Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Clin Invest ; 131(6)2021 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-33720039

RESUMO

In rheumatoid arthritis (RA), osteoclastic bone resorption causes structural joint damage as well as periarticular and systemic bone loss. Periarticular bone loss is one of the earliest indices of RA, often preceding the onset of clinical symptoms via largely unknown mechanisms. Excessive osteoclastogenesis induced by receptor activator of NF-κB ligand (RANKL) expressed by synovial fibroblasts causes joint erosion, whereas the role of RANKL expressed by lymphocytes in various types of bone damage has yet to be elucidated. In the bone marrow of arthritic mice, we found an increase in the number of RANKL-expressing plasma cells, which displayed an ability to induce osteoclastogenesis in vitro. Genetic ablation of RANKL in B-lineage cells resulted in amelioration of periarticular bone loss, but not of articular erosion or systemic bone loss, in autoimmune arthritis. We also show conclusive evidence for the critical contribution of synovial fibroblast RANKL to joint erosion in collagen-induced arthritis on the arthritogenic DBA/1J background. This study highlights the importance of plasma-cell RANKL in periarticular bone loss in arthritis and provides mechanistic insight into the early manifestation of bone lesion induced by autoimmunity.


Assuntos
Artrite Experimental/imunologia , Osteogênese/imunologia , Plasmócitos/imunologia , Animais , Artrite Experimental/patologia , Artrite Reumatoide/imunologia , Artrite Reumatoide/patologia , Doenças Autoimunes/imunologia , Doenças Autoimunes/patologia , Linfócitos B/imunologia , Linfócitos B/patologia , Células da Medula Óssea/imunologia , Células da Medula Óssea/patologia , Reabsorção Óssea/imunologia , Reabsorção Óssea/patologia , Feminino , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Transgênicos , Plasmócitos/patologia , Ligante RANK/deficiência , Ligante RANK/genética , Ligante RANK/imunologia , Membrana Sinovial/imunologia , Membrana Sinovial/patologia
2.
Nature ; 561(7722): 195-200, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30185903

RESUMO

Receptor activator of nuclear factor-kappa B (RANK) ligand (RANKL) binds RANK on the surface of osteoclast precursors to trigger osteoclastogenesis. Recent studies have indicated that osteocytic RANKL has an important role in osteoclastogenesis during bone remodelling; however, the role of osteoblastic RANKL remains unclear. Here we show that vesicular RANK, which is secreted from the maturing osteoclasts, binds osteoblastic RANKL and promotes bone formation by triggering RANKL reverse signalling, which activates Runt-related transcription factor 2 (Runx2). The proline-rich motif in the RANKL cytoplasmic tail is required for reverse signalling, and a RANKL(Pro29Ala) point mutation reduces activation of the reverse signalling pathway. The coupling of bone resorption and formation is disrupted in RANKL(Pro29Ala) mutant mice, indicating that osteoblastic RANKL functions as a coupling signal acceptor that recognizes vesicular RANK. RANKL reverse signalling is therefore a potential pharmacological target for avoiding the reduced bone formation associated with inhibition of osteoclastogenesis.


Assuntos
Reabsorção Óssea/metabolismo , Osteogênese , Ligante RANK/metabolismo , Transdução de Sinais , Substituição de Aminoácidos , Animais , Diferenciação Celular , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Reagentes de Ligações Cruzadas/química , Vesículas Citoplasmáticas/metabolismo , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteoclastos/citologia , Osteoclastos/metabolismo , Ligante RANK/química , Ligante RANK/deficiência , Ligante RANK/genética , Receptor Ativador de Fator Nuclear kappa-B/genética , Receptor Ativador de Fator Nuclear kappa-B/metabolismo
3.
Bone ; 114: 161-171, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29292230

RESUMO

The tooth root is essential for normal tooth physiological function. Studies on mice with mutations or targeted gene deletions revealed that osteoclasts (OCs) play an important role in tooth root development. However, knowledge on the cellular and molecular mechanism underlying how OCs mediate root formation is limited. During bone formation, growth factors (e.g. Insulin-like growth factor-1, IGF-1) liberated from bone matrix by osteoclastic bone resorption stimulate osteoblast differentiation. Thus, we hypothesize that OC-osteoblast coupling may also apply to OC-odontoblast coupling; therefore OCs may have a direct impact on odontoblast differentiation through the release of growth factor(s) from bone matrix, and consequently regulate tooth root formation. To test this hypothesis, we used a receptor activator of NF-κB ligand (RANKL) knockout mouse model in which OC differentiation and function was entirely blocked. We found that molar root formation and tooth eruption were defective in RANKL-/- mice. Disrupted elongation and disorganization of Hertwig's epithelial root sheath (HERS) was observed in RANKL-/- mice. Reduced expression of nuclear factor I C (NFIC), osterix, and dentin sialoprotein, markers essential for radicular (root) odontogenic cell differentiation indicated that odontoblast differentiation was disrupted in RANKL deficient mice likely contributing to the defect in root formation. Moreover, down-regulation of IGF/AKT/mTOR activity in odontoblast indicated that IGF signaling transduction in odontoblasts of the mutant mice was impaired. Treating odontoblast cells in vitro with conditioned medium from RANKL-/- OCs cultured on bone slices resulted in inhibition of odontoblast differentiation. Moreover, depletion of IGF-1 in bone resorption-conditioned medium (BRCM) from wild-type (WT) OC significantly compromised the ability of WT osteoclastic BRCM to induce odontoblast differentiation while addition of IGF-1 into RANKL-/- osteoclastic BRCM rescued impaired odontoblast differentiation, confirming that root and eruption defect in RANKL deficiency mice may result from failure of releasing of IGF-1 from bone matrix through OC bone resorption. These results suggest that OCs are important for odontoblast differentiation and tooth root formation, possibly through IGF/AKT/mTOR signaling mediated by cell-bone matrix interaction. These findings provide significant insights into regulatory mechanism of tooth root development, and also lay the foundation for root regeneration studies.


Assuntos
Reabsorção Óssea/metabolismo , Fator de Crescimento Insulin-Like I/deficiência , Mutação/fisiologia , Odontoblastos/metabolismo , Ligante RANK/deficiência , Raiz Dentária/metabolismo , Animais , Reabsorção Óssea/diagnóstico por imagem , Reabsorção Óssea/genética , Dentinogênese/efeitos dos fármacos , Dentinogênese/fisiologia , Fator de Crescimento Insulin-Like I/administração & dosagem , Fator de Crescimento Insulin-Like I/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Odontoblastos/efeitos dos fármacos , Ligante RANK/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Raiz Dentária/efeitos dos fármacos , Raiz Dentária/crescimento & desenvolvimento
4.
J Bone Miner Res ; 33(1): 167-181, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28914985

RESUMO

Currently, it is believed that osteoclasts positive for tartrate-resistant acid phosphatase (TRAP+) are the exclusive bone-resorbing cells responsible for focal bone destruction in inflammatory arthritis. Recently, a mouse model of cherubism (Sh3bp2KI/KI ) with a homozygous gain-of-function mutation in the SH3-domain binding protein 2 (SH3BP2) was shown to develop auto-inflammatory joint destruction. Here, we demonstrate that Sh3bp2KI/KI mice also deficient in the FBJ osteosarcoma oncogene (c-Fos) still exhibit noticeable bone erosion at the distal tibia even in the absence of osteoclasts at 12 weeks old. Levels of serum collagen I C-terminal telopeptide (ICTP), a marker of bone resorption generated by matrix metalloproteinases (MMPs), were elevated, whereas levels of serum cross-linked C-telopeptide (CTX), another resorption marker produced by cathepsin K, were not increased. Collagenolytic MMP levels were increased in the inflamed joints of the Sh3bp2KI/KI mice deficient in c-Fos. Resorption pits contained a large number of F4/80+ macrophages and genetic depletion of macrophages rescued these erosive changes. Importantly, administration of NSC405020, an MMP14 inhibitor targeted to the hemopexin (PEX) domain, suppressed bone erosion in c-Fos-deficient Sh3bp2KI/KI mice. After activation of the NF-κB pathway, macrophage colony-stimulating factor (M-CSF)-dependent macrophages from c-Fos-deficient Sh3bp2KI/KI mice expressed increased amounts of MMP14 compared with wild-type macrophages. Interestingly, receptor activator of NF-κB ligand (RANKL)-deficient Sh3bp2KI/KI mice failed to show notable bone erosion, whereas c-Fos deletion did restore bone erosion to the RANKL-deficient Sh3bp2KI/KI mice, suggesting that osteolytic transformation of macrophages requires both loss-of-function of c-Fos and gain-of-function of SH3BP2 in this model. These data provide the first genetic evidence that cells other than osteoclasts can cause focal bone destruction in inflammatory bone disease and suggest that MMP14 is a key mediator conferring pathological bone-resorbing capacity on c-Fos-deficient Sh3bp2KI/KI macrophages. In summary, the paradigm that osteoclasts are the exclusive cells executing inflammatory bone destruction may need to be reevaluated based on our findings with c-Fos-deficient cherubism mice lacking osteoclasts. © 2017 American Society for Bone and Mineral Research.


Assuntos
Osso e Ossos/patologia , Querubismo/patologia , Inflamação/patologia , Macrófagos/patologia , Metaloproteinase 14 da Matriz/metabolismo , Osteoclastos/metabolismo , Proteínas Proto-Oncogênicas c-fos/deficiência , Fosfatase Ácida Resistente a Tartarato/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Articulações/efeitos dos fármacos , Articulações/patologia , Fator Estimulador de Colônias de Macrófagos/farmacologia , Macrófagos/efeitos dos fármacos , Macrófagos/enzimologia , Metaloproteinase 2 da Matriz/metabolismo , Camundongos , NF-kappa B/metabolismo , Osteoclastos/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ligante RANK/deficiência , Ligante RANK/metabolismo , Transdução de Sinais/efeitos dos fármacos
5.
J Clin Periodontol ; 45(3): 285-292, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29220094

RESUMO

AIM: Periodontitis results from bacteria-induced inflammation. A key cytokine, RANKL, is produced by a number of cell types. The cellular source of RANKL critical for periodontitis has not been established. METHODS: We induced periodontal bone loss by oral inoculation of Porphyromonas gingivalis and Fusobacterium nucleatum in both normoglycaemic and streptozotocin-induced type 1 diabetic mice. Experimental transgenic mice had osteocyte-specific deletion of floxed receptor activator of nuclear factor kappa-B ligand (RANKL) mediated by DMP-1-driven Cre recombinase. Outcomes were assessed by micro-CT, histomorphometric analysis, immunofluorescent analysis of RANKL and tartrate-resistant acid phosphatase staining for osteoclasts and osteoclast activity. RESULTS: Oral infection stimulated RANKL expression in osteocytes of wild-type mice, which was increased by diabetes and blocked in transgenic mice. Infected wild-type mice had significant bone loss and increased osteoclast numbers and activity, which were further enhanced by diabetes. No bone loss or increase in osteoclastogenesis or activity was detected in transgenic mice with RANKL deletion in osteocytes that were normoglycaemic or diabetic. CONCLUSIONS: This study demonstrates for the first time the essential role of osteocytes in bacteria-induced periodontal bone loss and in diabetes-enhanced periodontitis.


Assuntos
Perda do Osso Alveolar/microbiologia , Infecções por Bacteroidaceae/complicações , Diabetes Mellitus Experimental/complicações , Proteínas da Matriz Extracelular/genética , Osteócitos/metabolismo , Periodontite/metabolismo , Porphyromonas gingivalis , Ligante RANK/metabolismo , Animais , Proteínas da Matriz Extracelular/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Periodontite/complicações , Periodontite/microbiologia , Ligante RANK/deficiência
6.
J Bone Miner Metab ; 36(3): 264-273, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-28589412

RESUMO

Src knockout (KO) and RANKL KO mice both exhibit near complete osteopetrosis in terms of 3D-bone volume (BV) fraction by micro-CT, whereas the serum CTX concentration of Src KO is apparently normal and that of RANKL KO is 30% of wild-type (WT) despite the fact that they lack osteoclasts. By histomorphometry we found that, whereas eroded surface (ES) and osteoid surface (OS) are zero values in RANKL KO, they are indistinguishable from WT in Src KO; because of marked increase in bone surface (BS), ES/BS and OS/BS of Src KO are 30-40% of WT. While RANKL KO lack both osteoclasts and osteoblasts, Src KO reveal increased numbers of osteoclasts and indistinguishable numbers of osteoblasts compared with WT; again, on the basis of BS, N.Oc/BS is comparable to WT and N.Ob/BS is markedly decreased in Src KO. The apparently increased number of total osteoclasts may be due to increased expression of RANKL found in Src KO bone in vivo. Src has a gene dosage-dependent effect on osteoclast function in vitro, with Src-/- osteoclasts completely lacking bone-resorbing function as determined by CTX release on dentin. Thus, Src KO osteoclasts retain some bone-resorbing function in vivo. The number of osteocytes is proportionally increased in RANKL KO, while Src KO mice have relative osteocyte deficiency, raising the possibility that RANKL and Src has an unrecognized role in osteocyte survival.


Assuntos
Colágeno Tipo I/sangue , Peptídeos/sangue , Ligante RANK/deficiência , Quinases da Família src/deficiência , Animais , Biomarcadores/metabolismo , Reabsorção Óssea/sangue , Reabsorção Óssea/genética , Reabsorção Óssea/metabolismo , Regulação da Expressão Gênica , Imageamento Tridimensional , Camundongos , Camundongos Knockout , Osteoblastos/metabolismo , Osteocalcina/sangue , Osteoclastos/metabolismo , Osteócitos/metabolismo , Fosfatase Ácida Resistente a Tartarato/sangue , Tíbia/diagnóstico por imagem , Tíbia/patologia , Microtomografia por Raio-X
7.
Stem Cells ; 35(5): 1365-1377, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28100034

RESUMO

Autosomal recessive osteopetrosis (ARO) is a severe bone disease characterized by increased bone density due to impairment in osteoclast resorptive function or differentiation. Hematopoietic stem cell transplantation is the only available treatment; however, this therapy is not effective in RANKL-dependent ARO, since in bone this gene is mainly expressed by cells of mesenchymal origin. Of note, whether lack of RANKL production might cause a defect also in the bone marrow (BM) stromal compartment, possibly contributing to the pathology, is unknown. To verify this possibility, we generated and characterized BM mesenchymal stromal cell (BM-MSC) lines from wild type and Rankl-/- mice, and found that Rankl-/- BM-MSCs displayed reduced clonogenicity and osteogenic capacity. The differentiation defect was significantly improved by lentiviral transduction of Rankl-/- BM-MSCs with a vector stably expressing human soluble RANKL (hsRANKL). Expression of Rankl receptor, Rank, on the cytoplasmic membrane of BM-MSCs pointed to the existence of an autocrine loop possibly activated by the secreted cytokine. Based on the close resemblance of RANKL-defective osteopetrosis in humans and mice, we expect that our results are also relevant for RANKL-dependent ARO patients. Data obtained in vitro after transduction with a lentiviral vector expressing hsRANKL would suggest that restoration of RANKL production might not only rescue the defective osteoclastogenesis of this ARO form, but also improve a less obvious defect in the osteoblast lineage, thus possibly achieving higher benefit for the patients, when the approach is translated to clinics. Stem Cells 2017;35:1365-1377.


Assuntos
Diferenciação Celular , Vetores Genéticos/metabolismo , Lentivirus/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Osteogênese , Ligante RANK/deficiência , Animais , Biomarcadores/metabolismo , Células Clonais , Imunofenotipagem , Camundongos Endogâmicos C57BL , Ligante RANK/metabolismo , Transdução de Sinais , Transdução Genética
8.
Immunity ; 43(6): 1174-85, 2015 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-26680207

RESUMO

The central nervous system (CNS) is an immunologically privileged site protected from uncontrolled access of T cells by the blood-brain barrier (BBB), which is breached upon autoimmune inflammation. Here we have shown that receptor activator of nuclear factor-κB (NF-κB) ligand (RANKL) on T cells regulates C-C type chemokine ligand 20 (CCL20) production by astrocytes and T cell localization in the CNS. Importantly, mice specifically lacking RANKL in T cells were resistant to experimental autoimmune encephalomyelitis (EAE) due to altered T cell trafficking. Pharmacological inhibition of RANKL prevented the development of EAE without affecting the peripheral immune response, indicating that RANKL is a potential therapeutic target for treating autoimmune diseases in the CNS.


Assuntos
Quimiotaxia de Leucócito/imunologia , Encefalomielite Autoimune Experimental/imunologia , Ligante RANK/imunologia , Linfócitos T/imunologia , Animais , Astrócitos/imunologia , Técnicas de Cocultura , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Imuno-Histoquímica , Ativação Linfocitária/imunologia , Camundongos , Camundongos Knockout , Ligante RANK/deficiência , Reação em Cadeia da Polimerase em Tempo Real
9.
J Immunol ; 194(9): 4144-53, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25825446

RESUMO

The immune and the skeletal system are tightly interconnected, and B lymphocytes are uniquely endowed with osteo-interactive properties. In this context, receptor activator of NF-κB (RANK) ligand (RANKL) plays a pivotal role in lymphoid tissue formation and bone homeostasis. Although murine models lacking RANK or RANKL show defects in B cell number, the role of the RANKL-RANK axis on B physiology is still a matter of debate. In this study, we have characterized in detail B cell compartment in Rankl(-/-) mice, finding a relative expansion of marginal zone B cells, B1 cells, and plasma cells associated with increased Ig serum levels, spontaneous germinal center formation, and hyperresponse to CD40 triggering. Such abnormalities were associated with an increased frequency of regulatory B cells and augmented B cell-derived IL-10 production. Remarkably, in vivo IL-10-R blockade reduced T cell-triggered plasma cell differentiation and restrained the expansion of regulatory B cells. These data point to a novel role of the RANKL-RANK axis in the regulation of B cell homeostasis and highlight an unexpected link between IL-10 CD40 signaling and the RANKL pathway.


Assuntos
Linfócitos B/imunologia , Interleucina-10/imunologia , Ligante RANK/deficiência , Ligante RANK/imunologia , Animais , Camundongos , Camundongos Knockout
10.
J Bone Miner Res ; 30(5): 855-68, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25431114

RESUMO

Receptor activator of NF-κB ligand (RANKL) is a TNFα-like cytokine that is produced by a diverse set of lineage-specific cells and is involved in a wide variety of physiological processes that include skeletal remodeling, lymph node organogenesis, mammary gland development, and thermal regulation. Consistent with these diverse functions, control of RANKL expression is accomplished in a cell-specific fashion via a set of at least 10 regulatory enhancers that are located up to 170 kb upstream of the gene's transcriptional start site. Here we examined the in vivo consequence of introducing a contiguous DNA segment containing these components into a genetically deleted RANKL null mouse strain. In contrast to RANKL null littermates, null mice containing the transgene exhibited normalized body size, skeletal development, and bone mass as well as normal bone marrow cavities, normalized spleen weights, and the presence of developed lymph nodes. These mice also manifested normalized reproductive capacity, including the ability to lactate and to produce normal healthy litters. Consistent with this, the transgene restored endogenous-like RANKL transcript levels in several RANKL-expressing tissues. Most importantly, restoration of RANKL expression from this segment of DNA was fully capable of rescuing the complex aberrant skeletal and immune phenotype of the RANKL null mouse. RANKL also restored appropriate levels of B220+ IgM+ and B220+ IgD+ B cells in spleen. Finally, we found that RANKL expression from this transgene was regulated by exogenously administered 1,25(OH)2 D3 , parathyroid hormone (PTH), and lipopolysaccharide (LPS), thus recapitulating the ability of these same factors to regulate the endogenous gene. These findings fully highlight the properties of the Tnfsf11 gene locus predicted through previous in vitro dissection. We conclude that the mouse Tnfsf11 gene locus identified originally through unbiased chromatin immunoprecipitation with DNA microarray (ChIP-chip) analysis contains the necessary genetic information to direct appropriate tissue-specific and factor-regulated RANKL expression in vivo.


Assuntos
DNA/genética , Ligante RANK/deficiência , Ligante RANK/genética , Sequências Reguladoras de Ácido Nucleico/genética , Transcrição Gênica , Animais , Linfócitos B/citologia , Linfócitos B/efeitos dos fármacos , Linfócitos B/metabolismo , Células da Medula Óssea/citologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Colecalciferol/farmacologia , Lâmina de Crescimento/efeitos dos fármacos , Lâmina de Crescimento/metabolismo , Humanos , Lipopolissacarídeos/farmacologia , Tecido Linfoide/metabolismo , Camundongos , Camundongos Transgênicos , Especificidade de Órgãos/efeitos dos fármacos , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Osteogênese/efeitos dos fármacos , Hormônio Paratireóideo/farmacologia , Fenótipo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Linfócitos T/citologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Transcrição Gênica/efeitos dos fármacos , Transgenes
11.
J Cell Biochem ; 113(1): 260-8, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21898548

RESUMO

The pathophysiology of osteoporosis in patients with Crohn's disease (CD) is still not completely elucidated. In this study, we evaluated osteoclastogenesis from peripheral blood cells of CD patients and studied the role of lymphocytes and inflammatory cytokines in this process. Peripheral blood mononuclear cells from seven patients with quiescent CD and matched healthy controls were isolated, and separated into T cells, B cells, and a T- and B-cell depleted fraction. In various culture combinations, osteoclast formation in the absence of the osteoclastogenic factors RANKL and M-CSF was assessed by scoring the number of tartrate-resistant acid phosphatase (TRACP) positive multinucleated cells (MNCs). Cytokine levels in culture supernatants were measured. Formation of heterogeneous cell clusters in culture was noticed; a process that was inhibited by anti-LFA-1. In CD cultures, mean cluster area was up to threefold higher than in control cultures, and shown to be induced by T cells. Over tenfold higher numbers of TRACP(+) MNCs were found in CD cultures, but exclusively in cultures containing T cells. Formation of cell clusters correlated strongly with formation of TRACP(+) MNCs. Both cell cluster formation and osteoclast formation were related to IL-17 levels in vitro. In conclusion, osteoclastogenesis, preceded by cell cluster formation, is T cell-mediated and increased in patients with quiescent CD. Our findings suggest heterotypic interactions between osteoclast precursors and T cells to be a triggering step in osteoclast formation in CD. Furthermore, our results propose a possible role for IL-17 in osteoclastogenesis in CD patients, and as such in CD-associated bone loss.


Assuntos
Reabsorção Óssea/patologia , Doença de Crohn/imunologia , Doença de Crohn/metabolismo , Osteoclastos/metabolismo , Linfócitos T/metabolismo , Adulto , Reabsorção Óssea/etiologia , Proliferação de Células , Células Cultivadas , Doença de Crohn/complicações , Citocinas/imunologia , Feminino , Humanos , Interleucina-17/imunologia , Fator Estimulador de Colônias de Macrófagos/deficiência , Masculino , Osteoclastos/citologia , Osteoclastos/imunologia , Osteoporose/fisiopatologia , Ligante RANK/deficiência
12.
Nat Med ; 17(10): 1231-4, 2011 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-21909105

RESUMO

Osteocytes embedded in bone have been postulated to orchestrate bone homeostasis by regulating both bone-forming osteoblasts and bone-resorbing osteoclasts. We find here that purified osteocytes express a much higher amount of receptor activator of nuclear factor-κB ligand (RANKL) and have a greater capacity to support osteoclastogenesis in vitro than osteoblasts and bone marrow stromal cells. Furthermore, the severe osteopetrotic phenotype that we observe in mice lacking RANKL specifically in osteocytes indicates that osteocytes are the major source of RANKL in bone remodeling in vivo.


Assuntos
Osso e Ossos/fisiologia , Homeostase/fisiologia , Osteócitos/metabolismo , Osteopetrose/metabolismo , Ligante RANK/metabolismo , Animais , Diferenciação Celular/fisiologia , Fêmur/citologia , Fêmur/diagnóstico por imagem , Citometria de Fluxo , Perfilação da Expressão Gênica , Proteínas de Fluorescência Verde/metabolismo , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Osteoclastos/metabolismo , Osteoclastos/fisiologia , Osteócitos/fisiologia , Ligante RANK/deficiência , Reação em Cadeia da Polimerase em Tempo Real , Microtomografia por Raio-X
13.
J Bone Miner Res ; 26(12): 2978-90, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21898588

RESUMO

Osteoclasts are derived from the monocyte/macrophage lineage, but little is known about osteoclast precursors in circulation. We previously showed that cell cycle-arrested quiescent osteoclast precursors (QOPs) were detected along bone surfaces as direct osteoclast precursors. Here we show that receptor activator of NF-κB (RANK)-positive cells isolated from bone marrow and peripheral blood possess characteristics of QOPs in mice. RANK-positive cells expressed c-Fms (receptors of macrophage colony-stimulating factor) at various levels, but scarcely expressed other monocyte/granulocyte markers. RANK-positive cells failed to exert phagocytic and proliferating activities, and differentiated into osteoclasts but not into dendritic cells. To identify circulating QOPs, collagen disks containing bone morphogenetic protein-2 (BMP disks) were implanted into mice, which were administered bromodeoxyuridine daily. Most nuclei of osteoclasts detected in BMP-2-induced ectopic bone were bromodeoxyuridine-negative. RANK-positive cells in peripheral blood proliferated more slowly and had a much longer lifespan than F4/80 (a macrophage marker)-positive macrophages. When BMP disks and control disks were implanted in RANK ligand-deficient mice, RANK-positive cells were observed in the BMP disks but not in the controls. F4/80-positive cells were distributed in both disks. Administration of FYT720, a sphingosine 1-phosphate agonist, promoted the egress of RANK-positive cells from hematopoietic tissues into bloodstream. These results suggest that lineage-determined QOPs circulate in the blood and settle in the bone.


Assuntos
Sangue/metabolismo , Osso e Ossos/citologia , Linhagem da Célula , Movimento Celular , Osteoclastos/citologia , Células-Tronco/citologia , Animais , Biomarcadores/metabolismo , Células da Medula Óssea/citologia , Proteínas Morfogenéticas Ósseas/metabolismo , Osso e Ossos/metabolismo , Bromodesoxiuridina/metabolismo , Núcleo Celular/metabolismo , Macrófagos/citologia , Macrófagos/metabolismo , Camundongos , Osteoclastos/metabolismo , Osteócitos/citologia , Osteócitos/metabolismo , Ligante RANK/deficiência , Ligante RANK/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo
14.
Am J Pathol ; 179(4): 1861-71, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21854748

RESUMO

Cryptopatches (CPs) and isolated lymphoid follicles (ILFs) are organized intestinal lymphoid tissues that develop postnatally in mice and include stromal cells expressing the receptor activator of nuclear factor kappa-B ligand (RANKL). We investigated how stromal RANKL influences the development and differentiation of CPs and ILFs by analyzing the development of these lymphoid structures in knockout mice lacking RANKL. We found that RANKL(-/-) mice had a fourfold reduction in the overall density of CPs in the small intestine compared to control mice, with the largest decrease in the proximal small intestine. No B cells were present in CPs from the small intestine of RANKL(-/-) mice and ILF formation was completely blocked. In sharp contrast, colonic ILFs containing B cells were present in RANKL(-/-) mice. Stromal cells within CPs in the small intestine of RANKL(-/-) mice did not express CXCL13 (originally called B lymphocyte chemoattractant) and often lacked other normally expressed stromal cell antigens, whereas colonic lymphoid aggregates in RANKL(-/-) mice retained stromal CXCL13 expression. The CXCL13-dependent maturation of precursor CPs into ILFs is differentially regulated in the small intestine and colon, with an absolute requirement for RANKL only in the small intestine.


Assuntos
Intestino Grosso/embriologia , Intestino Grosso/imunologia , Intestino Delgado/embriologia , Intestino Delgado/imunologia , Tecido Linfoide/embriologia , Organogênese , Ligante RANK/metabolismo , Animais , Antígenos/imunologia , Linfócitos B/patologia , Antígeno CD11c/metabolismo , Contagem de Células , Quimiocina CXCL13/metabolismo , Humanos , Intestino Grosso/patologia , Intestino Delgado/patologia , Tecido Linfoide/metabolismo , Tecido Linfoide/patologia , Receptor beta de Linfotoxina/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Tamanho do Órgão , Ligante RANK/deficiência , Células Estromais/imunologia , Células Estromais/patologia
15.
Nature ; 468(7320): 98-102, 2010 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-20881962

RESUMO

Breast cancer is one of the most common cancers in humans and will on average affect up to one in eight women in their lifetime in the United States and Europe. The Women's Health Initiative and the Million Women Study have shown that hormone replacement therapy is associated with an increased risk of incident and fatal breast cancer. In particular, synthetic progesterone derivatives (progestins) such as medroxyprogesterone acetate (MPA), used in millions of women for hormone replacement therapy and contraceptives, markedly increase the risk of developing breast cancer. Here we show that the in vivo administration of MPA triggers massive induction of the key osteoclast differentiation factor RANKL (receptor activator of NF-κB ligand) in mammary-gland epithelial cells. Genetic inactivation of the RANKL receptor RANK in mammary-gland epithelial cells prevents MPA-induced epithelial proliferation, impairs expansion of the CD49f(hi) stem-cell-enriched population, and sensitizes these cells to DNA-damage-induced cell death. Deletion of RANK from the mammary epithelium results in a markedly decreased incidence and delayed onset of MPA-driven mammary cancer. These data show that the RANKL/RANK system controls the incidence and onset of progestin-driven breast cancer.


Assuntos
Neoplasias Mamárias Experimentais/induzido quimicamente , Neoplasias Mamárias Experimentais/patologia , Progestinas/efeitos adversos , Ligante RANK/metabolismo , Animais , Apoptose/efeitos da radiação , Diferenciação Celular , Proliferação de Células/efeitos dos fármacos , Dano ao DNA , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/efeitos da radiação , Feminino , Raios gama , Integrina alfa6/metabolismo , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/metabolismo , Acetato de Medroxiprogesterona/administração & dosagem , Acetato de Medroxiprogesterona/efeitos adversos , Camundongos , NF-kappa B/metabolismo , Osteoclastos/citologia , Fosfoproteínas/análise , Fosfoproteínas/imunologia , Progestinas/administração & dosagem , Ligante RANK/deficiência , Ligante RANK/genética , Receptor Ativador de Fator Nuclear kappa-B/deficiência , Receptor Ativador de Fator Nuclear kappa-B/genética , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Transdução de Sinais , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo
16.
J Immunol ; 183(9): 5738-47, 2009 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-19828638

RESUMO

Microfold cells (M cells) are specialized epithelial cells situated over Peyer's patches (PP) and other organized mucosal lymphoid tissues that transport commensal bacteria and other particulate Ags into intraepithelial pockets accessed by APCs. The TNF superfamily member receptor activator of NF-kappaB ligand (RANKL) is selectively expressed by subepithelial stromal cells in PP domes. We found that RANKL null mice have <2% of wild-type levels of PP M cells and markedly diminished uptake of 200 nm diameter fluorescent beads. Ab-mediated neutralization of RANKL in adult wild-type mice also eliminated most PP M cells. The M cell deficit in RANKL null mice was corrected by systemic administration of exogenous RANKL. Treatment with RANKL also induced the differentiation of villous M cells on all small intestinal villi with the capacity for avid uptake of Salmonella and Yersinia organisms and fluorescent beads. The RANK receptor for RANKL is expressed by epithelial cells throughout the small intestine. We conclude that availability of RANKL is the critical factor controlling the differentiation of M cells from RANK-expressing intestinal epithelial precursor cells.


Assuntos
Antígenos/metabolismo , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Mucosa Intestinal/imunologia , Ligante RANK/fisiologia , Animais , Linhagem Celular , Feminino , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Intestino Delgado/citologia , Intestino Delgado/imunologia , Intestino Delgado/metabolismo , Intestino Delgado/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Microvilosidades/imunologia , Microvilosidades/metabolismo , Nódulos Linfáticos Agregados/citologia , Nódulos Linfáticos Agregados/imunologia , Nódulos Linfáticos Agregados/metabolismo , Lectinas de Plantas/biossíntese , Lectinas de Plantas/metabolismo , Ligante RANK/deficiência , Ligante RANK/genética , Receptor Ativador de Fator Nuclear kappa-B/fisiologia , Salmonella typhi/imunologia , Ulex/imunologia , Ulex/metabolismo , Yersinia enterocolitica/imunologia
17.
Hum Genet ; 124(6): 561-77, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18987890

RESUMO

Osteopetrosis is the result of mutations affecting osteoclast function. Careful analyses of osteopetrosis have provided instrumental information on bone remodeling, including the coupling of bone formation to bone resorption. Based on a range of novel genetic mutations and the resulting osteoclast phenotypes, we discuss how osteopetrosis models have clarified the function of the coupling of bone formation to bone resorption, and the pivotal role of the osteoclast and their function in this phenomenon. We highlight the distinct possibility that osteoclast activities can be divided into two separate avenues: bone resorption and control of bone formation.


Assuntos
Mutação , Osteoclastos/fisiologia , Osteopetrose/genética , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Proteínas Relacionadas à Autofagia , Remodelação Óssea/genética , Remodelação Óssea/fisiologia , Reabsorção Óssea/genética , Reabsorção Óssea/fisiopatologia , Anidrase Carbônica II/deficiência , Anidrase Carbônica II/genética , Catepsina K , Catepsinas/genética , Canais de Cloreto/genética , Modelos Animais de Doenças , Humanos , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Proteínas de Membrana/genética , Camundongos , Modelos Biológicos , Osteoblastos/patologia , Osteoblastos/fisiologia , Osteoclastos/patologia , Osteopetrose/etiologia , Osteopetrose/patologia , Osteopetrose/fisiopatologia , Ligante RANK/deficiência , Ligante RANK/genética , Receptor Ativador de Fator Nuclear kappa-B/deficiência , Receptor Ativador de Fator Nuclear kappa-B/genética , Ubiquitina-Proteína Ligases/genética , ATPases Vacuolares Próton-Translocadoras/genética
18.
Immunity ; 29(3): 423-37, 2008 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-18799149

RESUMO

Medullary thymic epithelial cells (mTECs) establish T cell self-tolerance through the expression of autoimmune regulator (Aire) and peripheral tissue-specific self-antigens. However, signals underlying mTEC development remain largely unclear. Here, we demonstrate crucial regulation of mTEC development by receptor activator of NF-kappaB (RANK) and CD40 signals. Whereas only RANK signaling was essential for mTEC development during embryogenesis, in postnatal mice, cooperation between CD40 and RANK signals was required for mTEC development to successfully establish the medullary microenvironment. Ligation of RANK or CD40 on fetal thymic stroma in vitro induced mTEC development in a tumor necrosis factor-associated factor 6 (TRAF6)-, NF-kappaB inducing kinase (NIK)-, and IkappaB kinase beta (IKKbeta)-dependent manner. These results show that developmental-stage-dependent cooperation between RANK and CD40 promotes mTEC development, thereby establishing self-tolerance.


Assuntos
Antígenos CD40/metabolismo , Ligante de CD40/metabolismo , Células Epiteliais/citologia , Células Epiteliais/imunologia , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Tolerância a Antígenos Próprios , Timo/citologia , Animais , Autoimunidade , Antígenos CD40/deficiência , Diferenciação Celular , Células Epiteliais/metabolismo , Camundongos , Camundongos Knockout , NF-kappa B/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Ligante RANK/deficiência , Ligante RANK/metabolismo , Transdução de Sinais , Fator 6 Associado a Receptor de TNF/metabolismo , Timo/embriologia , Timo/imunologia , Timo/fisiologia , Quinase Induzida por NF-kappaB
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...