Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
J Immunol ; 204(4): 868-878, 2020 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-31915261

RESUMEN

Osteoclasts (OC) originate from either bone marrow (BM)-resident or circulating myeloid OC progenitors (OCP) expressing the receptor CX3CR1. Multiple lines of evidence argue that OCP in homeostasis and inflammation differ. We investigated the relative contributions of BM-resident and circulating OCP to osteoclastogenesis during homeostasis and fracture repair. Using CX3CR1-EGFP/TRAP tdTomato mice, we found CX3CR1 expression in mononuclear cells, but not in multinucleated TRAP+ OC. However, CX3CR1-expressing cells generated TRAP+ OC on bone within 5 d in CX3CR1CreERT2/Ai14 tdTomato reporter mice. To define the role that circulating cells play in osteoclastogenesis during homeostasis, we parabiosed TRAP tdTomato mice (CD45.2) on a C57BL/6 background with wild-type (WT) mice (CD45.1). Flow cytometry (CD45.1/45.2) demonstrated abundant blood cell mixing between parabionts after 2 wk. At 4 wk, there were numerous tdTomato+ OC in the femurs of TRAP tdTomato mice but almost none in WT mice. Similarly, cultured BM stimulated to form OC demonstrated multiple fluorescent OC in cell cultures from TRAP tdTomato mice, but not from WT mice. Finally, flow cytometry confirmed low-level engraftment of BM cells between parabionts but significant engraftment in the spleens. In contrast, during fracture repair, we found that circulating CX3CR1+ cells migrated to bone, lost expression of CX3CR1, and became OC. These data demonstrate that OCP, but not mature OC, express CX3CR1 during both homeostasis and fracture repair. We conclude that, in homeostasis mature OC derive predominantly from BM-resident OCP, whereas during fracture repair, circulating CX3CR1+ cells can become OC.

2.
J Endocr Soc ; 4(10): bvaa112, 2020 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-33834150
3.
J Immunol ; 203(1): 105-116, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31109956

RESUMEN

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.


Asunto(s)
Enfermedades Óseas/inmunología , Inflamación/inmunología , Osteoclastos/fisiología , Receptor Notch2/metabolismo , Receptor PAR-1/metabolismo , Animales , Anticuerpos Neutralizantes/metabolismo , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteogénesis/genética , Ligando RANK/metabolismo , Receptor Notch2/inmunología , Receptor PAR-1/genética , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
4.
J Clin Invest ; 128(4): 1429-1441, 2018 04 02.
Artículo en Inglés | MEDLINE | ID: mdl-29504949

RESUMEN

Coupling is the process that links bone resorption to bone formation in a temporally and spatially coordinated manner within the remodeling cycle. Several lines of evidence point to the critical roles of osteoclast-derived coupling factors in the regulation of osteoblast performance. Here, we used a fractionated secretomic approach and identified the axon-guidance molecule SLIT3 as a clastokine that stimulated osteoblast migration and proliferation by activating ß-catenin. SLIT3 also inhibited bone resorption by suppressing osteoclast differentiation in an autocrine manner. Mice deficient in Slit3 or its receptor, Robo1, exhibited osteopenic phenotypes due to a decrease in bone formation and increase in bone resorption. Mice lacking Slit3 specifically in osteoclasts had low bone mass, whereas mice with either neuron-specific Slit3 deletion or osteoblast-specific Slit3 deletion had normal bone mass, thereby indicating the importance of SLIT3 as a local determinant of bone metabolism. In postmenopausal women, higher circulating SLIT3 levels were associated with increased bone mass. Notably, injection of a truncated recombinant SLIT3 markedly rescued bone loss after an ovariectomy. Thus, these results indicate that SLIT3 plays an osteoprotective role by synchronously stimulating bone formation and inhibiting bone resorption, making it a potential therapeutic target for metabolic bone diseases.


Asunto(s)
Comunicación Autocrina , Resorción Ósea/metabolismo , Proteínas de la Membrana/metabolismo , Osteoclastos/metabolismo , Osteogénesis , Animales , Enfermedades Óseas Metabólicas/genética , Enfermedades Óseas Metabólicas/metabolismo , Enfermedades Óseas Metabólicas/patología , Resorción Ósea/genética , Resorción Ósea/patología , Diferenciación Celular , Femenino , Humanos , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Osteoblastos/metabolismo , Osteoblastos/patología , Osteoclastos/patología , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Proteínas Roundabout
5.
Bone ; 67: 1-9, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24994594

RESUMEN

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.


Asunto(s)
Resorción Ósea/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Animales , Resorción Ósea/sangre , Resorción Ósea/genética , Colágeno Tipo I/sangre , Femenino , Ratones , Ratones Noqueados , Ovariectomía , Péptidos/sangre , Fosfatidilinositol 3-Quinasas/genética , Unión Proteica/genética , Unión Proteica/fisiología , Proteínas Proto-Oncogénicas c-cbl/genética
6.
J Cell Biochem ; 115(8): 1449-57, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24590570

RESUMEN

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.


Asunto(s)
Resorción Ósea/genética , Catepsina K/genética , Células Madre Hematopoyéticas/metabolismo , Osteogénesis , Animales , Resorción Ósea/patología , Catepsina K/metabolismo , Curación de Fractura/genética , Células Madre Hematopoyéticas/patología , Ratones , Ratones Noqueados , Osteoclastos/metabolismo , Osteoclastos/patología , Osteopetrosis/genética , Osteopetrosis/patología
7.
J Cell Biochem ; 115(7): 1277-89, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24470255

RESUMEN

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.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Fosfatidilinositol 3-Quinasa Clase Ia/metabolismo , Osteoclastos/citología , Proteínas Proto-Oncogénicas c-cbl/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Animales , Densidad Ósea/genética , Remodelación Ósea/genética , Resorción Ósea/genética , Diferenciación Celular/genética , Movimiento Celular/genética , Supervivencia Celular/genética , Células Cultivadas , Cromonas/farmacología , Fosfatidilinositol 3-Quinasa Clase Ia/biosíntesis , Fosfatidilinositol 3-Quinasa Clase Ia/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Morfolinas/farmacología , Mutación , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación/efectos de los fármacos , Unión Proteica , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-cbl/biosíntesis , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/antagonistas & inhibidores , Proteínas Proto-Oncogénicas p21(ras)/biosíntesis , Ligando RANK/farmacología , Transducción de Señal/genética
8.
J Bone Miner Res ; 28(5): 1203-13, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23165930

RESUMEN

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.


Asunto(s)
Células de la Médula Ósea/citología , Células Dendríticas/citología , Macrófagos/citología , Osteoclastos/citología , Animales , Masculino , Ratones , Ratones Endogámicos C57BL
9.
J Bone Miner Res ; 28(1): 135-49, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22893614

RESUMEN

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.


Asunto(s)
Antígeno CD11b/metabolismo , Antígenos CD18/metabolismo , Diferenciación Celular , Proteínas de Unión al ADN/metabolismo , Osteoclastos/citología , Transducción de Señal , Células Madre/citología , Animales , Recuento de Células , Diferenciación Celular/efectos de los fármacos , Fémur/diagnóstico por imagen , Fémur/efectos de los fármacos , Fémur/metabolismo , Fibrinógeno/metabolismo , Fibrinógeno/farmacología , Humanos , Integrina beta3/metabolismo , Ratones , Ratones Endogámicos C57BL , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Tamaño de los Órganos/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Osteogénesis/efectos de los fármacos , Fenotipo , Proteínas Proto-Oncogénicas c-bcl-6 , Ligando RANK/farmacología , Transducción de Señal/efectos de los fármacos , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Transcripción Genética/efectos de los fármacos , Microtomografía por Rayos X
10.
J Bone Miner Res ; 27(5): 1030-42, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22258693

RESUMEN

Interleukin-7 is a critical cytokine for lymphoid development and a direct inhibitor of in vitro osteoclastogenesis in murine bone marrow cultures. To explore the role of IL-7 in bone, we generated transgenic mouse lines bearing the 2.3-kb rat collagen 1α1 promoter driving the expression of human IL-7 specifically in osteoblasts. In addition, we crossed these mice with IL-7-deficient mice to determine if the alterations in lymphopoiesis, bone mass, and osteoclast formation observed in the IL-7 knockout (KO) mice could be rescued by osteoblast-specific overexpression of IL-7. Here, we show that mice overexpressing human IL-7 in the osteoblast lineage showed increased trabecular bone volume in vivo by µCT and decreased osteoclast formation in vitro. Furthermore, targeted overexpression of IL-7 in osteoblasts rescued the osteopenic bone phenotype and B-cell development of IL-7 KO mice but did not have an effect on T lymphopoiesis, which occurs in the periphery. The bone phenotypes in IL-7 KO mice and targeted IL-7-overexpressing mouse models were observed only in females. These results likely reflect both direct inhibitory effects of IL-7 on osteoclastogenesis in vivo and sex-specific differences in responses to IL-7.


Asunto(s)
Regulación de la Expresión Génica , Interleucina-7/deficiencia , Interleucina-7/genética , Osteoblastos/metabolismo , Animales , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Humanos , Linfopoyesis , Ratones , Ratones Noqueados , Ratones Transgénicos , Fenotipo , Reacción en Cadena de la Polimerasa , Ratas , Factores Sexuales
11.
J Bone Miner Res ; 26(6): 1207-16, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21611963

RESUMEN

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.


Asunto(s)
Células de la Médula Ósea/citología , Diferenciación Celular/efectos de los fármacos , Células Madre Hematopoyéticas/citología , Osteoclastos/citología , Osteoclastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Hormona Paratiroidea/farmacología , Fosfatasa Ácida/metabolismo , Animales , Bioensayo , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Resorción Ósea/metabolismo , Resorción Ósea/patología , Bovinos , Tamaño de la Célula/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/metabolismo , Isoenzimas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Osteoclastos/enzimología , Osteoclastos/patología , Hormona Paratiroidea/administración & dosificación , Bazo/efectos de los fármacos , Fosfatasa Ácida Tartratorresistente
12.
Bone ; 47(3): 472-9, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20601290

RESUMEN

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.


Asunto(s)
Linfocitos B/fisiología , Enfermedades Óseas Metabólicas , Huesos/citología , Huesos/metabolismo , Animales , Linfocitos B/citología , Enfermedades Óseas Metabólicas/metabolismo , Enfermedades Óseas Metabólicas/patología , Enfermedades Óseas Metabólicas/fisiopatología , Huesos/patología , Huesos/fisiopatología , Diferenciación Celular , Estrógenos/metabolismo , Células Madre Hematopoyéticas/metabolismo , Humanos , Sistema Inmunológico/fisiología , Osteoprotegerina/metabolismo , Ligando RANK/metabolismo , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo
13.
Am J Pathol ; 176(5): 2405-13, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20348238

RESUMEN

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.


Asunto(s)
Osteoblastos/citología , Osteoclastos/citología , Osteogénesis Imperfecta/metabolismo , Animales , Células de la Médula Ósea/citología , Linaje de la Célula , Modelos Animales de Enfermedad , Proteínas Fluorescentes Verdes/metabolismo , Homocigoto , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutación , Factor de Necrosis Tumoral alfa/metabolismo
14.
J Clin Endocrinol Metab ; 95(4): 1991-7, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20156921

RESUMEN

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.


Asunto(s)
Proteínas Morfogenéticas Óseas/sangre , Estrógenos/sangre , Hormona Paratiroidea/sangre , Posmenopausia/sangre , Proteínas Adaptadoras Transductoras de Señales , Adulto , Anciano , Envejecimiento/fisiología , Biomarcadores , Densidad Ósea/fisiología , Desarrollo Óseo/fisiología , Estudios Transversales , Femenino , Marcadores Genéticos , Hormonas Esteroides Gonadales/sangre , Humanos , Persona de Mediana Edad , Premenopausia/sangre , Adulto Joven
15.
Bone ; 44(4): 537-46, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19130908

RESUMEN

Ebf1 is a transcription factor essential for B cell fate specification and function and important for the development of olfactory sensory neurons. We show here that Ebf1 also plays an important role in regulating osteoblast and adipocyte development in vivo. Ebf1 mRNA and protein is expressed in MSCs, in OBs at most stages of differentiation, and in adipocytes. Tibiae and femora from Ebf1(-/-) mice had a striking increase in all bone formation parameters examined including the number of OBs, osteoid volume, and bone formation rate. Serum osteocalcin, a marker of bone formation, was significantly elevated in mutant mice. The numbers of osteoclasts in bone were normal in younger (4 week-old) Ebf1(-/-) mice but increased in older (12 week-old) Ebf1(-/-) mice. This correlated well with in vitro osteoclast development from bone marrow cells. In addition to the increased osteoblastogenesis, there was a dramatic increase in adipocyte numbers in the bone marrow of Ebf1(-/-) mice. Increased adiposity was also seen histologically in the liver but not in the spleen of these mice, and accompanied by decreased deposition of adipose to subcutaneous sites. Thus Ebf1-deficient mice appear to be a new model of lipodystrophy. Ebf1 is a rare example of a transcription factor that regulates both the osteoblast and adipocyte lineages similarly.


Asunto(s)
Adipocitos/citología , Diferenciación Celular/fisiología , Osteoblastos/citología , Osteogénesis/fisiología , Transactivadores/metabolismo , Adipocitos/metabolismo , Animales , Western Blotting , Linaje de la Célula , Inmunohistoquímica , Ratones , Ratones Noqueados , Osteoblastos/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transactivadores/genética
16.
Adv Exp Med Biol ; 602: 47-56, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17966387

RESUMEN

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.


Asunto(s)
Linfocitos B/metabolismo , Desarrollo Óseo/inmunología , Proteínas de Unión al ADN/metabolismo , Osteoclastos/metabolismo , Factor de Transcripción PAX5/metabolismo , Transactivadores/metabolismo , Animales , Desarrollo Óseo/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Ratones , Osteoblastos/metabolismo , Factor de Transcripción PAX5/genética , Factor de Transcripción PAX5/inmunología , Ligando RANK/metabolismo , Bazo/metabolismo , Transactivadores/genética , Transactivadores/inmunología , Factores de Transcripción/metabolismo
18.
Ann N Y Acad Sci ; 1117: 82-93, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17872391

RESUMEN

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.


Asunto(s)
Linfocitos B/fisiología , Huesos/metabolismo , Adipocitos/metabolismo , Animales , Linfocitos B/metabolismo , Diferenciación Celular , Linaje de la Célula , Linfocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Osteoblastos/metabolismo , Osteocalcina/metabolismo , Osteogénesis , Células Precursoras de Linfocitos B , Factores de Transcripción
19.
J Bone Miner Res ; 21(11): 1704-12, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17002560

RESUMEN

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.


Asunto(s)
Huesos/fisiología , Ovariectomía/métodos , Linfocitos T/fisiología , Animales , Células de la Médula Ósea/metabolismo , Huesos/metabolismo , Femenino , Linfocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Osteoclastos/metabolismo , Recombinación Genética , Células Madre/metabolismo , Linfocitos T/metabolismo
20.
Bone ; 39(5): 991-999, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16782418

RESUMEN

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.


Asunto(s)
Megacariocitos/citología , Osteoclastos/citología , Animales , Células de la Médula Ósea/citología , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Comunicación Celular , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo/métodos , Factor Estimulante de Colonias de Macrófagos/farmacología , Megacariocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteoblastos/citología , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , Ligando RANK/farmacología , Bazo/citología , Bazo/efectos de los fármacos , Bazo/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...