Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Bone ; 143: 115708, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33164872

RESUMEN

The cysteine knot protein sclerostin is an osteocyte-derived secreted inhibitor of the Wnt co-receptors LRP5 and LRP6. LRP5 plays a dominant role in bone homeostasis, but we previously reported that Sost/sclerostin suppression significantly increased osteogenesis regardless of Lrp5 presence or absence. Those observations suggested that the bone forming effects of sclerostin inhibition can occur through Lrp6 (when Lrp5 is suppressed), or through other yet undiscovered mechanisms independent of Lrp5/6. To distinguish between these two possibilities, we generated mice with compound deletion of Lrp5 and Lrp6 selectively in bone, and treated them with sclerostin monoclonal antibody (Scl-mAb). All mice were homozygous flox for both Lrp5 and Lrp6 (Lrp5f/f; Lrp6f/f), and varied only in whether or not they carried the Dmp1-Cre transgene. Positive (Cre+) and negative (Cre-) mice were injected with Scl-mAb or vehicle from 4.5 to 14 weeks of age. Vehicle-treated Cre+ mice exhibited significantly reduced skeletal properties compared to vehicle-treated Cre- mice, as assessed by DXA, µCT, pQCT, and histology, indicating that Lrp5/6 deletions were effective and efficient. Scl-mAb treatment improved nearly every bone-related parameter among Cre- mice, but the same treatment in Cre+ mice resulted in little to no improvement in skeletal properties. For the few endpoints where Cre+ mice responded to Scl-mAb, it is likely that antibody-induced promotion of Wnt signaling occurred in cell types earlier in the mesenchymal/osteoblast differentiation pathway than the Dmp1-expressing stage. This latter conclusion was supported by changes in some histomorphometric parameters. In conclusion, unlike with the deletion of Lrp5 alone, the bone-selective late-stage co-deletion of Lrp5 and Lrp6 significantly impairs or completely nullifies the osteogenic action of Scl-mAb, and highlights a major role for both Lrp5 and Lrp6 in the mechanism of action for the bone-building effects of sclerostin antibody.


Asunto(s)
Glicoproteínas , Péptidos y Proteínas de Señalización Intercelular , Proteínas Adaptadoras Transductoras de Señales , Animales , Huesos/metabolismo , Glicoproteínas/metabolismo , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/genética , Proteína-6 Relacionada a Receptor de Lipoproteína de Baja Densidad/metabolismo , Ratones , Osteoblastos/metabolismo
2.
J Cell Physiol ; 235(12): 9785-9794, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32529635

RESUMEN

Skeletal homeostasis is sensitive to perturbations in Wnt signaling. Beyond its role in the bone, Wnt is a major target for pharmaceutical inhibition in a wide range of diseases, most notably cancers. Numerous clinical trials for Wnt-based candidates are currently underway, and Wnt inhibitors will likely soon be approved for clinical use. Given the bone-suppressive effects accompanying Wnt inhibition, there is a need to expose alternate pathways/molecules that can be targeted to counter the deleterious effects of Wnt inhibition on bone properties. Activation of the Pi3k/Akt pathway via Pten deletion is one possible osteoanabolic pathway to exploit. We investigated whether the osteopenic effects of ß-catenin deletion from bone cells could be rescued by Pten deletion in the same cells. Mice carrying floxed alleles for Pten and ß-catenin were bred to Dmp1-Cre mice to delete Pten alone, ß-catenin alone, or both genes from the late-stage osteoblast/osteocyte population. The mice were assessed for bone mass, density, strength, and formation parameters to evaluate the potential rescue effect of Pten deletion in Wnt-impaired mice. Pten deletion resulted in high bone mass and ß-catenin deletion resulted in low bone mass. Compound mutants had bone properties similar to ß-catenin mutant mice, or surprisingly in some assays, were further compromised beyond ß-catenin mutants. Pten inhibition, or one of its downstream nodes, is unlikely to protect against the bone-wasting effects of Wnt/ßcat inhibition. Other avenues for preserving bone mass in the presence of Wnt inhibition should be explored to alleviate the skeletal side effects of Wnt inhibitor-based therapies.


Asunto(s)
Proteínas de la Matriz Extracelular/genética , Neoplasias/tratamiento farmacológico , Fosfohidrolasa PTEN/genética , beta Catenina/genética , Animales , Enfermedades Óseas Metabólicas/tratamiento farmacológico , Enfermedades Óseas Metabólicas/genética , Proliferación Celular/genética , Modelos Animales de Enfermedad , Humanos , Ratones , Neoplasias/genética , Osteoblastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Osteogénesis/genética , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Vía de Señalización Wnt/efectos de los fármacos
4.
PLoS Genet ; 16(5): e1008361, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32463812

RESUMEN

Osteocalcin (OCN), the most abundant noncollagenous protein in the bone matrix, is reported to be a bone-derived endocrine hormone with wide-ranging effects on many aspects of physiology, including glucose metabolism and male fertility. Many of these observations were made using an OCN-deficient mouse allele (Osc-) in which the 2 OCN-encoding genes in mice, Bglap and Bglap2, were deleted in ES cells by homologous recombination. Here we describe mice with a new Bglap and Bglap2 double-knockout (dko) allele (Bglap/2p.Pro25fs17Ter) that was generated by CRISPR/Cas9-mediated gene editing. Mice homozygous for this new allele do not express full-length Bglap or Bglap2 mRNA and have no immunodetectable OCN in their serum. FTIR imaging of cortical bone in these homozygous knockout animals finds alterations in the collagen maturity and carbonate to phosphate ratio in the cortical bone, compared with wild-type littermates. However, µCT and 3-point bending tests do not find differences from wild-type littermates with respect to bone mass and strength. In contrast to the previously reported OCN-deficient mice with the Osc-allele, serum glucose levels and male fertility in the OCN-deficient mice with the Bglap/2pPro25fs17Ter allele did not have significant differences from wild-type littermates. We cannot explain the absence of endocrine effects in mice with this new knockout allele. Possible explanations include the effects of each mutated allele on the transcription of neighboring genes, or differences in genetic background and environment. So that our findings can be confirmed and extended by other interested investigators, we are donating this new Bglap and Bglap2 double-knockout strain to the Jackson Laboratories for academic distribution.


Asunto(s)
Sistema Endocrino/fisiología , Osteocalcina/genética , Animales , Densidad Ósea/genética , Huesos/metabolismo , Sistema Endocrino/metabolismo , Femenino , Fertilidad/genética , Resistencia a la Insulina/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteocalcina/deficiencia
5.
Bone ; 131: 115084, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31648079

RESUMEN

Enhancing LRP5 signaling and inhibiting TGFß signaling have each been reported to increase bone mass and improve bone strength in wild-type mice. Monotherapy targeting LRP5 signaling, or TGFß signaling, also improved bone properties in mouse models of Osteogenesis Imperfecta (OI). We investigated whether additive or synergistic increases in bone properties would be attained if enhanced LRP5 signaling was combined with TGFß inhibition. We crossed an Lrp5 high bone mass (HBM) allele (Lrp5A214V) into the Col1a2G610C/+ mouse model of OI. At 6-weeks-of-age we began treating mice with an antibody that inhibits TGFß1, ß2, and ß3 (mAb 1D11), or with an isotype-matched control antibody (mAb 13C4). At 12-weeks-old, we observed that combining enhanced LRP5 signaling with inhibited TGFß signaling produced an additive effect on femoral and vertebral trabecular bone volumes, but not on cortical bone volumes. Although enhanced LRP5 signaling increased femur strength in a 3-point bending assay in Col1a2G610C/+ mice, femur strength did not improve further with TGFß inhibition. Neither enhanced LRP5 signaling nor TGFß inhibition, alone or in combination, improved femur 3-point-bending post-yield displacement in Col1a2G610C/+ mice. These pre-clinical studies indicate combination therapies that target LRP5 and TGFß signaling should increase trabecular bone mass in patients with OI more than targeting either signaling pathway alone. Whether additive increases in trabecular bone mass will occur in, and clinically benefit, patients with OI needs to be determined.


Asunto(s)
Osteogénesis Imperfecta , Animales , Hueso Esponjoso/diagnóstico por imagen , Colágeno Tipo I , Hueso Cortical , Modelos Animales de Enfermedad , Humanos , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Ratones , Osteogénesis Imperfecta/tratamiento farmacológico , Osteogénesis Imperfecta/genética , Transducción de Señal , Factor de Crecimiento Transformador beta
6.
Bone ; 120: 166-175, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30409757

RESUMEN

High-bone-mass (HBM)-causing missense mutations in the low density lipoprotein receptor-related protein-5 (Lrp5) are associated with increased osteoanabolic action and protection from disuse- and ovariectomy-induced osteopenia. These mutations (e.g., A214V and G171V) confer resistance to endogenous secreted Lrp5/6 inhibitors, such as sclerostin (SOST) and Dickkopf homolog-1 (DKK1). Cells in the osteoblast lineage are responsive to canonical Wnt stimulation, but recent work has indicated that osteoclasts exhibit both indirect and direct responsiveness to canonical Wnt. Whether Lrp5-HBM receptors, expressed in osteoclasts, might alter osteoclast differentiation, activity, and consequent net bone balance in the skeleton, is not known. To address this, we bred mice harboring heterozygous Lrp5 HBM-causing conditional knock-in alleles to Ctsk-Cre transgenic mice and studied the phenotype using DXA, µCT, histomorphometry, serum assays, and primary cell culture. Mice with HBM alleles induced in Ctsk-expressing cells (TG) exhibited higher bone mass and architectural properties compared to non-transgenic (NTG) counterparts. In vivo and in vitro measurements of osteoclast activity, population density, and differentiation yielded significant reductions in osteoclast-related parameters in female but not male TG mice. Droplet digital PCR performed on osteocyte enriched cortical bone tubes from TG and NTG mice revealed that ~8-17% of the osteocyte population (depending on sex) underwent recombination of the conditional Lrp5 allele in the presence of Ctsk-Cre. Further, bone formation parameters in the midshaft femur cortex show a small but significant increase in anabolic action on the endocortical but not periosteal surface. These findings suggest that Wnt/Lrp5 signaling in osteoclasts affects osteoclastogenesis and activity in female mice, but also that some of the changes in bone mass in TG mice might be due to Cre expression in the osteocyte population.


Asunto(s)
Huesos/metabolismo , Catepsina K/metabolismo , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Mutación/genética , Absorciometría de Fotón , Alelos , Animales , Biomarcadores/sangre , Células de la Médula Ósea/metabolismo , Resorción Ósea/sangre , Resorción Ósea/patología , Huesos/diagnóstico por imagen , Diferenciación Celular , Femenino , Integrasas/metabolismo , Masculino , Ratones Transgénicos , Tamaño de los Órganos/genética , Osteoclastos/metabolismo , Osteoclastos/patología , Osteogénesis/genética , Periostio/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Recombinación Genética/genética , Transgenes , Microtomografía por Rayos X
7.
JCI Insight ; 3(11)2018 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-29875318

RESUMEN

The WNT pathway has become an attractive target for skeletal therapies. High-bone-mass phenotypes in patients with loss-of-function mutations in the LRP5/6 inhibitor Sost (sclerosteosis), or in its downstream enhancer region (van Buchem disease), highlight the utility of targeting Sost/sclerostin to improve bone properties. Sclerostin-neutralizing antibody is highly osteoanabolic in animal models and in human clinical trials, but antibody-based inhibition of another potent LRP5/6 antagonist, Dkk1, is largely inefficacious for building bone in the unperturbed adult skeleton. Here, we show that conditional deletion of Dkk1 from bone also has negligible effects on bone mass. Dkk1 inhibition increases Sost expression, suggesting a potential compensatory mechanism that might explain why Dkk1 suppression lacks anabolic action. To test this concept, we deleted Sost from osteocytes in, or administered sclerostin neutralizing antibody to, mice with a Dkk1-deficient skeleton. A robust anabolic response to Dkk1 deletion was manifest only when Sost/sclerostin was impaired. Whole-body DXA scans, µCT measurements of the femur and spine, histomorphometric measures of femoral bone formation rates, and biomechanical properties of whole bones confirmed the anabolic potential of Dkk1 inhibition in the absence of sclerostin. Further, combined administration of sclerostin and Dkk1 antibody in WT mice produced a synergistic effect on bone gain that greatly exceeded individual or additive effects of the therapies, confirming the therapeutic potential of inhibiting multiple WNT antagonists for skeletal health. In conclusion, the osteoanabolic effects of Dkk1 inhibition can be realized if sclerostin upregulation is prevented. Anabolic therapies for patients with low bone mass might benefit from a strategy that accounts for the compensatory milieu of WNT inhibitors in bone tissue.


Asunto(s)
Anabolizantes/administración & dosificación , Glicoproteínas/antagonistas & inhibidores , Hiperostosis/tratamiento farmacológico , Osteogénesis/efectos de los fármacos , Sindactilia/tratamiento farmacológico , Vía de Señalización Wnt/efectos de los fármacos , Proteínas Adaptadoras Transductoras de Señales , Animales , Anticuerpos Neutralizantes/administración & dosificación , Proteínas Morfogenéticas Óseas/genética , Modelos Animales de Enfermedad , Femenino , Fémur/citología , Fémur/diagnóstico por imagen , Fémur/patología , Marcadores Genéticos/genética , Glicoproteínas/genética , Glicoproteínas/metabolismo , Humanos , Hiperostosis/diagnóstico por imagen , Hiperostosis/genética , Hiperostosis/patología , Péptidos y Proteínas de Señalización Intercelular/genética , Mutación con Pérdida de Función , Masculino , Ratones , Osteocitos , Columna Vertebral/citología , Columna Vertebral/diagnóstico por imagen , Columna Vertebral/patología , Sindactilia/diagnóstico por imagen , Sindactilia/genética , Sindactilia/patología , Resultado del Tratamiento , Regulación hacia Arriba/efectos de los fármacos , Microtomografía por Rayos X
8.
Calcif Tissue Int ; 100(4): 361-373, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28013361

RESUMEN

Recently, we demonstrated that osteoblast-specific overexpression of human WNT16 increased both cortical and trabecular bone mass and structure in mice. To further identify the cell-specific role of Wnt16 in bone homeostasis, we created transgenic (TG) mice overexpressing human WNT16 in osteocytes using Dmp1 promoter (Dmp1-hWNT16 TG) on C57BL/6 (B6) background. We analyzed bone phenotypes and serum bone biomarkers, performed gene expression analysis and measured dynamic bone histomorphometry in Dmp1-hWNT16 TG and wild-type (WT) mice. Compared to WT mice, Dmp1-hWNT16 TG mice exhibited significantly higher whole-body, spine and femoral aBMD, BMC and trabecular (BV/TV, Tb.N, and Tb.Th) and cortical (bone area and thickness) parameters in both male and female at 12 weeks of age. Femur stiffness and ultimate force were also significantly improved in the Dmp1-hWNT16 TG female mice, compared to sex-matched WT littermates. In addition, female Dmp1-hWNT16 TG mice displayed significantly higher MS/BS, MAR and BFR/BS compared to the WT mice. Gene expression analysis demonstrated significantly higher mRNA level of Alp in both male and female Dmp1-hWNT16 TG mice and significantly higher levels of Osteocalcin, Opg and Rankl in the male Dmp1-hWNT16 TG mice in bone tissue compared to sex-matched WT mice. These results indicate that WNT16 plays a critical role for acquisition of both cortical and trabecular bone mass and strength. Strategies designed to use WNT16 as a target for therapeutic interventions will be valuable to treat osteoporosis and other low bone mass conditions.


Asunto(s)
Densidad Ósea/fisiología , Osteocitos/metabolismo , Proteínas Wnt/metabolismo , Animales , Densidad Ósea/genética , Huesos/metabolismo , Femenino , Fémur/metabolismo , Fémur/patología , Humanos , Masculino , Ratones , Ratones Transgénicos , Osteoblastos/metabolismo , Osteocalcina/metabolismo , Osteoporosis/genética , Osteoporosis/metabolismo , Proteínas Wnt/genética
9.
Bone ; 90: 127-32, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27297606

RESUMEN

Osteogenesis Imperfecta (OI) comprises a group of genetic skeletal fragility disorders. The mildest form of OI, Osteogenesis Imperfecta type I, is frequently caused by haploinsufficiency mutations in COL1A1, the gene encoding the α1(I) chain of type 1 collagen. Children with OI type I have a 95-fold higher fracture rate compared to unaffected children. Therapies for OI type I in the pediatric population are limited to anti-catabolic agents. In adults with osteoporosis, anabolic therapies that enhance Wnt signaling in bone improve bone mass, and ongoing clinical trials are determining if these therapies also reduce fracture risk. We performed a proof-of-principle experiment in mice to determine whether enhancing Wnt signaling in bone could benefit children with OI type I. We crossed a mouse model of OI type I (Col1a1(+/Mov13)) with a high bone mass (HBM) mouse (Lrp5(+/p.A214V)) that has increased bone strength from enhanced Wnt signaling. Offspring that inherited the OI and HBM alleles had higher bone mass and strength than mice that inherited the OI allele alone. However, OI+HBM and OI mice still had bones with lower ductility compared to wild-type mice. We conclude that enhancing Wnt signaling does not make OI bone normal, but does improve bone properties that could reduce fracture risk. Therefore, agents that enhance Wnt signaling are likely to benefit children and adults with OI type 1.


Asunto(s)
Huesos/patología , Huesos/fisiopatología , Colágeno Tipo I/metabolismo , Osteogénesis Imperfecta/patología , Osteogénesis Imperfecta/fisiopatología , Vía de Señalización Wnt , Alelos , Animales , Fenómenos Biomecánicos , Densidad Ósea , Matriz Ósea/patología , Huesos/diagnóstico por imagen , Huesos/metabolismo , Calcificación Fisiológica , Hueso Esponjoso/diagnóstico por imagen , Hueso Esponjoso/patología , Hueso Esponjoso/fisiopatología , Colágeno Tipo I/genética , Cadena alfa 1 del Colágeno Tipo I , Hueso Cortical/diagnóstico por imagen , Hueso Cortical/patología , Hueso Cortical/fisiopatología , Modelos Animales de Enfermedad , Femenino , Fémur/diagnóstico por imagen , Fémur/patología , Fémur/fisiopatología , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Vértebras Lumbares/diagnóstico por imagen , Vértebras Lumbares/patología , Vértebras Lumbares/fisiopatología , Masculino , Ratones Endogámicos C57BL , Tamaño de los Órganos , Microtomografía por Rayos X
10.
Endocrinology ; 157(8): 3047-57, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27253995

RESUMEN

ß-Catenin (ßcat) is a major downstream signaling node in canonical Wingless-related integration site (Wnt) signaling pathway, and its activity is crucial for canonical Wnt signal transduction. Wnt signaling has recently been implicated in the osteo-anabolic response to PTH, a potent calcium-regulating factor. We investigated whether ßcat is essential for the anabolic action of intermittent PTH by generating male mice with adult-onset deletion of ßcat in a subpopulation of bone cells (osteocytes and late-stage osteoblasts), treating them with an anabolic regimen of PTH, and measuring the skeletal responses. Male (10kb)Dmp1-CreERt2 transgenic mice that also harbored floxed loss-of-function ßcat alleles (ßcat(f/f)) were induced for Cre activity using tamoxifen, then injected daily with human PTH 1-34 (30 µg/kg) or vehicle for 5 weeks. Mice in which ßcat was deleted showed either total lack of bone mineral density (BMD) gain, or BMD loss, and did not respond to PTH treatment. However, bone mass measurements in the trabecular compartment of the femur and spine revealed PTH-induced bone gain whether ßcat was deleted or not. PTH-stimulated increases in periosteal and cancellous bone formation rates were not impaired by ßcat deletion, but resorption markers and cortical porosity were significantly increased in induced mice, particularly induced mice treated with PTH. These results suggest that ßcat is required for net-positive BMD effects of PTH therapy but that the anabolic effects per se of PTH treatment might not require osteocytic/osteoblastic ßcat.


Asunto(s)
Proteínas de la Matriz Extracelular/genética , Eliminación de Gen , Osteogénesis , Hormona Paratiroidea/farmacología , beta Catenina/genética , Factores de Edad , Anabolizantes/metabolismo , Anabolizantes/farmacología , Animales , Densidad Ósea/efectos de los fármacos , Densidad Ósea/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutagénesis , Osteogénesis/efectos de los fármacos , Osteogénesis/genética , Hormona Paratiroidea/metabolismo
12.
Endocrinology ; 157(2): 722-36, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26584014

RESUMEN

Previous genome-wide association studies have identified common variants in genes associated with bone mineral density (BMD) and risk of fracture. Recently, we identified single nucleotide polymorphisms (SNPs) in Wingless-type mouse mammary tumor virus integration site (WNT)16 that were associated with peak BMD in premenopausal women. To further identify the role of Wnt16 in bone mass regulation, we created transgenic (TG) mice overexpressing human WNT16 in osteoblasts. We compared bone phenotypes, serum biochemistry, gene expression, and dynamic bone histomorphometry between TG and wild-type (WT) mice. Compared with WT mice, WNT16-TG mice exhibited significantly higher whole-body areal BMD and bone mineral content (BMC) at 6 and 12 weeks of age in both male and female. Microcomputer tomography analysis of trabecular bone at distal femur revealed 3-fold (male) and 14-fold (female) higher bone volume/tissue volume (BV/TV), and significantly higher trabecular number and trabecular thickness but lower trabecular separation in TG mice compared with WT littermates in both sexes. The cortical bone at femur midshaft also displayed significantly greater bone area/total area and cortical thickness in the TG mice in both sexes. Serum biochemistry analysis showed that male TG mice had higher serum alkaline phosphatase, osteocalcin, osteoprotegerin (OPG), OPG to receptor activator of NF-kB ligand (tumor necrosis family ligand superfamily, number 11; RANKL) ratio as compared with WT mice. Also, lower carboxy-terminal collagen cross-link (CTX) to tartrate-resistant acid phosphatase 5, isoform b (TRAPc5b) ratio was observed in TG mice compared with WT littermates in both male and female. Histomorphometry data demonstrated that both male and female TG mice had significantly higher cortical and trabecular mineralizing surface/bone surface and bone formation rate compared with sex-matched WT mice. Gene expression analysis demonstrated higher expression of Alp, OC, Opg, and Opg to Rankl ratio in bone tissue in the TG mice compared with WT littermates. Our data indicate that WNT16 is critical for positive regulation of both cortical and trabecular bone mass and structure and that this molecule might be targeted for therapeutic interventions to treat osteoporosis.


Asunto(s)
Densidad Ósea/genética , Fémur/diagnóstico por imagen , Osteoblastos/metabolismo , Osteogénesis/genética , ARN Mensajero/metabolismo , Proteínas Wnt/genética , Fosfatasa Ácida/genética , Fosfatasa Ácida/metabolismo , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Animales , Huesos/diagnóstico por imagen , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Femenino , Fémur/metabolismo , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Ratones , Ratones Transgénicos , Osteocalcina/genética , Osteocalcina/metabolismo , Osteoporosis , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , Péptidos/genética , Péptidos/metabolismo , Ligando RANK/genética , Ligando RANK/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Fosfatasa Ácida Tartratorresistente , Vía de Señalización Wnt , Microtomografía por Rayos X
13.
J Cell Physiol ; 231(1): 162-71, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26058470

RESUMEN

Core binding factor ß (Cbfß) is a partner protein of Runx family transcription factors with minimally characterized function in cartilage. Here we address the role of Cbfß in cartilage by generating chondrocyte-specific Cbfß-deficient mice (Cbfb(Δch/Δch) ) from Cbfb-floxed mice crossed with mice expressing Cre from the Col2a1 promoter. Cbfb(Δch/Δch) mice died soon after birth and exhibited delayed endochondral bone formation, shorter appendicular skeleton length with increased proliferative chondrocytes, and nearly absent hypertrophic chondrocyte zones. Immunohistochemical and quantitative real-time PCR analyses showed that the number and size of proliferative chondrocytes increased and the expression of chondrocyte maturation markers at the growth plates, including Runx2, osterix, and osteopontin, significantly diminished in Cbfb(Δch/Δch) mice compared to wild type mice. With regard to signaling pathways, both PTHrP-Ihh and BMP signaling were compromised in Cbfb(Δch/Δch) mice. Mechanistically, Cbfß deficiency in chondrocytes caused a decrease of protein levels of Runx transcription factors by accelerating polyubiquitination-mediated proteosomal degradation in vitro. Indeed, Runx2 and Runx3, but not Runx1, decreased in Cbfb(Δch/Δch) mice. Collectively, these findings indicate that Cbfß plays a critical role for chondrocyte differentiation through stabilizing Runx2 and Runx3 proteins in cartilage.


Asunto(s)
Diferenciación Celular/genética , Condrocitos/citología , Condrogénesis/genética , Subunidad beta del Factor de Unión al Sitio Principal/metabolismo , Placa de Crecimiento/metabolismo , Animales , Cartílago/fisiología , Subunidad beta del Factor de Unión al Sitio Principal/genética , Regulación del Desarrollo de la Expresión Génica/fisiología , Ratones Endogámicos C57BL , Ratones Transgénicos , Osteogénesis/fisiología
15.
Neurosci Lett ; 602: 95-8, 2015 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-26151587

RESUMEN

Neural progenitor cells generate various types of neurons and glia in a tightly regulated manner. During primary neurogenesis, retinoic acid (RA) acts earlier than Notch signaling and regulates differentiation and proliferation by upregulating proneural and neurogenic genes in the neural plate. However, the relationship between Notch signaling and the retinoid pathway during late neurogenesis remains unclear. We investigated the role of Mindbomb (Mib)-mediated Notch signaling in the differentiation of neural progenitors during late neurogenesis by overexpressing Mib and administering RA to Tg[hsp70-Mib:EGFP]. The majority of cells in the p3 domain differentiated into GABAergic Kolmer-Agduhr (KA) cells in Tg[hsp70-mib:EGFP] embryos heat-shocked during late neurogenesis, whereas these phenotypes were suppressed by exogenous RA. Our observations suggest that Mib-mediated Notch signaling plays a critical role in the temporal differentiation of neural progenitors, and that the generation of late-born KA″ cells is regulated by the interplay between Mib and RA.


Asunto(s)
Neuronas/citología , Receptores Notch/fisiología , Médula Espinal/citología , Tretinoina/fisiología , Animales , Animales Modificados Genéticamente , Diferenciación Celular , Embrión no Mamífero , Neuronas GABAérgicas/citología , Neuronas GABAérgicas/metabolismo , Proteínas HSP70 de Choque Térmico/genética , Interneuronas/citología , Interneuronas/metabolismo , Neuronas Motoras/citología , Neuronas Motoras/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis , Neuronas/metabolismo , Regiones Promotoras Genéticas , Células Receptoras Sensoriales/citología , Células Receptoras Sensoriales/metabolismo , Transducción de Señal , Médula Espinal/embriología , Médula Espinal/metabolismo , Tretinoina/farmacología , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/fisiología , Pez Cebra , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/fisiología
16.
J Bone Miner Res ; 30(4): 715-22, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25358268

RESUMEN

Core binding factor beta (Cbfß), the partner protein of Runx family transcription factors, enhances Runx function by increasing the binding of Runx to DNA. Null mutations of Cbfb result in embryonic death, which can be rescued by restoring fetal hematopoiesis but only until birth, where bone formation is still nearly absent. Here, we address a direct role of Cbfß in skeletal homeostasis by generating osteoblast-specific Cbfß-deficient mice (Cbfb(Δob/Δob) ) from Cbfb-floxed mice crossed with mice expressing Cre from the Col1a1 promoter. Cbfb(Δob/Δob) mice showed normal growth and development but exhibited reduced bone mass, particularly of cortical bone. The reduction of bone mass in Cbfb(Δob/Δob) mice is similar to the phenotype of mice with haploinsufficiency of Runx2. Although the number of osteoblasts remained unchanged, the number of active osteoblasts decreased in Cbfb(Δob/Δob) mice and resulted in lower mineral apposition rate. Immunohistochemical and quantitative real-time PCR analyses showed that the expression of osteogenic markers, including Runx2, osterix, osteocalcin, and osteopontin, was significantly repressed in Cbfb(Δob/Δob) mice compared with wild-type mice. Cbfß deficiency also reduced Runx2 protein levels in osteoblasts. The mechanism was revealed by forced expression of Cbfß, which increased Runx2 protein levels in vitro by inhibiting polyubiquitination-mediated proteosomal degradation. Collectively, these findings indicate that Cbfß stabilizes Runx2 in osteoblasts by forming a complex and thus facilitates the proper maintenance of bone mass, particularly cortical bone.


Asunto(s)
Huesos/fisiología , Subunidad beta del Factor de Unión al Sitio Principal/fisiología , Osteoblastos/metabolismo , Animales , Desarrollo Óseo/fisiología , Núcleo Celular/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal , Subunidad beta del Factor de Unión al Sitio Principal/metabolismo , Ratones , Ratones Endogámicos C57BL , Tamaño de los Órganos
17.
Bone ; 61: 91-101, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24401612

RESUMEN

Circulating osteoclast precursor cells highly express CX3C chemokine receptor 1 (CX3CR1), which is the only receptor for the unique CX3C membrane-anchored chemokine, fractalkine (CX3CL1). An irradiated murine model was used to evaluate the role of the CX3CL1-CX3CR1 axis in osteoclast recruitment and osteoclastogenesis. Ionizing radiation (IR) promoted the migration of circulating CD11b+ cells to irradiated bones and dose-dependently increased the number of differentiated osteoclasts in irradiated bones. Notably, CX3CL1 was dramatically upregulated in the vascular endothelium after IR. IR-induced production of CX3CL1 by skeletal vascular endothelium promoted chemoattraction of circulating CX3CR1+/CD11b+ cells and triggered homing of these osteoclast precursor cells toward the bone remodeling surface, a specific site for osteoclast differentiation. CX3CL1 also increased the endothelium-derived expression of other chemokines including stromal cell-derived factor-1 (CXCL12) and macrophage inflammatory protein-2 (CXCL2) by activating the hypoxia-inducible factor-1 α pathway. These effects may further enhance osteoclastogenesis. A series of in vivo experiments confirmed that knockout of CX3CR1 in bone marrow-derived cells and functional inhibition of CX3CL1 using a specific neutralizing antibody significantly ameliorated osteoclastogenesis and prevented bone loss after IR. These results demonstrate that the de novo CX3CL1-CX3CR1 axis plays a pivotal role in osteoclast recruitment and subsequent bone resorption, and verify its therapeutic potential as a new target for anti-resorptive treatment.


Asunto(s)
Resorción Ósea/metabolismo , Huesos/metabolismo , Huesos/efectos de la radiación , Quimiocina CX3CL1/metabolismo , Endotelio Vascular/metabolismo , Osteoclastos/metabolismo , Animales , Receptor 1 de Quimiocinas CX3C , Células Cultivadas , Modelos Animales de Enfermedad , Citometría de Flujo , Humanos , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Confocal , Osteoclastos/citología , Receptores de Quimiocina/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/citología , Células Madre/metabolismo
18.
PLoS One ; 8(12): e83584, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24349534

RESUMEN

The transdifferentiation of vascular smooth muscle cells (VSMCs) into osteoblast-like cells has been implicated in the context of vascular calcification. We investigated the roles of vitamin D receptor (Vdr) and runt-related transcription factor 2 (Runx2) in the osteoblastic differentiation of VSMCs in response to vitamin D3 using in vitro VSMCs cultures and in vivo in Vdr knockout (Vdr(-/-)) and Runx2 carboxy-terminus truncated heterozygous (Runx2(+/ΔC)) mice. Treatment of VSMCs with active vitamin D3 promoted matrix mineral deposition, and increased the expressions of Vdr, Runx2, and of osteoblastic genes but decreased the expression of smooth muscle myosin heavy chain in primary VSMCs cultures. Immunoprecipitation experiments suggested an interaction between Vdr and Runx2. Furthermore, silencing Vdr or Runx2 attenuated the procalcific effects of vitamin D3. Functional cooperation between Vdr and Runx2 in vascular calcification was also confirmed in in vivo mouse models. Vascular calcification induced by high-dose vitamin D3 was completely inhibited in Vdr(-/-) or Runx2(+/ΔC) mice, despite elevated levels of serum calcium or alkaline phosphatase. Collectively, these findings suggest that functional cooperation between Vdr and Runx2 is necessary for vascular calcification in response to vitamin D3.


Asunto(s)
Conservadores de la Densidad Ósea/efectos adversos , Colecalciferol/efectos adversos , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Receptores de Calcitriol/metabolismo , Calcificación Vascular , Animales , Conservadores de la Densidad Ósea/farmacología , Células Cultivadas , Colecalciferol/farmacología , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Ratones , Ratones Noqueados , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Ratas , Receptores de Calcitriol/genética , Calcificación Vascular/inducido químicamente , Calcificación Vascular/genética , Calcificación Vascular/metabolismo , Calcificación Vascular/patología
19.
Bone ; 50(1): 149-55, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22036912

RESUMEN

We investigated the therapeutic effect of topical Risedronate (RIS) on a mouse model of estrogen-deficient osteoporosis. Fourteen-week-old female mice were ovariectomized and assigned to 4 groups: SHAM-operated (SHAM), OVX mice treated with vehicle (OVX-V), OVX mice treated with 0.2% RIS (OVX-0.2% RIS), and OVX-mice treated with 0.02% RIS (OVX-0.02% RIS). Topical samples containing RIS were prepared in 10% (w/w) polyethylene glycol (PEG, MW 400) and 80 µg of sample was spread on the mice's mid-backs every 3 days for 5 weeks. Micro-CT analysis of femora demonstrated that OVX-0.2% RIS exhibited a 29% greater bone mineral density and 24% greater bone volume fraction than that of OVX-V group. Investigation of the trabecular bone in OVX-0.2% RIS revealed a 24% higher bone volume (BV/TV), 51% higher trabecular number (Tb.N), and 40% lower trabecular separation (Tb.Sp) compared to OVX-V mice. Additionally, bone phenotypes of tibiae were further confirmed by histological analysis. OVX-0.2% RIS group exhibited a 494% greater BV/TV, 464% less Tb.Sp, 81% greater active osteoclast surface (Oc.S/BS) and 26% less osteoclast number (N.Oc/BS) than that of OVX-V group. Collectively, these results indicated that topical delivery of RIS has powerful pharmaceutical effects on the prevention of osteoporosis and bone turnover.


Asunto(s)
Conservadores de la Densidad Ósea/administración & dosificación , Conservadores de la Densidad Ósea/uso terapéutico , Ácido Etidrónico/análogos & derivados , Osteoporosis/tratamiento farmacológico , Administración Tópica , Animales , Densidad Ósea/efectos de los fármacos , Conservadores de la Densidad Ósea/farmacología , Ácido Etidrónico/administración & dosificación , Ácido Etidrónico/farmacología , Ácido Etidrónico/uso terapéutico , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Ovariectomía , Ácido Risedrónico , Piel/citología , Piel/efectos de los fármacos , Tibia/efectos de los fármacos , Tibia/ultraestructura , Microtomografía por Rayos X
20.
Soc Work Public Health ; 25(2): 223-36, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20391263

RESUMEN

Little attention has been paid to health status issues among elderly individuals in Korea. The purpose of this secondary data analysis was to develop a better understanding of relationship between a number of socioeconomic factors and subjective social status affecting risk behaviors and health status among Korean elderly. Path analyses of 9,585 Korean elderly provided findings that those who reported a higher level of socioeconomic status were less likely to engage in risk behaviors and thus more likely to have better health status. Further, those who reported a higher level of socio-economic status were more likely to have a higher level of subjective social status and thus have better health status. Implications for policy and practice are discussed.


Asunto(s)
Estado de Salud , Asunción de Riesgos , Clase Social , Anciano , Femenino , Encuestas Epidemiológicas , Humanos , Corea (Geográfico) , Masculino , Programas Nacionales de Salud
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA