Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 183
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 175(7): 1756-1768.e17, 2018 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-30550785

RESUMEN

Irisin is secreted by muscle, increases with exercise, and mediates certain favorable effects of physical activity. In particular, irisin has been shown to have beneficial effects in adipose tissues, brain, and bone. However, the skeletal response to exercise is less clear, and the receptor for irisin has not been identified. Here we show that irisin binds to proteins of the αV class of integrins, and biophysical studies identify interacting surfaces between irisin and αV/ß5 integrin. Chemical inhibition of the αV integrins blocks signaling and function by irisin in osteocytes and fat cells. Irisin increases both osteocytic survival and production of sclerostin, a local modulator of bone remodeling. Genetic ablation of FNDC5 (or irisin) completely blocks osteocytic osteolysis induced by ovariectomy, preventing bone loss and supporting an important role of irisin in skeletal remodeling. Identification of the irisin receptor should greatly facilitate our understanding of irisin's function in exercise and human health.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Remodelación Ósea , Fibronectinas/metabolismo , Integrina alfaV/metabolismo , Osteocitos/metabolismo , Osteólisis/metabolismo , Adipocitos/patología , Animales , Línea Celular Tumoral , Femenino , Fibronectinas/genética , Células HEK293 , Humanos , Integrina alfaV/genética , Ratones , Osteocitos/patología , Osteólisis/genética
3.
Proc Natl Acad Sci U S A ; 120(46): e2312677120, 2023 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-37931101

RESUMEN

We have previously reported that the cortical bone thinning seen in mice lacking the Wnt signaling antagonist Sfrp4 is due in part to impaired periosteal apposition. The periosteum contains cells which function as a reservoir of stem cells and contribute to cortical bone expansion, homeostasis, and repair. However, the local or paracrine factors that govern stem cells within the periosteal niche remain elusive. Cathepsin K (Ctsk), together with additional stem cell surface markers, marks a subset of periosteal stem cells (PSCs) which possess self-renewal ability and inducible multipotency. Sfrp4 is expressed in periosteal Ctsk-lineage cells, and Sfrp4 global deletion decreases the pool of PSCs, impairs their clonal multipotency for differentiation into osteoblasts and chondrocytes and formation of bone organoids. Bulk RNA sequencing analysis of Ctsk-lineage PSCs demonstrated that Sfrp4 deletion down-regulates signaling pathways associated with skeletal development, positive regulation of bone mineralization, and wound healing. Supporting these findings, Sfrp4 deletion hampers the periosteal response to bone injury and impairs Ctsk-lineage periosteal cell recruitment. Ctsk-lineage PSCs express the PTH receptor and PTH treatment increases the % of PSCs, a response not seen in the absence of Sfrp4. Importantly, in the absence of Sfrp4, PTH-dependent increase in cortical thickness and periosteal bone formation is markedly impaired. Thus, this study provides insights into the regulation of a specific population of periosteal cells by a secreted local factor, and shows a central role for Sfrp4 in the regulation of Ctsk-lineage periosteal stem cell differentiation and function.


Asunto(s)
Osteogénesis , Nicho de Células Madre , Ratones , Animales , Catepsina K/metabolismo , Periostio/metabolismo , Diferenciación Celular/genética , Vía de Señalización Wnt , Proteínas Proto-Oncogénicas/metabolismo
4.
J Biol Chem ; 296: 100790, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34019873

RESUMEN

Deletion of c-Src, a ubiquitously expressed tyrosine kinase, results in osteoclast dysfunction and osteopetrosis, in which bones harden into "stone." In contrast, deletion of the genes encoding other members of the Src family kinase (SFK) fails to produce an osteopetrotic phenotype. This suggests that c-Src performs a unique function in the osteoclast that cannot be compensated for by other SFKs. We aimed to identify the molecular basis of this unique role in osteoclasts and bone resorption. We found that c-Src, Lyn, and Fyn were the most highly expressed SFKs in WT osteoclasts, whereas Hck, Lck, Blk, and Fgr displayed low levels of expression. Formation of the podosome belt, clusters of unique actin assemblies, was disrupted in src-/- osteoclasts; introduction of constitutively activated SFKs revealed that only c-Src and Fyn could restore this process. To identify the key structural domains responsible, we constructed chimeric Src-Hck and Src-Lyn constructs in which the unique, SH3, SH2, or catalytic domains had been swapped. We found that the Src unique, SH3, and kinase domains were each crucial to establish Src functionality. The SH2 domain could however be substituted with Lyn or Hck SH2 domains. Furthermore, we demonstrate that c-Src's functionality is, in part, derived from an SH3-proximal proline-rich domain interaction with c-Cbl, leading to phosphorylation of c-Cbl Tyr700. These data help clarify Src's unique functionality in the organization of the cytoskeleton in osteoclasts, required for efficient bone resorption and explain why c-Src cannot be replaced, in osteoclasts, by other SFKs.


Asunto(s)
Osteoclastos/metabolismo , Podosomas/metabolismo , Dominios Homologos src , Familia-src Quinasas/metabolismo , Animales , Resorción Ósea/genética , Resorción Ósea/metabolismo , Diferenciación Celular , Células HEK293 , Humanos , Ratones , Osteoclastos/citología , Familia-src Quinasas/genética
5.
Connect Tissue Res ; 63(2): 97-111, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-31868022

RESUMEN

Introduction: With age, the number of adipocytes and osteoclasts increases, the number of osteoblasts decreases, and mechano-adaptation is impaired.Objectives: Using marrow aspiration, which has a known osteogenic effect in young mice, we sought to recruit osteoblast progenitors to mediate the mechano-adaptive response to in vivo tibial loading.Methods: First, we assessed bone formation and marrow adiposity in the tibiae of old mice (>20 months) sacrificed 1, 2, and 4 weeks after unilateral marrow aspiration. Then, we examined the effects of marrow aspiration on mechano-adaptation in aged mice using tibial loading.Results: Two weeks after aspiration, aspirated tibiae had more bone than contralateral tibiae due to the formation of bone in the medullary canal. Two weeks and four weeks after marrow aspiration, the volume of marrow adipose tissue was higher in the aspirated tibiae, compared to contralateral tibiae. Histomorphometry indicated that aspiration increased non-periosteal (endosteal, intracortical, intramedullary) bone formation, compared to the contralateral tibia.  Mice with marrow aspiration had reduced periosteal bone formation in the contralateral tibia, compared to mice that had loading alone. Loading-induced periosteal bone formation was higher in mice that had loading alone, compared to mice that had aspiration + loading, indicating that aspiration further reduced the mechano-adaptive response.Conclusion: These data demonstrate that, in old mice, bone forms in the medullary canal following aspiration. Adiposity is increased following marrow aspiration, and periosteal mechano-adaptation is reduced.


Asunto(s)
Médula Ósea , Osteogénesis , Tejido Adiposo , Animales , Ratones , Ratones Endogámicos C57BL , Osteogénesis/fisiología , Tibia
6.
Surg Endosc ; 36(9): 6984-6996, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35226161

RESUMEN

BACKGROUND: Bariatric surgery has been shown to result in weight loss, improved hemoglobin A1C, and decreased mortality but can also lead to bone loss and increased fracture rates. Serum IGFBP-2 is elevated in patients after bariatric surgery and although it may lead to improved blood glucose, may also drive bone resorption, and inhibit IGF-I action. This study tested the hypothesis that Igfbp2-/- mice were acutely protected from bone loss after vertical sleeve gastrectomy (VSG). METHODS: Thirty-four mice, 17 Igfbp2-/- and 17 + / + underwent a hand-sewn VSG or sham surgery, at 16 weeks of age. Mice were harvested at 20 weeks of age. DXA was measured for body composition, areal bone mineral density (aBMD), areal bone mineral content (aBMC), femoral bone mineral density (fBMD), and femoral bone mineral content (fBMC) at 15, 18, and 20 weeks of age. Micro-computed tomography and serum ELISA assays were measured and analyzed at 20 weeks of age. RESULTS: Both Igfbp2-/- and + / + mice lost significant weight (P = 0.0251, P = 0.0003, respectively) and total fat mass (P = 0.0082, P = 0.0004, respectively) at 4 weeks after VSG. Igfbp2+/+ mice lost significant aBMD, fBMD, fBMC, trabecular BMD, trabecular BV/TV and cortical tissue mineral density (P = 0.0150, P = 0.0313, P = 0.0190, P = 0.0072, and 0.0320 respectively). The Igfbp2-/- mice did not show significant bone loss in these parameters nor in trabecular BV/TV. Both Igfbp2-/- and + / + mice had less cortical bone area (P = 0.0181, P = < .00001), cortical area over total area (P = 0.0085, P = 0.0007), and cortical thickness (P = 0.0050, P = < 0.0001), respectively. Igfbp2+/+ mice demonstrated significantly lower polar, minimum, and maximum moments of inertia (P = 0.0031, P = 0.0239, and P = 0.0037, respectively). Igfbp2+/+ had significantly higher levels of IGFBP-2 at 2 weeks postoperatively after VSG (P = 0.035), and elevated levels of CTx and P1NP (P = 0.0127, P = 0.0058, respectively). CONCLUSIONS: Igfbp2-/- mice were protected against trabecular bone loss and had attenuated cortical bone loss 4 weeks after VSG.


Asunto(s)
Hueso Esponjoso , Gastrectomía/efectos adversos , Proteína 2 de Unión a Factor de Crecimiento Similar a la Insulina , Osteoporosis/genética , Animales , Densidad Ósea , Hueso Esponjoso/diagnóstico por imagen , Proteína 2 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Ratones , Osteoporosis/patología , Microtomografía por Rayos X
7.
Curr Osteoporos Rep ; 20(2): 153-161, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35182301

RESUMEN

PURPOSE OF REVIEW: Periosteal apposition and endosteal remodeling regulate cortical bone expansion and thickness, both critical determinants of bone strength. Yet, the cellular characteristics and local or paracrine factors that regulate the periosteum and endosteum remain largely elusive. Here we discuss novel insights in cortical bone growth, expansion, and homeostasis, provided by the study of Secreted Frizzled Receptor Protein 4 (Sfrp4), a decoy receptor for Wnt ligands. RECENT FINDINGS: SFRP4 loss-of function mutations cause Pyle disease, a rare skeletal disorder characterized by cortical bone thinning and increased fragility fractures despite increased trabecular bone density. On the endosteal surface, Sfrp4-mediated repression of non-canonical Wnt signaling regulates endosteal resorption. On the periosteum, Sfrp4 identifies as a critical functional mediator of periosteal stem cell/progenitor expansion and differentiation. Analysis of signaling pathways regulating skeletal stem cells/progenitors provides an opportunity to advance our understanding of the mechanisms involved in cortical bone biology.


Asunto(s)
Hueso Cortical , Receptores Frizzled , Biología , Diferenciación Celular , Humanos , Periostio , Proteínas Proto-Oncogénicas
8.
Proc Natl Acad Sci U S A ; 116(28): 14138-14143, 2019 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-31239337

RESUMEN

Loss-of-function mutations in the Wnt inhibitor secreted frizzled receptor protein 4 (SFRP4) cause Pyle's disease (OMIM 265900), a rare skeletal disorder characterized by wide metaphyses, significant thinning of cortical bone, and fragility fractures. In mice, we have shown that the cortical thinning seen in the absence of Sfrp4 is associated with decreased periosteal and endosteal bone formation and increased endocortical resorption. While the increase in Rankl/Opg in cortical bone of mice lacking Sfrp4 suggests an osteoblast-dependent effect on endocortical osteoclast (OC) activity, whether Sfrp4 can cell-autonomously affect OCs is not known. We found that Sfrp4 is expressed during bone marrow macrophage OC differentiation and that Sfrp4 significantly suppresses the ability of early and late OC precursors to respond to Rankl-induced OC differentiation. Sfrp4 deletion in OCs resulted in activation of canonical Wnt/ß-catenin and noncanonical Wnt/Ror2/Jnk signaling cascades. However, while inhibition of canonical Wnt/ß-catenin signaling did not alter the effect of Sfrp4 on OCgenesis, blocking the noncanonical Wnt/Ror2/Jnk cascade markedly suppressed its regulation of OC differentiation in vitro. Importantly, we report that deletion of Ror2 exclusively in OCs (CtskCreRor2fl/fl ) in Sfrp4 null mice significantly reversed the increased number of endosteal OCs seen in these mice and reduced their cortical thinning. Altogether, these data show autocrine and paracrine effects of Sfrp4 in regulating OCgenesis and demonstrate that the increase in endosteal OCs seen in Sfrp4-/- mice is a consequence of noncanonical Wnt/Ror2/Jnk signaling activation in OCs overriding the negative effect that activation of canonical Wnt/ß-catenin signaling has on OCgenesis.


Asunto(s)
Resorción Ósea/genética , MAP Quinasa Quinasa 4/genética , Osteoclastos/metabolismo , Proteínas Proto-Oncogénicas/genética , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/genética , Animales , Comunicación Autocrina/genética , Resorción Ósea/patología , Huesos/metabolismo , Diferenciación Celular/genética , Hueso Cortical/crecimiento & desarrollo , Hueso Cortical/patología , Regulación del Desarrollo de la Expresión Génica , Humanos , Ratones , Ratones Noqueados , Osteoblastos/metabolismo , Osteoblastos/patología , Osteocondrodisplasias/genética , Osteoclastos/patología , Comunicación Paracrina/genética , Eliminación de Secuencia , Vía de Señalización Wnt/genética
9.
Stem Cells ; 38(6): 769-781, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32053258

RESUMEN

Bone remodeling and regeneration are dependent on resident stem/progenitor cells with the ability to replenish mature osteoblasts and repair the skeleton. Using lineage tracing approaches, we identified a population of Dmp1+ cells that reside within cortical bone and are distinct from osteocytes. Our aims were to characterize this stromal population of transcortical perivascular cells (TPCs) in their resident niche and evaluate their osteogenic potential. To distinguish this population from osteoblasts/osteocytes, we crossed mice containing inducible DMP1CreERT2/Ai9 Tomato reporter (iDMP/T) with Col2.3GFP reporter (ColGFP), a marker of osteoblasts and osteocytes. We observed iDMP/T+;ColGFP- TPCs within cortical bone following tamoxifen injection. These cells were perivascular and located within transcortical channels. Ex vivo bone outgrowth cultures showed TPCs migrated out of the channels onto the plate and expressed stem cell markers such as Sca1, platelet derived growth factor receptor beta (PDGFRß), and leptin receptor. In a cortical bone transplantation model, TPCs migrate from their vascular niche within cortical bone and contribute to new osteoblast formation and bone tube closure. Treatment with intermittent parathyroid hormone increased TPC number and differentiation. TPCs were unable to differentiate into adipocytes in the presence of rosiglitazone in vitro or in vivo. Altogether, we have identified and characterized a novel stromal lineage-restricted osteoprogenitor that is associated with transcortical vessels of long bones. Functionally, we have demonstrated that this population can migrate out of cortical bone channels, expand, and differentiate into osteoblasts, therefore serving as a source of progenitors contributing to new bone formation.


Asunto(s)
Huesos/fisiopatología , Osteoblastos/metabolismo , Osteocitos/metabolismo , Animales , Diferenciación Celular , Humanos , Ratones
10.
Nat Immunol ; 9(2): 186-93, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18084294

RESUMEN

Immunoreceptor tyrosine-based activation motif (ITAM)-coupled receptors modulate the amplitude and nature of macrophage responses to Toll-like receptor and cytokine receptor stimulation. However, the molecular mechanisms enabling this receptor crosstalk are not known. Here we investigated the function of the calcium-dependent kinases CaMK and Pyk2 'downstream' of ITAM-associated receptors in the regulation of cytokine-induced activation of Jak kinases and STAT transcription factors. CaMK and Pyk2 relayed signals from integrins and the ITAM-containing adaptor DAP12 to augment interleukin 10- and interferon-alpha-induced Jak activation and STAT1-dependent gene expression. CaMK inhibition suppressed STAT1-mediated interferon-alpha signaling in a mouse model of systemic lupus erythematosus. Our results associate Pyk2 and Jak kinases with the linkage of signals emanating from cytokine and heterologous ITAM-dependent receptors.


Asunto(s)
Quinasa de la Proteína Quinasa Dependiente de Calcio-Calmodulina/metabolismo , Quinasa 2 de Adhesión Focal/metabolismo , Interferón Tipo I/farmacología , Quinasas Janus/metabolismo , Macrófagos/efectos de los fármacos , Factor de Transcripción STAT1/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Calcio/metabolismo , Células Cultivadas , Humanos , Macrófagos/inmunología , Proteínas de la Membrana , Ratones , Fosforilación , Receptores Inmunológicos/metabolismo , Transducción de Señal , Tirosina/metabolismo
11.
N Engl J Med ; 374(26): 2553-2562, 2016 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-27355534

RESUMEN

BACKGROUND: Cortical-bone fragility is a common feature in osteoporosis that is linked to nonvertebral fractures. Regulation of cortical-bone homeostasis has proved elusive. The study of genetic disorders of the skeleton can yield insights that fuel experimental therapeutic approaches to the treatment of rare disorders and common skeletal ailments. METHODS: We evaluated four patients with Pyle's disease, a genetic disorder that is characterized by cortical-bone thinning, limb deformity, and fractures; two patients were examined by means of exome sequencing, and two were examined by means of Sanger sequencing. After a candidate gene was identified, we generated a knockout mouse model that manifested the phenotype and studied the mechanisms responsible for altered bone architecture. RESULTS: In all affected patients, we found biallelic truncating mutations in SFRP4, the gene encoding secreted frizzled-related protein 4, a soluble Wnt inhibitor. Mice deficient in Sfrp4, like persons with Pyle's disease, have increased amounts of trabecular bone and unusually thin cortical bone, as a result of differential regulation of Wnt and bone morphogenetic protein (BMP) signaling in these two bone compartments. Treatment of Sfrp4-deficient mice with a soluble Bmp2 receptor (RAP-661) or with antibodies to sclerostin corrected the cortical-bone defect. CONCLUSIONS: Our study showed that Pyle's disease was caused by a deficiency of sFRP4, that cortical-bone and trabecular-bone homeostasis were governed by different mechanisms, and that sFRP4-mediated cross-regulation between Wnt and BMP signaling was critical for achieving proper cortical-bone thickness and stability. (Funded by the Swiss National Foundation and the National Institutes of Health.).


Asunto(s)
Densidad Ósea/genética , Remodelación Ósea/genética , Osteocondrodisplasias/genética , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/genética , Adolescente , Animales , Biomarcadores/sangre , Proteínas Morfogenéticas Óseas/metabolismo , Remodelación Ósea/fisiología , Huesos/patología , Huesos/fisiología , Preescolar , Modelos Animales de Enfermedad , Femenino , Eliminación de Gen , Homeostasis , Humanos , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Osteocondrodisplasias/fisiopatología , Análisis de Secuencia de ADN , Transducción de Señal , Proteínas Wnt/metabolismo
12.
Blood ; 130(5): 619-624, 2017 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-28615219

RESUMEN

The concept that tumor-initiating cells can co-opt the self-renewal program of endogenous stem cells as a means of enforcing their unlimited proliferative potential is widely accepted, yet identification of specific factors that regulate self-renewal of normal and cancer stem cells remains limited. Using a comparative transcriptomic approach, we identify ZNF521/Zfp521 as a conserved hematopoietic stem cell (HSC)-enriched transcription factor in human and murine hematopoiesis whose function in HSC biology remains elusive. Competitive serial transplantation assays using Zfp521-deficient mice revealed that ZFP521 regulates HSC self-renewal and differentiation. In contrast, ectopic expression of ZFP521 in HSCs led to a robust maintenance of progenitor activity in vitro. Transcriptional analysis of human acute myeloid leukemia (AML) patient samples revealed that ZNF521 is highly and specifically upregulated in AMLs with MLL translocations. Using an MLL-AF9 murine leukemia model and serial transplantation studies, we show that ZFP521 is not required for leukemogenesis, although its absence leads to a significant delay in leukemia onset. Furthermore, knockdown of ZNF521 reduced proliferation in human leukemia cell lines possessing MLL-AF9 translocations. Taken together, these results identify ZNF521/ZFP521 as a critical regulator of HSC function, which facilitates MLL-AF9-mediated leukemic disease in mice.


Asunto(s)
Proliferación Celular , Proteínas de Unión al ADN/metabolismo , Células Madre Hematopoyéticas/metabolismo , Neoplasias Experimentales/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Factores de Transcripción/metabolismo , Animales , Línea Celular Tumoral , Proteínas de Unión al ADN/genética , Células Madre Hematopoyéticas/patología , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Ratones , Ratones Noqueados , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Neoplasias Experimentales/patología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Factores de Transcripción/genética , Translocación Genética
13.
Calcif Tissue Int ; 105(3): 316-330, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31243483

RESUMEN

The mechano-adaptive response of bone to loading in the murine uniaxial tibial loading model is impaired in aged animals. Previous studies have shown that in aged mice, the amount of bone formed in response to loading is augmented when loads are applied following sciatic neurectomy. The synergistic effect of neurectomy and loading remains to be elucidated. We hypothesize that sciatic neurectomy increases cellular presence, thereby augmenting the response to load in aged mice. We examined bone adaptation in four groups of female C57BL/6J mice, 20-22 months old: (1) sham surgery + 9N loading; (2) sciatic neurectomy, sacrificed after 5 days; (3) sciatic neurectomy, sacrificed after 19 days; (4) sciatic neurectomy + 9N loading. We examined changes in bone cross sectional properties with micro-CT images, and static and dynamic histomorphometry with histological sections taken at the midpoint between tibiofibular junctions. The response to loading at 9N was not detectable with quantitative micro-CT data, but surface-specific histomorphometry captured an increase in bone formation in specific regions. 5 days following sciatic neurectomy, the amount of bone in the neurectomized leg was the same as the contralateral leg, but 19 days following sciatic neurectomy, there was significant bone loss in the neurectomized leg, and both osteoclasts and osteoblasts were recruited to the endosteal surfaces. When sciatic neurectomy and loading at 9N were combined, 3 out of 4 bone quadrants had increased bone formation, on the endosteal and periosteal surfaces (increased osteoid surface and mineralizing surface respectively). These data demonstrate that sciatic neurectomy increases cellular presence on the endosteal surface. With long-term sciatic-neurectomy, both osteoclasts and osteoblasts were recruited to the endosteal surface, which resulted in increased bone formation when combined with a sufficient mechanical stimulus. Controlled and localized recruitment of both osteoblasts and osteoclasts combined with appropriate mechanical loading could inform therapies for mechanically-directed bone formation.


Asunto(s)
Adaptación Fisiológica/fisiología , Envejecimiento/fisiología , Proliferación Celular/fisiología , Fenómenos Mecánicos , Osteogénesis/fisiología , Nervio Ciático/lesiones , Nervio Ciático/patología , Animales , Desnervación , Femenino , Ratones , Ratones Endogámicos C57BL , Tibia , Soporte de Peso/fisiología , Microtomografía por Rayos X
14.
Proc Natl Acad Sci U S A ; 113(9): E1226-35, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26884171

RESUMEN

Proper tuning of ß-catenin activity in osteoblasts is required for bone homeostasis, because both increased and decreased ß-catenin activity have pathologic consequences. In the classical pathway for ß-catenin activation, stimulation with WNT ligands suppresses constitutive phosphorylation of ß-catenin by glycogen synthase kinase 3ß, preventing ß-catenin ubiquitination and proteasomal degradation. Here, we have found that mitogen-activated protein kinase kinase kinase 2 (MAP3K2 or MEKK2) mediates an alternative pathway for ß-catenin activation in osteoblasts that is distinct from the canonical WNT pathway. FGF2 activates MEKK2 to phosphorylate ß-catenin at serine 675, promoting recruitment of the deubiquitinating enzyme, ubiquitin-specific peptidase 15 (USP15). USP15 in turn prevents the basal turnover of ß-catenin by inhibiting its ubiquitin-dependent proteasomal degradation, thereby enhancing WNT signaling. Analysis of MEKK2-deficient mice and genetic interaction studies between Mekk2- and ß-catenin-null alleles confirm that this pathway is an important physiologic regulator of bone mass in vivo. Thus, an FGF2/MEKK2 pathway mediates an alternative nonclassical pathway for ß-catenin activation, and this pathway is a key regulator of bone formation by osteoblasts.


Asunto(s)
Desarrollo Óseo , MAP Quinasa Quinasa Quinasa 2/metabolismo , beta Catenina/metabolismo , Animales , Ratones , Tamaño de los Órganos , Osteoblastos/citología , Fosforilación
15.
J Cell Physiol ; 233(2): 1585-1600, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28631813

RESUMEN

There is a growing and alarming prevalence of obesity and the metabolic syndrome in type I diabetic patients (T1DM), particularly in adolescence. In general, low bone mass, higher fracture risk, and increased marrow adipose tissue (MAT) are features of diabetic osteopathy in insulin-deficient subjects. On the other hand, type 2 diabetes (T2DM) is associated with normal or high bone mass, a greater risk of peripheral fractures, and no change in MAT. Therefore, we sought to determine the effect of weight gain on bone turnover in insulin-deficient mice. We evaluated the impact of a 6-week high-fat (HFD) rich in medium chain fatty acids or low-fat diet (LFD) on bone mass and MAT in a streptozotocin (STZ)-induced model using male C57BL/6J mice at 8 weeks of age. Dietary intervention was initiated after diabetes confirmation. At the endpoint, lower non-fasting glucose levels were observed in diabetic mice fed with high fat diet compared to diabetic mice fed the low fat diet (STZ-LFD). Compared to euglycemic controls, the STZ-LFD had marked polydipsia and polyphagia, as well as reduced lean mass, fat mass, and bone parameters. Interestingly, STZ-HFD mice had higher bone mass, namely less cortical bone loss and more trabecular bone than STZ-LFD. Thus, we found that a HFD, rich in medium chain fatty acids, protects against bone loss in a T1DM mouse model. Whether this may also translate to T1DM patients who are overweight or obese in respect to maintenance of bone mass remains to be determined through longitudinal studies.


Asunto(s)
Glucemia/metabolismo , Composición Corporal , Remodelación Ósea , Diabetes Mellitus Experimental/dietoterapia , Diabetes Mellitus Tipo 1/dietoterapia , Dieta Alta en Grasa , Ácidos Grasos/administración & dosificación , Osteoporosis/prevención & control , Estreptozocina , Adiposidad , Animales , Biomarcadores/sangre , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/fisiopatología , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/inducido químicamente , Diabetes Mellitus Tipo 1/fisiopatología , Insulina/sangre , Cetonas/sangre , Masculino , Ratones Endogámicos C57BL , Osteoporosis/sangre , Osteoporosis/inducido químicamente , Osteoporosis/fisiopatología , Factores de Tiempo , Pérdida de Peso
16.
Calcif Tissue Int ; 102(1): 93-104, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29032463

RESUMEN

Calcium is essential for normal bone growth and development. Inadequate calcium intake increases the risk of osteoporosis and fractures. Kit ligand/c-Kit signaling plays an important role in regulating bone homeostasis. Mice with c-Kit mutations are osteopenic. The present study aimed to investigate whether impairment of or reduction in c-Kit signaling affects bone turnover during calcium deprivation. Three-week-old male WBB6F1/J-Kit W /Kit W-v /J (W/W v ) mice with c-Kit point mutation, Kit W-sh /HNihrJaeBsmJ (W sh /W sh ) mice with an inversion mutation in the regulatory elements upstream of the c-Kit promoter region, and their wild-type controls (WT) were fed either a normal (0.6% calcium) or a low calcium diet (0.02% calcium) for 3 weeks. µCT analysis indicated that both mutants fed normal calcium diet had significantly decreased cortical thickness and cancellous bone volume compared to WT. The low calcium diet resulted in a comparable reduction in cortical bone volume and cortical thickness in the W/W v and W sh /W sh mice, and their corresponding controls. As expected, the low calcium diet induced cancellous bone loss in the W/W v mice. In contrast, W sh /W sh cancellous bone did not respond to this diet. This c-Kit mutation prevented cancellous bone loss by antagonizing the low calcium diet-induced increase in osteoblast and osteoclast numbers in the W sh /W sh mice. Gene expression profiling showed that calcium deficiency increased Osx, Ocn, Alp, type I collagen, c-Fms, M-CSF, and RANKL/OPG mRNA expression in controls; however, the W sh mutation suppressed these effects. Our findings indicate that although calcium restriction increased bone turnover, leading to osteopenia, the decreased c-Kit expression levels in the W sh /W sh mice prevented the low calcium diet-induced increase in cancellous bone turnover and bone loss but not the cortical bone loss.


Asunto(s)
Huesos/metabolismo , Calcio/metabolismo , Hueso Esponjoso/metabolismo , Mutación/genética , Proteínas Proto-Oncogénicas c-kit/genética , Animales , Desarrollo Óseo/genética , Enfermedades Óseas Metabólicas/metabolismo , Remodelación Ósea/genética , Calcio de la Dieta/metabolismo , Ratones Transgénicos , Osteoblastos/metabolismo , Osteoclastos/metabolismo
17.
Calcif Tissue Int ; 103(6): 686-697, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30116830

RESUMEN

Patients with systemic lupus erythematosus (SLE), a chronic inflammatory disease characterized by loss of T- and B-cell tolerance to autoantigens, are at increased risk for osteoporosis and fractures. Mice deficient in Fc gamma receptor IIb (FcγRIIB) exhibit spontaneous SLE and its restoration rescues the disease. To determine whether deleting FcγRIIB affects cortical bone mass and mechanical properties, we analyzed cortical bone phenotype of FcγRIIB knockouts at different ages. FACS analysis revealed that 6-month-old FcγRIIB-/- mice had increased B220lowCD138+ cells, markers of plasma cells, indicating active SLE disease. In contrast, 3-month-old FcγRIIB-/- mice did not develop the active SLE disease. µCT analysis indicated that FcγRIIB deletion did not affect cortical bone in 3-month-old mutants. However, 6- and 10-month-old FcγRIIB-/- males and females had osteopenic cortical bone and the severity of bone loss increased with disease duration. FcγRIIB deletion decreased cross-sectional area, cortical area, and marrow area in 6-month-old males. Cortical area and cortical thickness were decreased in 10-month-old FcγRIIB-/- males. Lack of FcγRIIB decreased cortical thickness without affecting cortical area in females. However, deletion of a single FcγRIIB allele was insufficient to induce cortical bone loss. The bending strength was decreased in 6- and 10-month-old FcγRIIB-deficient males compared to WT controls. A microindentation analysis demonstrated significantly decreased hardness in both 10-month-old FcγRIIB-/- males and females. Our data indicate that FcγRIIB contributes to the regulation of cortical bone homeostasis subsequent to SLE development and that deletion of FcγRIIB in mice leads to SLE-like disease associated with cortical bone loss and decreased bending strength and hardness.


Asunto(s)
Hueso Cortical/patología , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/patología , Receptores de IgG/deficiencia , Animales , Enfermedades Óseas Metabólicas/genética , Enfermedades Óseas Metabólicas/metabolismo , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de IgG/genética
18.
FASEB J ; 31(5): 2050-2064, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28183805

RESUMEN

Circulating levels of bone-derived fibroblast growth factor 23 (FGF23) increase early during acute and chronic kidney disease and are associated with adverse outcomes. Membrane-bound Klotho acts as a permissive coreceptor for FGF23, and its expression was recently found in osteoblasts/osteocytes. We hypothesized that Klotho in bone cells is part of an autocrine feedback loop that regulates FGF23 expression during renal failure. Thus, we induced renal failure in mice with targeted deletion of Klotho in long bones. Uremic wild-type (KLfl/fl ) and knockout (Prx1-Cre;KLfl/fl ) mice both responded with reduced body weight, kidney atrophy, hyperphosphatemia, and increased bone turnover. Importantly, long bones of Prx1-Cre;KLfl/fl mice but not their axial skeleton failed to increase FGF23 expression as observed in uremic KLfl/fl mice. Consequently, Prx1-Cre;KLfl/fl mice had significantly lower serum FGF23 and parathyroid hormone levels, and higher renal 1-α-hydroxylase expression, serum 1,25-dihydroxyvitamin D, and calcium levels than KLfl/fl mice. These results were confirmed in two independent models of renal failure, adenine diet induced and 5/6 nephrectomy. Moreover, FGF23-treated bone cells required Klotho to increase FGF23 mRNA and ERK phosphorylation. In summary, our novel findings show that Klotho in bone is crucial for inducing FGF23 production upon renal failure. We propose the presence of an autocrine feedback loop in which Klotho senses the need for FGF23.-Kaludjerovic, J., Komaba, H., Sato, T., Erben, R. G., Baron, R., Olauson, H., Larsson, T. E., Lanske, B. Klotho expression in long bones regulates FGF23 production during renal failure.


Asunto(s)
Factores de Crecimiento de Fibroblastos/metabolismo , Glucuronidasa/metabolismo , Riñón/metabolismo , Insuficiencia Renal/metabolismo , Animales , Huesos/metabolismo , Calcitriol/metabolismo , Calcio/metabolismo , Factor-23 de Crecimiento de Fibroblastos , Glucuronidasa/deficiencia , Proteínas Klotho , Ratones Noqueados , Osteoblastos/metabolismo , Hormona Paratiroidea/genética , Fosfatos/metabolismo
19.
J Am Soc Nephrol ; 28(4): 1073-1078, 2017 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27799484

RESUMEN

Tight control of extracellular and intracellular inorganic phosphate (Pi) levels is critical to most biochemical and physiologic processes. Urinary Pi is freely filtered at the kidney glomerulus and is reabsorbed in the renal tubule by the action of the apical sodium-dependent phosphate transporters, NaPi-IIa/NaPi-IIc/Pit2. However, the molecular identity of the protein(s) participating in the basolateral Pi efflux remains unknown. Evidence has suggested that xenotropic and polytropic retroviral receptor 1 (XPR1) might be involved in this process. Here, we show that conditional inactivation of Xpr1 in the renal tubule in mice resulted in impaired renal Pi reabsorption. Analysis of Pi transport in primary cultures of proximal tubular cells or in freshly isolated renal tubules revealed that this Xpr1 deficiency significantly affected Pi efflux. Further, mice with conditional inactivation of Xpr1 in the renal tubule exhibited generalized proximal tubular dysfunction indicative of Fanconi syndrome, characterized by glycosuria, aminoaciduria, calciuria, and albuminuria. Dramatic alterations in the renal transcriptome, including a significant reduction in NaPi-IIa/NaPi-IIc expression, accompanied these functional changes. Additionally, Xpr1-deficient mice developed hypophosphatemic rickets secondary to renal dysfunction. These results identify XPR1 as a major regulator of Pi homeostasis and as a potential therapeutic target in bone and kidney disorders.


Asunto(s)
Síndrome de Fanconi/etiología , Nefronas , Receptores Acoplados a Proteínas G/fisiología , Receptores Virales/fisiología , Raquitismo Hipofosfatémico/etiología , Animales , Femenino , Masculino , Ratones , Receptor de Retrovirus Xenotrópico y Politrópico
20.
Kidney Int ; 92(3): 599-611, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28396120

RESUMEN

Osteocytes within the mineralized bone matrix control bone remodeling by regulating osteoblast and osteoclast activity. Osteocytes express the aging suppressor Klotho, but the functional role of this protein in skeletal homeostasis is unknown. Here we identify Klotho expression in osteocytes as a potent regulator of bone formation and bone mass. Targeted deletion of Klotho from osteocytes led to a striking increase in bone formation and bone volume coupled with enhanced osteoblast activity, in sharp contrast to what is observed in Klotho hypomorphic (kl/kl) mice. Conversely, overexpression of Klotho in cultured osteoblastic cells inhibited mineralization and osteogenic activity during osteocyte differentiation. Further, the induction of chronic kidney disease with high-turnover renal osteodystrophy led to downregulation of Klotho in bone cells. This appeared to offset the skeletal impact of osteocyte-targeted Klotho deletion. Thus, our findings establish a key role of osteocyte-expressed Klotho in regulating bone metabolism and indicate a new mechanism by which osteocytes control bone formation.


Asunto(s)
Envejecimiento/metabolismo , Trastorno Mineral y Óseo Asociado a la Enfermedad Renal Crónica/patología , Glucuronidasa/metabolismo , Osteocitos/metabolismo , Osteogénesis/fisiología , Animales , Densidad Ósea , Huesos/metabolismo , Huesos/patología , Diferenciación Celular , Modelos Animales de Enfermedad , Regulación hacia Abajo , Femenino , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/metabolismo , Glucuronidasa/genética , Humanos , Inmunohistoquímica , Proteínas Klotho , Ratones , Ratones Noqueados , Osteoblastos/fisiología , Osteoclastos/fisiología , Cultivo Primario de Células , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA