP (3.6GFP; preosteoblast) and pOBCol2.3GFP (2.3GFP; osteoblast/osteocytes), into the OIM model, we assessed osteoblast maturation and the mechanism of increased osteoclastogenesis. Cultures from oim/oim;2.3GFP mice showed a marked reduction of cells expressing GFP relative to +/+;2.3GFP littermates. No significant difference in expression of 3.6GFP between the +/+ and oim/oim mice was observed. Histological analysis of the oim/oim;3.6GFP mice showed an increased area of GFP-positive cells lining the endocortical surface compared with +/+;3.6GFP mice. In contrast GFP expression was similar between oim/oim;2.3GFP and +/+;2.3GFP mice. These data indicate that the osteoblastic lineage is under continuous stimulation; however, only a proportion of cells attain the mature osteoblast stage. Indeed, immature osteoblasts exhibit a stronger potential to support osteoclast formation and differentiation. We detected a higher Rankl/Opg ratio and higher expression of TNF-alpha in sorted immature osteoblasts. In addition, increased osteoclast formation was observed when osteoclast progenitors were cocultured with oim/oim-derived osteoblasts compared with osteoblasts derived from +/+ mice. Taken together, our data indicate that osteoblast lineage maturation is a critical aspect underlying the pathophysiology of OI.


Assuntos
Osteoblastos/citologia , Osteoclastos/citologia , Osteogênese Imperfeita/metabolismo , Animais , Células da Medula Óssea/citologia , Linhagem da Célula , Modelos Animais de Doenças , Proteínas de Fluorescência Verde/metabolismo , Homozigoto , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Fator de Necrose Tumoral alfa/metabolismo
10.
J Clin Endocrinol Metab ; 95(4): 1991-7, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20156921

RESUMO

CONTEXT: Sclerostin is a negative regulator of bone formation. OBJECTIVE: The aim of the study was to compare serum sclerostin levels in premenopausal and postmenopausal women and evaluate its relationship to estrogen, TH, bone turnover, and bone mass. DESIGN, SETTING, AND PARTICIPANTS: We conducted a cross-sectional observational study of healthy community-dwelling pre- and postmenopausal women. INTERVENTION(S): There were no interventions. MAIN OUTCOME MEASURE(S): We compared serum sclerostin levels in pre- and postmenopausal women and correlated sclerostin levels with female sex hormones, calciotropic hormones, bone turnover markers, and bone mineral density. RESULTS: Premenopausal women were 26.8 yr old, and postmenopausal women were 56.8 yr old. Postmenopausal women had lower values for estradiol (30 +/- 23 vs. 10 +/- 4 pg/ml; P < 0.001), estrone (61 +/- 24 vs. 29 +/- 10 pg/ml; P <0.001), and free estrogen index (FEI) (6 +/- 4 vs. 3 +/- 2 pmol/nmol; P = 0.008) and significantly lower bone mineral density at all sites compared to premenopausal women, with no significant differences in levels of PTH, 25-hydroxy or 1,25-dihydroxy vitamin D levels. Postmenopausal women had significantly higher serum sclerostin levels (1.16 +/- 0.38 ng/ml vs. 0.48 +/- 0.15 ng/ml; P < 0.001). Because most of the premenopausal women were on oral contraceptives, subsequent analyses were limited to postmenopausal women. There were significant negative correlations between sclerostin and FEI and sclerostin and PTH in this group. Using multiple regression analysis, both FEI (beta = -0.629; P = 0.002) and PTH (beta = -0.554; P = 0.004) were found to be independent predictors of sclerostin levels in postmenopausal women. CONCLUSIONS: Our findings suggest that serum sclerostin levels are regulated by both estrogens and PTH in postmenopausal women. These findings need to be explored further in larger prospective studies.


Assuntos
Proteínas Morfogenéticas Ósseas/sangue , Estrogênios/sangue , Hormônio Paratireóideo/sangue , Pós-Menopausa/sangue , Proteínas Adaptadoras de Transdução de Sinal , Adulto , Idoso , Envelhecimento/fisiologia , Biomarcadores , Densidade Óssea/fisiologia , Desenvolvimento Ósseo/fisiologia , Estudos Transversais , Feminino , Marcadores Genéticos , Hormônios Esteroides Gonadais/sangue , Humanos , Pessoa de Meia-Idade , Pré-Menopausa/sangue , Adulto Jovem
11.
Adv Exp Med Biol ; 602: 47-56, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17966387

RESUMO

A regulatory network comprised of transcription factors PU.1, Ikaros, E2A, EBF, and Pax5 control B cell fate specification and differentiation. Early B Cell Factor-1 (EBF-1) is essential for B cell fate specification while Pax5 is required for B cell development. Mice deficient in Pax5 or EBF-1 have a developmental arrest of B cell differentiation at the pro-B cell stage, which results in the absence of mature B cells. We analyzed the bone phenotype of Pax5 and EBF-1 wild-type (+/+) and homozygous mutant (-/-) mice to determine if the loss of these transcription factors regulated bone cell development. Bones from Pax5-/- mice were strikingly osteopenic 15 days after birth, with increased numbers of osteoclasts, and decreased trabecular number. The number of osteoblasts in Pax5-/- bones and their function in vitro were not different from controls. In addition, Pax5 was not expressed by wild-type osteoblasts. To investigate the origin of the in vivo increase in osteoclasts, Pax5-/- or +/+ spleen cells were cultured with M-CSF and RANKL and multinucleated, TRAP' cells counted. Cells from Pax5-/- spleen produced 5-10 times more osteoclasts than did controls. Tibia from EBF-1-/- mice had a striking increase in osteoblasts lining bone surfaces. Consistent with this was an increase in osteoid thickness and in the bone formation rate. This correlated with a 2-fold increase in serum osteocalcin. However, in vitro proliferation and ALP of mutant osteoblasts did not differ from control. In contrast, osteoclast number was similar in 4 week-old +/+ and -/- mice; however, at 12 weeks the number of osteoclasts was more than twice that of controls These data correlated with an increase in bone volume at 12 weeks of age. The most striking aspect of the EBF-1-/- bones was the presence of adipocytes, which filled the marrow space. The adipocytes in the marrow were present at both 4 and 12 weeks of age. Increased fat was also seen in the liver of mutant mice. However, subcutaneous fat was almost absent in EBF-1-/- mice. Importantly, EBF-1 mRNA was expressed in wild-type osteoblasts and in adipocytes. Loss of EBF-1 and Pax5 causes distinct, non-overlapping bone phenotypes. It is important to understand why this network of transcription factors, which are so important for B cell development, have such striking effects on bone cell growth and development.


Assuntos
Linfócitos B/metabolismo , Desenvolvimento Ósseo/imunologia , Proteínas de Ligação a DNA/metabolismo , Osteoclastos/metabolismo , Fator de Transcrição PAX5/metabolismo , Transativadores/metabolismo , Animais , Desenvolvimento Ósseo/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/imunologia , Camundongos , Osteoblastos/metabolismo , Fator de Transcrição PAX5/genética , Fator de Transcrição PAX5/imunologia , Ligante RANK/metabolismo , Baço/metabolismo , Transativadores/genética , Transativadores/imunologia , Fatores de Transcrição/metabolismo
12.
Ann N Y Acad Sci ; 1117: 82-93, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17872391

RESUMO

Mesenchymal lineage cells arise from pluripotent stem cells in the bone marrow (BM) and transition through a series of developmental stages resulting in mature functional cells. This specification results in the development of osteoblast, adipocytes, myoblasts, chondroblasts, and stromal cells (part of the recticular network). The osteoblast developmental pathway is well understood particularly at the later stages of development. However, less is known about the very early stages, where cell fate decisions that lead to commitment to the osteoblast lineage occur. Adipocytes, the cells that produce fat, likely share a common early progenitor with osteoblasts, although little is known about the molecular control of this lineage bifurcation. Growing evidence indicates that transcription factors required for B lymphocyte development from hematopoietic stem cells are critical for proper skeletal development although as yet none have been implicated in osteoblast differentiation. We have discovered that O/E-1, a transcription factor essential for B cell development, is expressed in osteoblasts and plays a critical role in controlling osteoblast development. O/E-1-deficient mice are runted, have increased bone formation parameters, and have a striking increase in osteoblasts. Remarkably, these mice also exhibit a dramatic expansion of adipocytes in the medullary canal of long bones.


Assuntos
Linfócitos B/fisiologia , Osso e Ossos/metabolismo , Adipócitos/metabolismo , Animais , Linfócitos B/metabolismo , Diferenciação Celular , Linhagem da Célula , Linfócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Osteoblastos/metabolismo , Osteocalcina/metabolismo , Osteogênese , Células Precursoras de Linfócitos B , Fatores de Transcrição
13.
J Bone Miner Res ; 21(11): 1704-12, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17002560

RESUMO

UNLABELLED: We examined OVX-induced bone loss in three TLD mouse models. In TLD mice, OVX caused trabecular bone loss equivalent to that of WT. In contrast, cortical bone loss with OVX was variable. We conclude that T lymphocytes do not influence OVX-induced trabecular bone loss. INTRODUCTION: We examined ovariectomy (OVX)-induced bone loss in three T lymphocyte-deficient (TLD) mouse models: nude mice, recombination activating gene 2-deficient (RAG2 KO) mice, and T cell receptor alpha chain-deficient (TCRalpha KO) mice. MATERIALS AND METHODS: Bone mass was examined by DXA, microCT, and histomorphometry. We also examined the effect of OVX on T lymphocytes in the bone marrow and spleens of wildtype (WT) mice and on in vitro osteoclastogenesis and colony forming unit-granulocyte macrophage (CFU-GM) activity in the bone marrow of WT and nude mice. RESULTS: In WT mice, OVX did not alter T lymphocyte number in the bone marrow but did increase T lymphocytes in the spleen. Comparison of bone mass in nude, RAG2 KO, and TCRalpha KO mice with WT as measured by DXA showed decreased femoral bone mass in nude mice and increased vertebral bone mass in RAG2 KO mice. In TCRalpha KO mice, femoral, tibial, and vertebral bone mass were decreased. In vertebrae and long bones, bone loss with OVX was consistently present in WT mice but variably present in TLD mice as measured by DXA. In contrast, microCT and histomorphometry showed similar trabecular bone loss after OVX in all mice. However, femoral cortical bone loss occurred only in WT and RAG2 KO mice. OVX produced similar trabecular bone loss in WT and TCRalpha KO mice and also induced cortical bone loss in both. Histomorphometry showed that TRACP(+) area in bones was increased by OVX in femurs from both WT and nude mice as was in vitro osteoclast-like cell formation and CFU-GM activity. CONCLUSIONS: These results show that OVX caused similar trabecular bone loss in both WT and TLD mice. The ability of DXA and measurement of cortical bone loss to show OVX-induced effects on bone mass was variable. It seems that T lymphocytes are not critical for OVX-induced trabecular bone loss in these mouse models.


Assuntos
Osso e Ossos/fisiologia , Ovariectomia/métodos , Linfócitos T/fisiologia , Animais , Células da Medula Óssea/metabolismo , Osso e Ossos/metabolismo , Feminino , Linfócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Nus , Osteoclastos/metabolismo , Recombinação Genética , Células-Tronco/metabolismo , Linfócitos T/metabolismo
14.
Bone ; 39(5): 991-999, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16782418

RESUMO

A growing body of evidence indicates that megakaryocytes (MK) or their growth factors play a role in skeletal homeostasis. We previously identified a novel regulatory pathway that controls bone formation, which is mediated by MK. In vivo megakaryocytosis resulted in massive bone formation. The co-culture of MK with osteoblasts (OB) resulted in increased OB proliferation in vitro, by a mechanism that required direct cell-to-cell contact. Here, we examined a second MK-mediated pathway that regulates osteoclast (OC) development. We have begun examining the unique inhibitory effect of MK on OC development. Spleen or bone marrow (BM) cells from C57BL/6 mice, as a source of OC precursors, were cultured with M-CSF and RANKL to induce OC development. MK were prepared by culturing fetal liver cells with thrombopoietin and separating cells into MK and non-MK populations. MK were titrated into spleen cell cultures and OC were identified as tartrate-resistant acid phosphatase-positive giant cells with >3 nuclei. There was a significant, P < 0.001, up to 10-fold reduction in OC formed when MK were added to the spleen cell cultures. We determined that 30% (vol:vol) MK conditioned media (CM) were able to completely block OC development from precursors, whereas 3% MK CM resulted in up to a 10-fold reduction in OC development, P < 0.001. These data indicate that a soluble factor(s) was responsible, at least in part, for the inhibition. We examined MK CM for known inhibitors of OC formation, using ELISAs. IL-4 was undetectable in MK CM, whereas IL-10 and IFN-gamma levels were similar in MK and non-MK CM. TGFbeta-1 levels were increased 2-fold in MK CM compared to control CM but were not responsible for the inhibition in OC development. Although, we found a significant increase in the levels of osteoprotegerin (OPG) in MK CM, antibody neutralization studies, MK derived from OPG-deficient mice, and tandem mass spectrophotometry, all confirm that OPG was not responsible for the MK-mediated inhibition of OC development. Overall, these data suggest that an unidentified factor(s) is present in MK CM that inhibits OC development. These studies indicate that MK can play a dual role in skeletal homeostasis by stimulating OB proliferation and simultaneously inhibiting OC development.


Assuntos
Megacariócitos/citologia , Osteoclastos/citologia , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Comunicação Celular , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura/métodos , Fator Estimulador de Colônias de Macrófagos/farmacologia , Megacariócitos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , Ligante RANK/farmacologia , Baço/citologia , Baço/efeitos dos fármacos , Baço/metabolismo , Fator de Crescimento Transformador beta1/metabolismo
15.
J Bone Miner Res ; 21(5): 695-702, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16734384

RESUMO

UNLABELLED: IL-7 is produced by stromal cells in bone marrow and is a major regulator of B and T lymphopoiesis. It is also a direct inhibitor of osteoclastogenesis in vitro. In this study we show that IL-7-deficient mice have increased OC and decreased trabecular bone volume compared with WT mice but mimic WT mice in the amount of trabecular but not cortical bone lost after ovariectomy. INTRODUCTION: Interleukin (IL)-7 is a potent regulator of lymphocyte development, which has significant effects on bone. Bone marrow cell cultures from IL-7 deficient (IL-7KO) mice produced significantly more TRACP(+) osteoclasts (OCs) than did cells from wildtype (WT) mice. A previous study found that treatment of mice with a neutralizing antibody to IL-7 blocked ovariectomy (OVX)-induced bone loss. We examined if differences exist between the bones of WT and IL-7KO mice and if OVX altered bone mass in IL-7KO mice. MATERIALS AND METHODS: Studies were in 2-month-old sham-operated (SHAM) and OVX female mice that were killed 4 weeks after surgery. IL-7KO mice and WT controls were in a C57BL/6 background. Both vertebrae (L(1)) and femora were evaluated by DXA, muCT, and histomorphometry. IL-7KO mice were confirmed as IL-7 deficient by their almost total lack of mature B cells in their bone marrow. RESULTS: There was significantly less trabecular bone volume in the vertebrae of IL-7KO mice than in WT mice. In addition, IL-7KO mice had significantly decreased (p < 0.05) trabecular number (13%) and increased trabecular spacing (15%). OVX decreased vertebral trabecular bone volume (TBV) by 21% (p < 0.05) in WT mice and by 22% (p < 0.05) in IL-7KO mice compared with SHAM. IL-7KO SHAM mice also had significantly less (30%) TBV (TA/TTA) in their femurs, as measured histomorphometrically, than did WT SHAM mice. Femurs from IL-7KO SHAM mice had significantly increased percent OC surface (23%) compared with WT SHAM. As in the vertebrae, OVX significantly decreased femoral TBV in both WT and IL-7KO mice by similar amounts (47% and 48%, respectively, p < 0.05 for both) compared with SHAM. However, OVX decreased cortical bone mass in WT but not in IL-7KO bones. We also examined bone marrow cells from WT and IL-7KO mice. Bone marrow cells from IL-7KO animals showed a significant increase in the number of TRACP(+) osteoclast-like cells (OCLs), which formed in cultures that were stimulated with macrophage-colony stimulating factor (M-CSF) and RANKL (both at 30 ng/ml). However, there was no significant difference in the number of OCLs that formed in B lymphocyte-depleted (B220(-)) bone marrow cell cultures from WT and IL-7KO mice. CONCLUSIONS: IL-7 deficiency in mice caused increased OC number in bone and decreased bone mass. OVX-induced bone loss in IL-7-deficient mice was selective and occurred in trabecular but not cortical bone.


Assuntos
Interleucina-7/fisiologia , Osteoclastos/citologia , Osteoporose/etiologia , Ovariectomia , Absorciometria de Fóton , Animais , Feminino , Citometria de Fluxo , Interleucina-7/genética , Camundongos , Camundongos Knockout
16.
J Bone Miner Res ; 21(1): 67-77, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16355275

RESUMO

UNLABELLED: Murine BM was fractionated using a series of hematopoietic markers to characterize its osteoclast progenitor populations. We found that the early osteoclastogenic activity in total BM was recapitulated by a population of cells contained within the CD11b(-/low) CD45R- CD3- CD115high fraction. INTRODUCTION: Osteoclasts are of hematopoietic origin and they have been shown to share the same lineage as macrophages. We further characterized the phenotype of osteoclast progenitor populations in murine bone marrow (BM) by analyzing their cell surface markers. MATERIALS AND METHODS: We used fluorescence-activated cell sorting (FACS) to identify the subsets of BM cells that contained osteoclast progenitors. We fractionated BM according to several markers and cultured the sorted populations for a period of 2-6 days with macrophage-colony stimulating factor (M-CSF) and RANKL. The numbers of multinucleated osteoclast-like cells (OCLs) that formed in the cultures were counted. RESULTS: We found that the CD45R- CD11b(-/low) population recapitulated the early osteoclastogenic activity of total BM. In addition, although previous experiments indicated that osteoclastogenic activity was enriched within the CD45R+ population, we found that highly purified CD45R+ BM was incapable of differentiating into osteoclasts in vitro. We also found that CD45R- CD11b(high) BM cells were an inefficient source of osteoclast progenitors. However, CD11b was transiently upregulated by cells of the CD45R- CD11b(-/low) fraction early (within 24 h) during culture with M-CSF. Finally, further fractionation of BM using CD115 and CD117 showed that, as osteoclast precursor cells matured, they downregulate CD117 but remain CD115+. Curiously, pure populations of CD117- (CD115high) cells isolated fresh from BM have low osteoclastogenic activity in vitro. CONCLUSIONS: We provided a refined analysis of the precise subpopulations of murine BM that are capable of differentiating into OCLs in vitro when treated with M-CSF and RANKL.


Assuntos
Células da Medula Óssea/fisiologia , Diferenciação Celular/fisiologia , Osteoclastos/fisiologia , Células-Tronco/fisiologia , Animais , Antígenos CD/metabolismo , Antígenos de Diferenciação/metabolismo , Células da Medula Óssea/citologia , Proteínas de Transporte/metabolismo , Proteínas de Transporte/farmacologia , Diferenciação Celular/efeitos dos fármacos , Linhagem da Célula , Células Cultivadas , Citometria de Fluxo/métodos , Fator Estimulador de Colônias de Macrófagos/metabolismo , Fator Estimulador de Colônias de Macrófagos/farmacologia , Masculino , Glicoproteínas de Membrana/metabolismo , Glicoproteínas de Membrana/farmacologia , Camundongos , Osteoclastos/citologia , Osteogênese , Ligante RANK , Receptor Ativador de Fator Nuclear kappa-B , Células-Tronco/citologia
17.
Bone ; 38(5): 678-85, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16309985

RESUMO

Receptor activator of NF-kappaB ligand (RANKL) and interleukin-1 (IL-1) individually plays a critical role in the differentiation and activation of osteoclasts in bone. In addition, both RANKL and IL-1 activate similar signal transduction pathways including p38 MAP kinase and c-Jun NH(2) terminal kinase (JNK). We examined if endogenously produced IL-1 influenced osteoclast-like cell (OCL) formation in murine bone marrow and bone marrow monocyte (BMM) cultures that were stimulated with M-CSF and RANKL. RANKL stimulated OCL formation in a dose-dependent manner in bone marrow cultures, and this response was significantly inhibited by IL-1 RA (100 ng/ml), a specific IL-1 antagonist. Interleukin-1 further increased OCL formation in BMM cultures that were treated with M-CSF (30 ng/ml) and RANKL (1, 3, 10 and 30 ng/ml). In addition, BMM cultures from IL-1 type I receptor-deficient mice, which do not respond to IL-1, demonstrated significantly less OCL formation compared to wild-type BMM cultures. We examined the time course and dose response of IL-1alpha protein expression by ELISA in BMM cultures that were treated with or without M-CSF and RANKL. RANKL dose dependently stimulated IL-1alpha protein significantly (up to 46%) in 6-day cultures. The interaction of RANKL and IL-1 on osteoclastogenesis did not appear significantly dependent on prostaglandin synthesis since PGE(2) expression in the conditioned medium of BMM cultures was nearly undetectable and the PGHS-2 specific inhibitor, NS-398, was without effect. We also investigated the effect of IL-1 on p38 MAP kinase and JNK in BMM cultures. The combination of RANKL and IL-1 had additive effects on JNK but not p38 MAP kinase compared to results in cultures treated with RANKL or IL-1 alone. In addition, SP600125, a specific JNK inhibitor, markedly reduced OCL formation in BMM cultures that were treated with RANKL or the combination of RANKL and IL-1. These findings demonstrate that endogenously produced IL-1 augments the response of bone marrow cells to RANKL, and this effect appears mediated by mechanisms that are associated with enhancement of JNK activity.


Assuntos
Células da Medula Óssea/efeitos dos fármacos , Proteínas de Transporte/farmacologia , Diferenciação Celular , Interleucina-1/fisiologia , Glicoproteínas de Membrana/farmacologia , Osteoclastos/citologia , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Proteína Antagonista do Receptor de Interleucina 1 , Interleucina-1/antagonistas & inibidores , Interleucina-1/farmacologia , MAP Quinase Quinase 4/antagonistas & inibidores , MAP Quinase Quinase 4/metabolismo , Fator Estimulador de Colônias de Macrófagos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Osteoclastos/metabolismo , Ligante RANK , Receptor Ativador de Fator Nuclear kappa-B , Sialoglicoproteínas/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
18.
J Exp Med ; 202(5): 589-95, 2005 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-16147974

RESUMO

Osteoclasts are derived from myeloid lineage cells, and their differentiation is supported by various osteotropic factors, including the tumor necrosis factor (TNF) family member TNF-related activation-induced cytokine (TRANCE). Genetic deletion of TRANCE or its receptor, receptor activator of nuclear factor kappaB (RANK), results in severely osteopetrotic mice with no osteoclasts in their bones. TNF receptor-associated factor (TRAF) 6 is a key signaling adaptor for RANK, and its deficiency leads to similar osteopetrosis. Hence, the current paradigm holds that TRANCE-RANK interaction and subsequent signaling via TRAF6 are essential for the generation of functional osteoclasts. Surprisingly, we show that hematopoietic precursors from TRANCE-, RANK-, or TRAF6-null mice can become osteoclasts in vitro when they are stimulated with TNF-alpha in the presence of cofactors such as TGF-beta. We provide direct evidence against the current paradigm that the TRANCE-RANK-TRAF6 pathway is essential for osteoclast differentiation and suggest the potential existence of alternative routes for osteoclast differentiation.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Células-Tronco Hematopoéticas/efeitos dos fármacos , Osteoclastos/citologia , Osteoclastos/metabolismo , Transdução de Sinais/fisiologia , Fator de Necrose Tumoral alfa/farmacologia , Animais , Proteínas de Transporte/genética , Primers do DNA , Deleção de Genes , Técnicas Histológicas , Linfotoxina-alfa/farmacologia , Masculino , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ligante RANK , Receptor Ativador de Fator Nuclear kappa-B , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator 6 Associado a Receptor de TNF/deficiência , Fator 6 Associado a Receptor de TNF/genética
19.
Mol Aspects Med ; 26(3): 181-201, 2005 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-15811434

RESUMO

Osteoporosis, a key predictor of hip fractures can be treated using a variety of safe and effective interventions. Nevertheless, optimally effective strategies for the prevention of hip fractures must also incorporate efforts to address a broad range of other potentially reversible factors. Hyperthyroidism, anticonvulsants, caffeine and smoking may decrease bone mass and increase fracture risk at any age. In older individuals it is important to also consider additional risk factors, including long-acting benzodiazepines, poor vision and sarcopenia. The presence of sarcopenia, an age-related decline in muscle bulk and quality enhances the risk of frailty and possibly also hip fracture, particularly if associated with diminished functional mobility, lower quadriceps strength and poor balance or body sway. In this review we examine evidence which indicates the presence of endocrine-immune dysregulation in both osteoporosis and sarcopenia. Post-menopausal declines in serum estrogen and androgen levels contribute to increases in local bone levels of cytoclastic cytokines, followed by increased osteoclastogenesis and bone loss. Similarly, the presence of decreased gonadal hormones and IGF-1, combined with unusually high peripheral levels of cytokines, inflammatory mediators and coagulation markers all enhance the risk of sarcopenia and frailty. We propose that a translational research approach which emphasizes common pathophysiologic mechanisms in osteoporosis and sarcopenia could accelerate the speed of discovery of effective strategies for both frailty and hip fracture prevention.


Assuntos
Fraturas do Quadril/imunologia , Fraturas do Quadril/fisiopatologia , Músculos/imunologia , Músculos/patologia , Osteoporose/imunologia , Osteoporose/fisiopatologia , Idoso , Animais , Idoso Fragilizado , Fraturas do Quadril/metabolismo , Fraturas do Quadril/prevenção & controle , Humanos , Músculos/metabolismo , Músculos/fisiopatologia , Osteoporose/metabolismo , Osteoporose/patologia , Fatores de Risco
20.
J Bone Miner Res ; 20(1): 23-9, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15619666

RESUMO

UNLABELLED: We examined the effect of PGE2 on OC formation from spleen cells treated with M-CSF and RANKL. PGE2 decreased OC number at 5-6 days of culture and increased OC number, size, and resorptive activity at 7-8 days. A selective EP2 receptor agonist mimicked these effects. Deletion of the EP2 receptor or depletion of T-cells abrogated the increase in OC number. INTRODUCTION: Prostaglandin E2 (PGE2) has been reported to increase osteoclast (OC) number in spleen cells cultured with RANKL and macrophage-colony-stimulating factor (M-CSF). In this study, we examined the time course of PGE2 effects on spleen cells cultured with RANKL and M-CSF. We then investigated which PGE receptors and cell types were involved in these effects. MATERIALS AND METHODS: Spleen cells were cultured from wildtype C57BL/6 mice and EP2 or EP4 receptor-deficient (-/-) and wildtype (+/+) mice on a mixed genetic background. Spleen cells were cultured with M-CSF and RANKL for 5-9 days with or without PGE2 or selective agonists for the four PGE2 receptors (EP1A, EP2A, EP3A, or EP4A). Some cultures were performed using T-cell-depleted spleen cells. OC number and size were quantitated. OC apoptosis and pit formation were measured at 7 or 8 days. RESULTS: PGE2 decreased the number of OCs formed in the presence of RANKL and M-CSF at 5-6 days of culture and increased OC number at 8-9 days compared with cultures without PGE2. PGE2 also increased OC size at 7 and 8 days, decreased apoptosis of OC at 7 days, and increased pit formation at 8 days. EP1A or EP4A had no effect on OC. EP3A decreased OC number. EP2A mimicked effect of PGE2. EP2(-/-) spleen cells showed no increase in OC number in response to PGE2, whereas deletion of EP4 had no effect. Depletion of T-cells abrogated the late increase of OC number. CONCLUSIONS: We conclude that PGE2 has an initial inhibitory effect on OC formation in spleen cell cultures, possibly mediated by both EP2 and EP3 receptors, and a later stimulatory effect, mediated by the EP2 receptor, possibly acting on T-cells.


Assuntos
Dinoprostona/farmacologia , Osteoclastos/citologia , Receptores de Prostaglandina E/fisiologia , Baço/citologia , Animais , Reabsorção Óssea , Proteínas de Transporte/farmacologia , Contagem de Células , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Feminino , Depleção Linfocítica , Fator Estimulador de Colônias de Macrófagos/farmacologia , Glicoproteínas de Membrana/farmacologia , Camundongos , Camundongos Mutantes , Osteoclastos/efeitos dos fármacos , Ligante RANK , Receptor Ativador de Fator Nuclear kappa-B , Receptores de Prostaglandina E/agonistas , Receptores de Prostaglandina E Subtipo EP2 , Receptores de Prostaglandina E Subtipo EP3 , Baço/efeitos dos fármacos , Linfócitos T/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA