Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
1.
J Immunol ; 204(4): 868-878, 2020 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-31915261

RESUMO

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 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
3.
Handb Exp Pharmacol ; 262: 177-230, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32006259

RESUMO

Cytokines and hematopoietic growth factors have traditionally been thought of as regulators of the development and function of immune and blood cells. However, an ever-expanding number of these factors have been discovered to have major effects on bone cells and the development of the skeleton in health and disease (Table 1). In addition, several cytokines have been directly linked to the development of osteoporosis in both animal models and in patients. In order to understand the mechanisms regulating bone cells and how this may be dysregulated in disease states, it is necessary to appreciate the diverse effects that cytokines and inflammation have on osteoblasts, osteoclasts, and bone mass. This chapter provides a broad overview of this topic with extensive references so that, if desired, readers can access specific references to delve into individual topics in greater detail.


Assuntos
Citocinas , Osteoporose , Animais , Osso e Ossos , Humanos , Osteoblastos/química , Osteoclastos/química
4.
J Biol Chem ; 291(8): 3882-94, 2016 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-26703472

RESUMO

Continuous parathyroid hormone (PTH) blocks its own osteogenic actions in marrow stromal cell cultures by inducing Cox2 and receptor activator of nuclear factor κB ligand (RANKL) in the osteoblastic lineage cells, which then cause the hematopoietic lineage cells to secrete an inhibitor of PTH-stimulated osteoblast differentiation. To identify this inhibitor, we used bone marrow macrophages (BMMs) and primary osteoblasts (POBs) from WT and Cox2 knock-out (KO) mice. Conditioned medium (CM) from RANKL-treated WT, but not KO, BMMs blocked PTH-stimulated cAMP production in POBs. Inhibition was reversed by pertussis toxin (PTX), which blocks Gαi/o activation. Saa3 was the most highly differentially expressed gene in a microarray comparison of RANKL-treated WT versus Cox2 KO BMMs, and RANKL induced Saa3 protein secretion only from WT BMMs. CM from RANKL-stimulated BMMs with Saa3 knockdown did not inhibit PTH-stimulated responses in POBs. SAA added to POBs inhibited PTH-stimulated cAMP responses, which was reversed by PTX. Selective agonists and antagonists of formyl peptide receptor 2 (Fpr2) suggested that Fpr2 mediated the inhibitory actions of Saa3 on osteoblasts. In BMMs committed to become osteoclasts by RANKL treatment, Saa3 expression peaked prior to appearance of multinucleated cells. Flow sorting of WT marrow revealed that Saa3 was secreted only from the RANKL-stimulated B220(-) CD3(-)CD11b(-/low) CD115(+) preosteoclast population. We conclude that Saa3 secretion from preosteoclasts, induced by RANKL in a Cox2-dependent manner, inhibits PTH-stimulated cAMP signaling and osteoblast differentiation via Gαi/o signaling. The induction of Saa3 by PTH may explain the suppression of bone formation when PTH is applied continuously and may be a new therapeutic target for osteoporosis.


Assuntos
AMP Cíclico/metabolismo , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Hormônio Paratireóideo/farmacologia , Sistemas do Segundo Mensageiro/efeitos dos fármacos , Proteína Amiloide A Sérica/metabolismo , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , AMP Cíclico/genética , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Camundongos , Camundongos Knockout , Osteoblastos/citologia , Osteoclastos/citologia , Osteogênese/efeitos dos fármacos , Osteogênese/genética , Hormônio Paratireóideo/metabolismo , Ligante RANK/genética , Ligante RANK/metabolismo , Receptores de Formil Peptídeo/genética , Receptores de Formil Peptídeo/metabolismo , Sistemas do Segundo Mensageiro/genética , Proteína Amiloide A Sérica/genética
5.
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
6.
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
7.
J Orthop Res ; 42(6): 1231-1243, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38111181

RESUMO

Osteoporosis is a chronic disease characterized by reduced bone mass and increased fracture risk, estimated to affect over 10 million people in the United States alone. Drugs used to treat bone loss often come with significant limitations and/or long-term safety concerns. Proteoglycan-4 (PRG4, also known as lubricin) is a mucin-like glycoprotein best known for its boundary lubricating function of articular cartilage. In more recent years, it has been shown that PRG4 has anti-inflammatory properties, contributes to the maintenance of subchondral bone integrity, and patients with PRG4 mutations are osteopenic. However, it remains unknown how PRG4 impacts mechanical and material properties of bone. Therefore, our objective was to perform a phenotyping study of bone in a Prg4 gene trap (GT) mouse (PRG4 deficient). We found that femurs of Prg4 GT mice have altered mechanical, structural, and material properties relative to wildtype littermates. Additionally, Prg4 GT mice have a greater number of calvarial osteoclasts than wildtype mice, but do not have a notable inflammatory serum profile. Finally, Prg4 GT mice do not have an altered rate of bone formation, and exogenous recombinant human PRG4 (rhPRG4) administration inhibited osteoclastogenesis in vitro, suggesting that the skeletal phenotype may be due to changes in bone resorption. Overall, this work demonstrates that PRG4 deficiency affects several integral properties of bone structure, mechanics, and skeletal cell activity, and provides the foundation and insight toward future work evaluating PRG4 as a potential therapeutic target in treating bone loss.


Assuntos
Osteoclastos , Osteogênese , Proteoglicanas , Animais , Osteogênese/efeitos dos fármacos , Osteoclastos/efeitos dos fármacos , Camundongos , Humanos , Masculino , Camundongos Endogâmicos C57BL , Crânio , Feminino , Proteínas Recombinantes/farmacologia , Proteínas Recombinantes/uso terapêutico , Fêmur/efeitos dos fármacos
8.
Nat Med ; 12(12): 1403-9, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17128270

RESUMO

Matrix-producing osteoblasts and bone-resorbing osteoclasts maintain bone homeostasis. Osteoclasts are multinucleated, giant cells of hematopoietic origin formed by the fusion of mononuclear pre-osteoclasts derived from myeloid cells. Fusion-mediated giant cell formation is critical for osteoclast maturation; without it, bone resorption is inefficient. To understand how osteoclasts differ from other myeloid lineage cells, we previously compared global mRNA expression patterns in these cells and identified genes of unknown function predominantly expressed in osteoclasts, one of which is the d2 isoform of vacuolar (H(+)) ATPase (v-ATPase) V(0) domain (Atp6v0d2). Here we show that inactivation of Atp6v0d2 in mice results in markedly increased bone mass due to defective osteoclasts and enhanced bone formation. Atp6v0d2 deficiency did not affect differentiation or the v-ATPase activity of osteoclasts. Rather, Atp6v0d2 was required for efficient pre-osteoclast fusion. Increased bone formation was probably due to osteoblast-extrinsic factors, as Atp6v02 was not expressed in osteoblasts and their differentiation ex vivo was not altered in the absence of Atp6v02. Our results identify Atp6v0d2 as a regulator of osteoclast fusion and bone formation, and provide genetic data showing that it is possible to simultaneously inhibit osteoclast maturation and stimulate bone formation by therapeutically targeting the function of a single gene.


Assuntos
Osteoclastos/fisiologia , Osteogênese/fisiologia , Bombas de Próton/genética , ATPases Vacuolares Próton-Translocadoras/fisiologia , Animais , Diferenciação Celular/efeitos dos fármacos , Fusão Celular , Células Cultivadas , Humanos , Fator Estimulador de Colônias de Macrófagos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteoclastos/citologia , Osteoclastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Osteogênese/genética , Isoformas de Proteínas , Estrutura Terciária de Proteína , Ligante RANK/farmacologia , ATPases Vacuolares Próton-Translocadoras/química
9.
Bone ; 168: 116296, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-34942359

RESUMO

Bone remodeling occurs through the interactions of three major cell lineages, osteoblasts, which mediate bone formation, osteocytes, which derive from osteoblasts, sense mechanical force and direct bone turnover, and osteoclasts, which mediate bone resorption. However, multiple additional cell types within the bone marrow, including macrophages, T lymphocytes and B lymphocytes influence the process. The bone marrow microenvironment, which is supported, in part, by bone cells, forms a nurturing network for B lymphopoiesis. In turn, developing B lymphocytes influence bone cells. Bone health during homeostasis depends on the normal interactions of bone cells with other lineages in the bone marrow. In disease state these interactions become pathologic and can cause abnormal function of bone cells and inadequate repair of bone after a fracture. This review summarizes what is known about the development of B lymphocytes and the interactions of B lymphocytes with bone cells in both health and disease.


Assuntos
Reabsorção Óssea , Osteócitos , Humanos , Osteócitos/metabolismo , Osteoclastos/metabolismo , Osteoblastos/metabolismo , Reabsorção Óssea/metabolismo , Remodelação Óssea/fisiologia , Linfócitos B
10.
Endocr Rev ; 29(4): 403-40, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18451259

RESUMO

Bone and the immune system are both complex tissues that respectively regulate the skeleton and the body's response to invading pathogens. It has now become clear that these organ systems often interact in their function. This is particularly true for the development of immune cells in the bone marrow and for the function of bone cells in health and disease. Because these two disciplines developed independently, investigators in each don't always fully appreciate the significance that the other system has on the function of the tissue they are studying. This review is meant to provide a broad overview of the many ways that bone and immune cells interact so that a better understanding of the role that each plays in the development and function of the other can develop. It is hoped that an appreciation of the interactions of these two organ systems will lead to better therapeutics for diseases that affect either or both.


Assuntos
Osso e Ossos/fisiologia , Comunicação Celular/imunologia , Sistema Imunitário/fisiologia , Animais , Linfócitos B/fisiologia , Transplante de Medula Óssea/fisiologia , Remodelação Óssea/imunologia , Remodelação Óssea/fisiologia , Comunicação Celular/fisiologia , Diferenciação Celular , Citocinas/fisiologia , Hematopoese/fisiologia , Humanos , Megacariócitos/fisiologia , Modelos Biológicos , Osteoblastos/fisiologia , Osteoclastos/fisiologia , Células-Tronco/fisiologia
11.
Bone ; 165: 116576, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36195243

RESUMO

Osteoclasts are the only cells that can efficiently resorb bone. They do so by sealing themselves on to bone and removing the mineral and organic components. Osteoclasts are essential for bone homeostasis and are involved in the development of diseases associated with decreased bone mass, like osteoporosis, or abnormal bone turnover, like Paget's disease of bone. In addition, compromise of their development or resorbing machinery is pathogenic in multiple types of osteopetrosis. However, osteoclasts also have functions other than bone resorption. Like cells of the innate immune system, they are derived from myeloid precursors and retain multiple immune cell properties. In addition, there is now strong evidence that osteoclasts regulate osteoblasts through a process known as coupling, which coordinates rates of bone resorption and bone formation during bone remodeling. In this article we review the non-resorbing functions of osteoclasts and highlight their importance in health and disease.


Assuntos
Reabsorção Óssea , Osteoclastos , Humanos , Osteoclastos/fisiologia , Osteoblastos , Remodelação Óssea , Osso e Ossos
12.
Endocrinology ; 163(9)2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-35880727

RESUMO

Many metabolic bone diseases arise as a result excessive osteoclastic bone resorption, which has motivated efforts to identify new molecular targets that can inhibit the formation or activity of these bone-resorbing cells. Mounting evidence indicates that the transcription factor Runx1 acts as a transcriptional repressor of osteoclast formation. Prior studies using a conditional knockout approach suggested that Runx1 in osteoclast precursors acts as an inhibitor of osteoclastogenesis; however, the effects of upregulation of Runx1 on osteoclast formation remain unknown. In this study, we investigated the skeletal effects of conditional overexpression of Runx1 in preosteoclasts by crossing novel Runx1 gain-of-function mice (Rosa26-LSL-Runx1) with LysM-Cre transgenic mice. We observed a sex-dependent effect whereby overexpression of Runx1 in female mice increased trabecular bone microarchitectural indices and improved torsion biomechanical properties. These effects were likely mediated by delayed osteoclastogenesis and decreased bone resorption. Transcriptomics analyses during osteoclastogenesis revealed a distinct transcriptomic profile in the Runx1-overexpressing cells, with enrichment of genes related to redox signaling, apoptosis, osteoclast differentiation, and bone remodeling. These data further confirm the antiosteoclastogenic activities of Runx1 and provide new insight into the molecular targets that may mediate these effects.


Assuntos
Reabsorção Óssea , Osteoclastos , Animais , Densidade Óssea , Reabsorção Óssea/genética , Reabsorção Óssea/metabolismo , Diferenciação Celular/genética , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Feminino , Camundongos , Osteogênese/genética , Ligante RANK/metabolismo
13.
J Biol Chem ; 285(4): 2834-46, 2010 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-19933269

RESUMO

FGF2 transgenic mice were developed in which type I collagen regulatory sequences drive the nuclear high molecular weight FGF2 isoforms in osteoblasts (TgHMW). The phenotype of TgHMW mice included dwarfism, decreased bone mineral density (BMD), osteomalacia, and decreased serum phosphate (P(i)). When TgHMW mice were fed a high P(i) diet, BMD was increased, and dwarfism was partially reversed. The TgHMW phenotype was similar to mice overexpressing FGF23. Serum FGF23 was increased in TgHMW mice. Fgf23 mRNA in bones and fibroblast growth factor receptors 1c and 3c and Klotho mRNAs in kidneys were increased in TgHMW mice, whereas the renal Na(+)/P(i) co-transporter Npt2a mRNA was decreased. Immunohistochemistry and Western blot analyses of TgHMW kidneys showed increased KLOTHO and decreased NPT2a protein. The results suggest that overexpression of HMW FGF2 increases FGF23/FGFR/KLOTHO signaling to down-regulate NPT2a, causing P(i) wasting, osteomalacia, and decreased BMD. We assessed whether HMW FGF2 expression was altered in the Hyp mouse, a mouse homolog of the human disease X-linked hypophosphatemic rickets/osteomalacia. Fgf2 mRNA was increased in bones, and Western blots showed increased FGF2 protein in nuclear fractions from osteoblasts of Hyp mice. In addition, immunohistochemistry demonstrated co-localization of FGF23 and HMW FGF2 protein in osteoblasts and osteocytes from Hyp mice. This study reveals a novel mechanism of regulation of the FGF23-P(i) homeostatic axis.


Assuntos
Fator 2 de Crescimento de Fibroblastos/genética , Fator 2 de Crescimento de Fibroblastos/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Glucuronidase/metabolismo , Hipofosfatemia/metabolismo , Osteomalacia/metabolismo , Absorciometria de Fóton , Animais , Núcleo Celular/metabolismo , Fator 2 de Crescimento de Fibroblastos/química , Fator de Crescimento de Fibroblastos 23 , Homeostase/fisiologia , Humanos , Hipofosfatemia/fisiopatologia , Isomerismo , Rim/fisiologia , Proteínas Klotho , Masculino , Camundongos , Camundongos Transgênicos , Peso Molecular , Osteoblastos/fisiologia , Osteomalacia/fisiopatologia , Fenótipo , Fosfatos/metabolismo , Regiões Promotoras Genéticas/fisiologia , Crânio/diagnóstico por imagem , Crânio/patologia , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIa/metabolismo , Microtomografia por Raio-X
14.
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
15.
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
16.
J Clin Invest ; 131(5)2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-33645543

RESUMO

Osteoporosis is a serious clinical problem that often follows the accelerated bone loss that occurs after the estrogen withdrawal of menopause. In order to better understand the mechanism that produces estrogen withdrawal-induced bone loss, Yu and Pal et al., as reported in this issue of the JCI, examined mice that underwent ovariectomy (OVX). In C57BL/6 mice with enhanced Th17 cells in gut tissue, the authors demonstrated that OVX increased migration of TNF-expressing Th17 cells from the gut to the bone marrow. Furthermore, they found that manipulation of the pathways by which lymphocytes migrate and home to bone marrow prevented the increase of TNF+, Th17 cells in bone marrow after OVX in mice and the trabecular, but not cortical, bone loss in this model. These results argue that interactions of the gut microbiota with the immune system are involved in the effects of estrogen withdrawal on trabecular bone.


Assuntos
Microbioma Gastrointestinal , Osteoporose Pós-Menopausa , Animais , Osso e Ossos , Feminino , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Ovariectomia , Células Th17
17.
J Endocr Soc ; 5(9): bvab069, 2021 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-34268462

RESUMO

BACKGROUND: Giant prolactinomas are an exceedingly uncommon type of pituitary adenomas that usually occur in men, and cause extremely high prolactin levels and mass-related symptoms. Rarely, patients may experience neurological deficits resembling ischemic events. METHODS: We describe an unusual case of a young man who presented with stroke-like symptoms and was found to have a giant prolactinoma. CLINICAL CASE: A 25-year-old man presented with left facial droop and gradually progressing upper and lower extremity weakness for evaluation of stroke. He reported recent weight gain and erectile dysfunction. Physical examination revealed left homonymous hemianopsia, left VII nerve palsy, and left hemiparesis. Magnetic resonance imaging of the brain showed an enormous mass in the sella turcica, which invaded the sphenoid sinus and right side of the skull base. Prolactin level was elevated at 13 580 ng/mL, and the testosterone level was low. The patient was started on cabergoline and had marked improvement in his symptoms in a few months. Fifteen months after starting treatment, he has had more than 90% reduction in tumor volume and a 93% reduction in prolactin level. CONCLUSION: Giant prolactinomas are uncommon and present with compressive symptoms that can be mistaken for a stroke. Our case is a unique report of a facial nerve palsy and hemiparesis secondary to giant prolactinoma in the absence of stroke or pituitary apoplexy.

18.
J Bone Miner Res ; 36(6): 1104-1116, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33567098

RESUMO

Sexual dimorphism of the skeleton is well documented. At maturity, the male skeleton is typically larger and has a higher bone density than the female skeleton. However, the underlying mechanisms for these differences are not completely understood. In this study, we examined sexual dimorphism in the formation of osteoclasts between cells from female and male mice. We found that the number of osteoclasts in bones was greater in females. Similarly, in vitro osteoclast differentiation was accelerated in female osteoclast precursor (OCP) cells. To further characterize sex differences between female and male osteoclasts, we performed gene expression profiling of cultured, highly purified, murine bone marrow OCPs that had been treated for 3 days with macrophage colony-stimulating factor (M-CSF) and receptor activator of NF-κB ligand (RANKL). We found that 125 genes were differentially regulated in a sex-dependent manner. In addition to genes that are contained on sex chromosomes, transcriptional sexual dimorphism was found to be mediated by genes involved in innate immune and inflammatory response pathways. Furthermore, the NF-κB-NFATc1 axis was activated earlier in female differentiating OCPs, which partially explains the differences in transcriptomic sexual dimorphism in these cells. Collectively, these findings identify multigenic sex-dependent intrinsic difference in differentiating OCPs, which results from an altered response to osteoclastogenic stimulation. In humans, these differences could contribute to the lower peak bone mass and increased risk of osteoporosis that females demonstrate relative to males. © 2021 American Society for Bone and Mineral Research (ASBMR).


Assuntos
Osteoclastos , Caracteres Sexuais , Animais , Células da Medula Óssea , Diferenciação Celular , Células Cultivadas , Feminino , Fator Estimulador de Colônias de Macrófagos , Masculino , Camundongos , Fatores de Transcrição NFATC , Osteogênese , Ligante RANK
19.
Biochem Biophys Res Commun ; 403(1): 73-8, 2010 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-21040703

RESUMO

Bone homeostasis is maintained through the balanced action of bone-forming osteoblasts and bone-resorbing osteoclasts. Under pathological conditions or with age, excessive bone loss is often observed due to increased bone resorption. Since osteoclasts are the primary cells in the body that can resorb bone, molecular understanding of osteoclast fate has important clinical implications. Over the past 20 years, many molecular players that govern osteoclast differentiation during normal development have been identified. However, whether the same molecules regulate bone loss occurring under pathological conditions remains largely unknown. We report here that although ATP6v0d2-deficient (ATP6v0d2 KO) mice exhibit an osteopetrotic phenotype due to inefficient osteoclast maturation, this deficiency fails to protect mice from ovariectomy (OVX)-induced bone loss, a model for post-menopause-associated osteoporosis. Moreover, we show that an OVX-induced increase in the number of colony forming unit-granulocyte/macrophage (CFU-GM) in bone marrow cells and subsequent osteoclast formation in vitro was not affected in the absence of ATP6v0d2. However, even after OVX, formation of large osteoclasts (>100 µm in diameter) with actin rings was still reduced in the absence of ATP6v0d2. Taken together, these findings suggest that the critical role of ATP6v0d2 may be limited to the control of bone homeostasis under normal development, and that OVX-induced bone loss is likely to be governed mostly by the increase in osteoclast precursors rather than increased efficiency of osteoclast maturation.


Assuntos
Densidade Óssea , Osso e Ossos/citologia , Diferenciação Celular , ATPases Vacuolares Próton-Translocadoras/fisiologia , Animais , Camundongos , Camundongos Knockout , Osteoclastos/citologia , Osteoporose/metabolismo , Osteoporose/patologia , ATPases Vacuolares Próton-Translocadoras/genética
20.
Cells ; 9(9)2020 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-32932615

RESUMO

Osteoclasts are the principal mediators of bone resorption. They form through the fusion of mononuclear precursor cells under the principal influence of the cytokines macrophage colony stimulating factor (M-CSF, aka CSF-1) and receptor activator of NF-κB ligand (RANKL, aka TNFSF11). Sexual dimorphism in the development of the skeleton and in the incidence of skeletal diseases is well described. In general, females, at any given age, have a lower bone mass than males. The reasons for the differences in the bone mass of the skeleton between women and men at various ages, and the incidence of certain metabolic bone diseases, are multitude, and include the actions of sex steroids, genetics, age, environment and behavior. All of these influence the rate that osteoclasts form, resorb and die, and frequently produce different effects in females and males. Hence, a variety of factors are responsible for the sexual dimorphism of the skeleton and the activity of osteoclasts in bone. This review will provide an overview of what is currently known about these factors and their effects on osteoclasts.


Assuntos
Osteoclastos/metabolismo , Caracteres Sexuais , Androgênios/metabolismo , Animais , Reabsorção Óssea/metabolismo , Estrogênios/metabolismo , Feminino , Expressão Gênica , Humanos , Inflamação/metabolismo , Masculino , Camundongos , Fenótipo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA