Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Nature ; 589(7842): 442-447, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33361811

RESUMO

Successful pregnancies rely on adaptations within the mother1, including marked changes within the immune system2. It has long been known that the thymus, the central lymphoid organ, changes markedly during pregnancy3. However, the molecular basis and importance of this process remain largely obscure. Here we show that the osteoclast differentiation receptor RANK4,5 couples female sex hormones to the rewiring of the thymus during pregnancy. Genetic deletion of Rank (also known as Tnfrsf11a) in thymic epithelial cells results in impaired thymic involution and blunted expansion of natural regulatory T (Treg) cells in pregnant female mice. Sex hormones, in particular progesterone, drive the development of thymic Treg cells through RANK in a manner that depends on AIRE+ medullary thymic epithelial cells. The depletion of Rank in the mouse thymic epithelium results in reduced accumulation of natural Treg cells in the placenta, and an increase in the number of miscarriages. Thymic deletion of Rank also results in impaired accumulation of Treg cells in visceral adipose tissue, and is associated with enlarged adipocyte size, tissue inflammation, enhanced maternal glucose intolerance, fetal macrosomia, and a long-lasting transgenerational alteration in glucose homeostasis, which are all key hallmarks of gestational diabetes. Transplantation of Treg cells rescued fetal loss, maternal glucose intolerance and fetal macrosomia. In human pregnancies, we found that gestational diabetes also correlates with a reduced number of Treg cells in the placenta. Our findings show that RANK promotes the hormone-mediated development of thymic Treg cells during pregnancy, and expand the functional role of maternal Treg cells to the development of gestational diabetes and the transgenerational metabolic rewiring of glucose homeostasis.


Assuntos
Diabetes Gestacional/imunologia , Morte Fetal/etiologia , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Linfócitos T Reguladores/imunologia , Timo/imunologia , Adipócitos/patologia , Animais , Proliferação de Células , Diabetes Gestacional/etiologia , Diabetes Gestacional/metabolismo , Diabetes Gestacional/patologia , Células Epiteliais/imunologia , Feminino , Feto/imunologia , Feto/metabolismo , Feto/patologia , Glucose/metabolismo , Intolerância à Glucose/genética , Humanos , Gordura Intra-Abdominal/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Placenta/imunologia , Placenta/patologia , Gravidez , Receptor Ativador de Fator Nuclear kappa-B/deficiência , Receptor Ativador de Fator Nuclear kappa-B/genética , Linfócitos T Reguladores/citologia , Timo/citologia , Fatores de Transcrição/metabolismo , Proteína AIRE
2.
Kidney Blood Press Res ; 43(4): 1149-1159, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30036881

RESUMO

BACKGROUND/AIMS: Podocyte injury and loss contribute to proteinuria, glomerulosclerosis and eventually kidney failure. Receptor activator of NF-κB (RANK) belongs to the TNF receptor superfamily, which plays a key role in the pathogenesis of podocyte injury. However, the mechanism underlying the effect of RANK in podocyte injury remains unclear. Here, we sought to explore the possible molecular mechanisms involved in podocyte injury caused by RANK. METHODS: Immortalized mouse podocytes were treated with siRNA targeting RANK for 48 h or ionomycin for 24 h before harvest. Western blot, quantitative RT-PCR and immunofluorescence staining were used to evaluate the expression and function of RANK, nuclear factor of activated T cells c1 (NFATc1), transient receptor potential cation channel, subfamily C, member 6 (TRPC6) and calcineurin in podocytes. The Calcineurin Cellular Activity Assay kit was used to detect the phosphatase activity of calcineurin in cultured podocytes. A Ca2+ influx assay was performed to analyze alterations in Ca2+ entry under different conditions. Co-immunoprecipitation assays were used to observe the relationship between RANK and TRPC6. RESULTS: RANK mRNA and protein expression were markedly increased in injured podocytes (ionomycin stimulation). Further study found that translocation of NFATc1 to the nucleus was significantly reduced after knocking down RANK by siRNA. Meanwhile, we also demonstrated that loss of RANK suppressed the phosphatase activity of calcineurin and attenuated the ionomycin-induced increase in Ca2+ influx. In addition, we showed that RANK knockdown in cultured podocytes decreased TRPC6 protein expression. Co-immunoprecipitation experiments suggested that RANK binds to TRPC6 and that ionomycin enhanced the binding of RANK to TRPC6. CONCLUSION: Our findings demonstrated that RANK deficiency ameliorates podocyte injury by suppressing calcium/calcineurin/NFATc1 signaling, which may present a promising target for therapeutic intervention.


Assuntos
Podócitos/patologia , Receptor Ativador de Fator Nuclear kappa-B/farmacologia , Transdução de Sinais/efeitos dos fármacos , Ferimentos e Lesões/metabolismo , Animais , Calcineurina/metabolismo , Cálcio/metabolismo , Linhagem Celular , Camundongos , Fatores de Transcrição NFATC/metabolismo , Podócitos/química , RNA Interferente Pequeno/farmacologia , Receptor Ativador de Fator Nuclear kappa-B/análise , Receptor Ativador de Fator Nuclear kappa-B/deficiência , Receptor Ativador de Fator Nuclear kappa-B/genética
3.
Acta Neuropathol Commun ; 6(1): 31, 2018 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-29699580

RESUMO

Although there is a strong association between osteoporosis and skeletal muscle atrophy/dysfunction, the functional relevance of a particular biological pathway that regulates synchronously bone and skeletal muscle physiopathology is still elusive. Receptor-activator of nuclear factor κB (RANK), its ligand RANKL and the soluble decoy receptor osteoprotegerin (OPG) are the key regulators of osteoclast differentiation and bone remodelling. We thus hypothesized that RANK/RANKL/OPG, which is a key pathway for bone regulation, is involved in Duchenne muscular dystrophy (DMD) physiopathology. Our results show that muscle-specific RANK deletion (mdx-RANK mko ) in dystrophin deficient mdx mice improves significantly specific force [54% gain in force] of EDL muscles with no protective effect against eccentric contraction-induced muscle dysfunction. In contrast, full-length OPG-Fc injections restore the force of dystrophic EDL muscles [162% gain in force], protect against eccentric contraction-induced muscle dysfunction ex vivo and significantly improve functional performance on downhill treadmill and post-exercise physical activity. Since OPG serves a soluble receptor for RANKL and as a decoy receptor for TRAIL, mdx mice were injected with anti-RANKL and anti-TRAIL antibodies to decipher the dual function of OPG. Injections of anti-RANKL and/or anti-TRAIL increase significantly the force of dystrophic EDL muscle [45% and 17% gains in force, respectively]. In agreement, truncated OPG-Fc that contains only RANKL domains produces similar gains, in terms of force production, than anti-RANKL treatments. To corroborate that full-length OPG-Fc also acts independently of RANK/RANKL pathway, dystrophin/RANK double-deficient mice were treated with full-length OPG-Fc for 10 days. Dystrophic EDL muscles exhibited a significant gain in force relative to untreated dystrophin/RANK double-deficient mice, indicating that the effect of full-length OPG-Fc is in part independent of the RANKL/RANK interaction. The sarco/endoplasmic reticulum Ca2+ ATPase (SERCA) activity is significantly depressed in dysfunctional and dystrophic muscles and full-length OPG-Fc treatment increased SERCA activity and SERCA-2a expression. These findings demonstrate the superiority of full-length OPG-Fc treatment relative to truncated OPG-Fc, anti-RANKL, anti-TRAIL or muscle RANK deletion in improving dystrophic muscle function, integrity and protection against eccentric contractions. In conclusion, full-length OPG-Fc represents an efficient alternative in the development of new treatments for muscular dystrophy in which a single therapeutic approach may be foreseeable to maintain both bone and skeletal muscle functions.


Assuntos
Músculo Esquelético/metabolismo , Distrofias Musculares/metabolismo , Distrofias Musculares/patologia , Distrofias Musculares/terapia , Osteoprotegerina/uso terapêutico , Receptor Ativador de Fator Nuclear kappa-B/deficiência , Animais , Creatina Quinase/metabolismo , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Camundongos Transgênicos , Contração Muscular/efeitos dos fármacos , Contração Muscular/fisiologia , Distrofias Musculares/genética , Osteoprotegerina/química , Osteoprotegerina/metabolismo , RNA Mensageiro/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
4.
Exp Hematol ; 43(2): 110-4.e1-2, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25461255

RESUMO

The contribution of osteoclasts to hematopoietic stem/progenitor cell (HSPC) retention in the bone marrow is controversial. Studies of HSPC trafficking in osteoclast-deficient mice are limited by osteopetrosis. Here, we employed two non-osteopetrotic mouse models to assess the contribution of osteoclasts to basal and granulocyte colony-stimulating factor (G-CSF)-induced HSPC mobilization. We generated Rank(-/-) fetal liver chimeras using Csf3r(-/-) recipients to produce mice lacking G-CSF receptor expression in osteoclasts. Basal and G-CSF-induced HSPC mobilization was normal in these chimeras. We next acutely depleted osteoclasts in wild-type mice using the RANK ligand inhibitor osteoprotegerin. Marked suppression of osteoclasts was observed after a single injection of osteoprotegerin-Fc. Basal and G-CSF-induced HSPC mobilization in osteoprotegerin-Fc-treated mice was comparable to that in control mice. Together, these data indicate that osteoclasts are not required for the efficient retention of HSPCs in the bone marrow and are dispensable for HSPC mobilization by G-CSF.


Assuntos
Células da Medula Óssea/efeitos dos fármacos , Fator Estimulador de Colônias de Granulócitos/farmacologia , Mobilização de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/efeitos dos fármacos , Osteoclastos/efeitos dos fármacos , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Quimera/genética , Feto , Expressão Gênica , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Fragmentos Fc das Imunoglobulinas/administração & dosagem , Fragmentos Fc das Imunoglobulinas/genética , Fígado/citologia , Fígado/metabolismo , Masculino , Camundongos , Camundongos Knockout , Osteoclastos/citologia , Osteoclastos/metabolismo , Osteoprotegerina/administração & dosagem , Osteoprotegerina/genética , Ligante RANK/antagonistas & inibidores , Ligante RANK/genética , Ligante RANK/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/deficiência , Receptor Ativador de Fator Nuclear kappa-B/genética , Receptores de Fator Estimulador de Colônias/deficiência , Receptores de Fator Estimulador de Colônias/genética , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/genética
5.
J Bone Miner Res ; 27(12): 2501-10, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22836362

RESUMO

In the last decades the molecular basis of monogenic diseases has been largely unraveled, although their treatment has often remained unsatisfactory. Autosomal recessive osteopetrosis (ARO) belongs to the small group of genetic diseases that are usually treated with hematopoietic stem cell transplantation (HSCT). However, this approach is not effective in the recently identified form carrying mutations in the receptor activator of NF-κB ligand (RANKL) gene. In this subset, therapy replacement approach based on RANKL delivery has a strong rationale. Here we demonstrate that the systematic administration of RANKL for 1 month to Rankl(-/-) mice, which closely resemble the human disease, significantly improves the bone phenotype and has beneficial effects on bone marrow, spleen and thymus; major adverse effects arise only when mice are clearly overtreated. Overall, we provide evidence that the pharmacological administration of RANKL represents the appropriate treatment option for RANKL-deficient ARO patients, to be validated in a pilot clinical trial.


Assuntos
Osteopetrose/tratamento farmacológico , Osteopetrose/genética , Ligante RANK/uso terapêutico , Animais , Células da Medula Óssea/efeitos dos fármacos , Reabsorção Óssea/induzido quimicamente , Osso e Ossos/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Osteopetrose/patologia , Fenótipo , Ligante RANK/administração & dosagem , Ligante RANK/efeitos adversos , Ligante RANK/genética , Receptor Ativador de Fator Nuclear kappa-B/deficiência , Receptor Ativador de Fator Nuclear kappa-B/genética
6.
J Immunol ; 188(3): 1201-5, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22219325

RESUMO

RANKL-RANK signaling regulates numerous physiologic processes such as bone remodeling, lymph node organogenesis, central thermoregulation, and formation of a lactating mammary gland in pregnancy. Recently, a receptor activator of NF-κB ligand (RANKL)-blocking Ab has been approved for human use in potentially millions of osteoporosis and cancer patients. However, germline deficiencies in RANKL or receptor activator of NF-κB (RANK) also lead to strong B cell defects in mice and human patients, suggesting that RANKL-RANK inhibition could interfere with B cell physiology and thereby trigger immunologic side-effects. To address this key question--that is, whether RANKL-RANK signaling affects B cell physiology directly or the observed defects are secondary because of the severe osteopetrosis--we generated B cell-specific RANK knockout mice. We show that B cells deficient for RANK undergo normal development and do not show any obvious defects in Ab secretion, class switch recombination, or somatic hypermutation. Our data indicate that ablation of the RANKL-RANK pathway has no direct adverse effect on B cell physiology.


Assuntos
Linfócitos B/fisiologia , Receptor Ativador de Fator Nuclear kappa-B/imunologia , Animais , Formação de Anticorpos , Linfócitos B/metabolismo , Humanos , Switching de Imunoglobulina , Camundongos , Camundongos Knockout , Ligante RANK , Receptor Ativador de Fator Nuclear kappa-B/deficiência , Transdução de Sinais , Hipermutação Somática de Imunoglobulina
7.
Nature ; 470(7335): 548-53, 2011 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-21326202

RESUMO

Inflammatory mechanisms influence tumorigenesis and metastatic progression even in cancers whose aetiology does not involve pre-existing inflammation or infection, such as breast and prostate cancers. For instance, prostate cancer metastasis is associated with the infiltration of lymphocytes into advanced tumours and the upregulation of two tumour-necrosis-factor family members: receptor activator of nuclear factor-κB (RANK) ligand (RANKL) and lymphotoxin. But the source of RANKL and its role in metastasis have not been established. RANKL and its receptor RANK control the proliferation of mammary lobuloalveolar cells during pregnancy through inhibitor of nuclear factor-κB (IκB) kinase-α (IKK-α), a protein kinase that is needed for the self-renewal of mammary cancer progenitors and for prostate cancer metastasis. We therefore examined whether RANKL, RANK and IKK-α are also involved in mammary/breast cancer metastasis. Indeed, RANK signalling in mammary carcinoma cells that overexpress the proto-oncogene Erbb2 (also known as Neu), which is frequently amplified in metastatic human breast cancers, was important for pulmonary metastasis. Metastatic spread of Erbb2-transformed carcinoma cells also required CD4(+)CD25(+) T cells, whose major pro-metastatic function was RANKL production. Most RANKL-producing T cells expressed forkhead box P3 (FOXP3), a transcription factor produced by regulatory T cells, and were located next to smooth muscle actin (SMA)(+) stromal cells in mouse and human breast cancers. The dependence of pulmonary metastasis on T cells was replaceable by exogenous RANKL, which also stimulated pulmonary metastasis of RANK(+) human breast cancer cells. These results are consistent with the adverse impact of tumour-infiltrating CD4(+) or FOXP3(+) T cells on human breast cancer prognosis and suggest that the targeting of RANKL-RANK can be used in conjunction with the therapeutic elimination of primary breast tumours to prevent recurrent metastatic disease.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linfócitos do Interstício Tumoral/metabolismo , Ligante RANK/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Transdução de Sinais , Linfócitos T Reguladores/metabolismo , Animais , Antígenos CD4/genética , Antígenos CD4/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Antígenos CD8/genética , Antígenos CD8/metabolismo , Linhagem Celular Tumoral , Feminino , Fatores de Transcrição Forkhead/metabolismo , Genes RAG-1/genética , Humanos , Quinase I-kappa B/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Endogâmicos C57BL , Metástase Neoplásica/patologia , Proto-Oncogene Mas , Ligante RANK/antagonistas & inibidores , Ligante RANK/farmacologia , Receptor Ativador de Fator Nuclear kappa-B/deficiência , Receptor Ativador de Fator Nuclear kappa-B/genética , Receptor ErbB-2/genética , Receptor ErbB-2/metabolismo , Transdução de Sinais/efeitos dos fármacos
8.
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
9.
J Biol Chem ; 285(36): 28164-73, 2010 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-20558734

RESUMO

PTH stimulates osteoblastic cells to form new bone and to produce osteoblast-osteoclast coupling factors such as RANKL. Whether osteoclasts or their activity are needed for PTH anabolism remains uncertain. We treated ovariectomized huRANKL knock-in mice with a human RANKL inhibitor denosumab (DMAb), alendronate (Aln), or vehicle for 4 weeks, followed by co-treatment with intermittent PTH for 4 weeks. Loss of bone mass and microarchitecture was prevented by Aln and further significantly improved by DMAb. PTH improved bone mass, microstructure, and strength, and was additive to Aln but not to DMAb. Aln inhibited biochemical and histomorphometrical indices of bone turnover,--i.e. osteocalcin and bone formation rate (BFR) on cancellous bone surfaces-, and Dmab inhibited them further. However Aln increased whereas Dmab suppressed osteoclast number and surfaces. PTH significantly increased osteocalcin and bone formation indices, in the absence or presence of either antiresorptive, although BFR remained lower in presence of Dmab. To further evaluate PTH effects in the complete absence of osteoclasts, high dose PTH was administered to RANK(-/-) mice. PTH increased osteocalcin similarly in RANK(-/-) and WT mice. It also increased BMD in RANK(-/-) mice, although less than in WT. These results further indicate that osteoclasts are not strictly required for PTH anabolism, which presumably still occurs via stimulation of modeling-based bone formation. However the magnitude of PTH anabolic effects on the skeleton, in particular its additive effects with antiresorptives, depends on the extent of the remodeling space, as determined by the number and activity of osteoclasts on bone surfaces.


Assuntos
Alendronato/farmacologia , Anticorpos Monoclonais/farmacologia , Osso e Ossos/metabolismo , Técnicas de Introdução de Genes , Osteoclastos/efeitos dos fármacos , Hormônio Paratireóideo/farmacologia , Ligante RANK/farmacologia , Receptor Ativador de Fator Nuclear kappa-B/genética , Alendronato/administração & dosagem , Animais , Anticorpos Monoclonais/administração & dosagem , Anticorpos Monoclonais Humanizados , Biomarcadores/metabolismo , Densidade Óssea/efeitos dos fármacos , Reabsorção Óssea/tratamento farmacológico , Osso e Ossos/citologia , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/fisiologia , Denosumab , Relação Dose-Resposta a Droga , Feminino , Expressão Gênica , Humanos , Masculino , Camundongos , Osteoclastos/metabolismo , Osteogênese/efeitos dos fármacos , Ovariectomia , Hormônio Paratireóideo/administração & dosagem , Ligante RANK/administração & dosagem , Receptor Ativador de Fator Nuclear kappa-B/antagonistas & inibidores , Receptor Ativador de Fator Nuclear kappa-B/deficiência , Receptor Ativador de Fator Nuclear kappa-B/metabolismo
10.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA