Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 201
Filtrar
Más filtros

Intervalo de año de publicación
1.
Calcif Tissue Int ; 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-39085428

RESUMEN

Bone formation is tightly modulated by genetically encoded molecular proteins that interact to regulate cellular differentiation and secretion of bony matrix. Many transcription factors are known to coordinate these events by controlling gene transcription within networks. However, not all factors involved are known. Here, we identified a novel function for Zinc Finger Homeobox 3 (Zfhx3), a gene encoding a transcription factor, as a regulator of bone metabolism. We knocked out Zfhx3 conditionally in mice in either chondrocytes or osteoblasts and characterized their bones by micro-CT in 12-week-old mice. We observed a negative effect in linear bone growth in both knockout mice but reduced bone mass only in mice with Zfhx3 deleted in osteoblasts. Loss of Zfhx3 expression in osteoblasts affected trabecular bone mass in femurs and vertebrae in both sexes but influenced cortical bone volume fraction only in females. Moreover, transcriptional analysis of femoral bones in osteoblast Zfhx3 conditional knockout mice revealed a reduced expression of osteoblast genes, and histological evaluation of trabecular bones suggests that Zfhx3 causes changes in bone formation and not resorption. The loss of Zfhx3 causes reductions in trabecular bone area and osteoid volume, but no changes in the expression of osteoclast differentiation markers or number of TRAP stained osteoclasts. These studies introduce Zfhx3 as a relevant factor toward understanding gene regulatory networks that control bone formation and development of peak bone mass.

2.
Am J Physiol Endocrinol Metab ; 322(6): E528-E539, 2022 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-35466691

RESUMEN

T-cell-like factor (TCF)7l2, a key effector of canonical Wnt signaling, is highly expressed in bone but nothing is known about its role in regulating osteoblast function. To test this, we generated mice with conditional disruption of Tcf7l2 gene in osteoblast lineages using Tcf7l2 floxed and Col1α2-Cre mice. Skeletal parameters were evaluated using heterozygous conditional knockdown (HCKD) mice since homozygous conditional knockout died during pregnancy or immediately after birth. At 5 wk of age, trabecular bone mass of long bones was reduced by 35% as measured by microcomputed tomography (µCT). Histology data showed a 42% reduction in femur trabecular bone mass caused by reduced bone formation. Knockdown of Tcf7l2 expression in osteoblasts decreased proliferation and differentiation by 20%-40%. Expression levels of genes (Hif1α, Vegf, and ß-catenin) targeted by TCF7L2 were decreased by 50% in Tcf7l2-deficient osteoblasts and bones of HCKD mice. We found that the Hif1α gene promoter contained multiple putative TCF7L2 motifs and stabilization of HIF1α protein levels rescued expression of TCF7L2 target genes and alkaline phosphatase (ALP) activity in Tcf7l2-deficient osteoblasts. Furthermore, Tcf7l2 overexpression increased proliferation in the presence of canonical Wnt3a that was not affected by ß-catenin inhibitor providing evidence for a noncanonical signaling in mediating TCF7L2 effects. Tcf7l2 expression was increased in response to mechanical strain (MS) in vitro and in vivo, and disruption of Tcf7l2 expression in osteoblasts reduced MS-induced ALP activity by 35%. We conclude that Tcf7l2, a mechanoresponsive gene, is an important regulator of osteoblast function acting, in part, via hypoxia signaling.NEW & NOTEWORTHY TCF7L2 is expressed by bone but it was not known whether TCF7L2 expression influenced bone development. By using a mouse model with conditional disruption of Tcf7l2 in osteoblast lineage cells, we have demonstrated for the first time, that TCF7L2 plays an important role in regulating osteoblasts via a noncanonical pathway.


Asunto(s)
Osteoblastos , Proteína 2 Similar al Factor de Transcripción 7 , beta Catenina , Animales , Diferenciación Celular/fisiología , Hipoxia de la Célula , Línea Celular , Ratones , Ratones Noqueados , Osteoblastos/citología , Osteoblastos/metabolismo , Proteína 2 Similar al Factor de Transcripción 7/biosíntesis , Proteína 2 Similar al Factor de Transcripción 7/genética , Proteína 2 Similar al Factor de Transcripción 7/metabolismo , Vía de Señalización Wnt , Microtomografía por Rayos X , beta Catenina/metabolismo
3.
Development ; 144(11): 2009-2020, 2017 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-28455377

RESUMEN

Lmx1b is a homeodomain transcription factor responsible for limb dorsalization. Despite striking double-ventral (loss-of-function) and double-dorsal (gain-of-function) limb phenotypes, no direct gene targets in the limb have been confirmed. To determine direct targets, we performed a chromatin immunoprecipitation against Lmx1b in mouse limbs at embryonic day 12.5 followed by next-generation sequencing (ChIP-seq). Nearly 84% (n=617) of the Lmx1b-bound genomic intervals (LBIs) identified overlap with chromatin regulatory marks indicative of potential cis-regulatory modules (PCRMs). In addition, 73 LBIs mapped to CRMs that are known to be active during limb development. We compared Lmx1b-bound PCRMs with genes regulated by Lmx1b and found 292 PCRMs within 1 Mb of 254 Lmx1b-regulated genes. Gene ontological analysis suggests that Lmx1b targets extracellular matrix production, bone/joint formation, axonal guidance, vascular development, cell proliferation and cell movement. We validated the functional activity of a PCRM associated with joint-related Gdf5 that provides a mechanism for Lmx1b-mediated joint modification and a PCRM associated with Lmx1b that suggests a role in autoregulation. This is the first report to describe genome-wide Lmx1b binding during limb development, directly linking Lmx1b to targets that accomplish limb dorsalization.


Asunto(s)
Tipificación del Cuerpo/genética , Extremidades/embriología , Proteínas con Homeodominio LIM/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos/genética , Factores de Transcripción/metabolismo , Animales , Pollos , Inmunoprecipitación de Cromatina , Secuencia Conservada/genética , Elementos de Facilitación Genéticos/genética , Regulación del Desarrollo de la Expresión Génica , Genoma , Factor 5 de Diferenciación de Crecimiento/genética , Factor 5 de Diferenciación de Crecimiento/metabolismo , Proteínas con Homeodominio LIM/genética , Ratones Endogámicos C57BL , Modelos Biológicos , Reproducibilidad de los Resultados , Análisis de Secuencia de ARN , Factores de Transcripción/genética
4.
J Cell Biochem ; 120(8): 13974-13984, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30977156

RESUMEN

Chemokines are secreted by a wide variety of cells; their functions are dependent on the binding to their chemokine receptors (CCRs) which induce directed chemotaxis in nearby responsive cells. Chemokines and their receptors can be induced under several different conditions. Based on data from clinical studies showing an increased expression of chemokine receptor 3 (CCR3) in circulating monocytes of human subjects with lower bone mineral density (BMD) as compared to those with high BMD, we predicted a role for CCR3 in the development of peak bone mass. We, therefore, first evaluated the expression pattern of Ccr3 in bone cells, in comparison to other CCRs, that have common ligands with CCR3. While Ccr1 and Ccr3 messenger RNA (mRNA) levels increased during both RANKL-induced osteoclast differentiation and AA-induced osteoblast differentiation, the levels of Ccr5 mRNA only increased during osteoblast differentiation. To examine if CCR3 influences osteoclast and/or osteoblast differentiation, we evaluated the consequence of blocking CCR3 function using neutralizing antibody on the expression of osteoclast and osteoblast differentiation markers. Treatment with CCR3 neutralizing antibody increased mRNA levels of Trap and cathepsin K in osteoclasts and osteocalcin in osteoblasts compared to cells treated with control IgG. Based on these in vitro findings, we next assessed the role of CCR3 in vivo by evaluating the skeletal phenotypes of Ccr3 knockout and corresponding control littermate mice. Disruption of CCR3 resulted in a significant increase in femur areal BMD at 5 and 8 weeks of age by dual-energy X-ray absorptiometry. Micro-CT analysis revealed a 25% increase in trabecular bone mass at 10 weeks of age caused by corresponding changes in trabecular number and thickness compared to wild type mice. Based on our findings, we conclude that disruption of CCR3 function favors bone mass accumulation, in part via enhancement of bone metabolism. Understanding the molecular pathways through which CCR3 acts to regulate osteoclast and osteoblast functions could lead to new therapeutic approaches to prevent inflammation-induced bone loss.


Asunto(s)
Hueso Esponjoso/anatomía & histología , Hueso Esponjoso/metabolismo , Receptores CCR3/metabolismo , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Anticuerpos Neutralizantes/farmacología , Ácido Ascórbico/farmacología , Biomarcadores/metabolismo , Peso Corporal/efectos de los fármacos , Hueso Esponjoso/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Femenino , Fémur/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Ratones Noqueados , Tamaño de los Órganos/efectos de los fármacos , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteoclastos/citología , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Fenotipo , Ligando RANK/farmacología , Células RAW 264.7 , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores CCR3/genética
5.
Histochem Cell Biol ; 151(1): 43-55, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30250975

RESUMEN

To identify the repertoire of ephrin genes that might regulate endochondral bone fracture repair, we examined changes in ephrin ligand and receptor (Eph) gene expression in fracture callus tissues during bone fracture healing. Ephrin and Eph proteins were then localized in the fracture callus tissues present when changes in gene expression were observed. Ephrin gene expression was widespread in fracture tissues, but the repertoire of ephrin genes with significant changes in expression that might suggest a regulatory role in fracture callus development was restricted to the ephrin A family members Epha4, Epha5 and the ephrin B family member Efnb1. After 3 weeks of healing, Epha4 fracture expression was downregulated from 1.3- to 0.8-fold and Epha5 fracture expression was upregulated from 1.2- to 1.5-fold of intact contralateral femur expression, respectively. Efnb1 expression was downregulated from 1.5- to 1.2-fold after 2 weeks post-fracture. These ephrin proteins were localized to fracture callus prehypertrophic chondrocytes and osteoblasts, as well as to the periosteum and fibrous tissues. The observed positive correlation between mRNA levels of EfnB1 with Col10 and Epha5 with Bglap, together with colocalized expression with their respective proteins, suggest that EfnB1 is a positive mediator of prehypertrophic chondrocyte development and that Epha5 contributes to osteoblast-mediated mineralization of fracture callus. In contrast, mRNA levels of Epha4 and Efnb1 correlated negatively with Bglap, thus suggesting a negative role for these two ephrin family members in mature osteoblast functions. Given the number of family members and widespread expression of the ephrins, a characterization of changes in ephrin gene expression provides a basis for identifying ephrin family members that might regulate the molecular pathways of bone fracture repair. This approach suggests that a highly restricted repertoire of ephrins, EfnB1 and EphA5, are the major mediators of fracture callus cartilage hypertrophy and ossification, respectively, and proposes candidates for additional functional study and eventual therapeutic application.


Asunto(s)
Huesos/metabolismo , Efrinas/genética , Osteogénesis/genética , Animales , Huesos/patología , Efrinas/metabolismo , Perfilación de la Expresión Génica , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
6.
Immunology ; 155(1): 85-98, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29574762

RESUMEN

In the past there have been a multitude of studies that ardently support the role of arginase II (Arg II) in vascular and endothelial disorders; however, the regulation and function of Arg II in autoimmune diseases has thus far remained unclear. Here we report that a global Arg II null mutation in mice suppressed experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis. During EAE, both Arg I and Arg II were induced in spinal cords, but only Arg II was induced in spleens and splenic dendritic cells (DCs). DC activation by lipopolysaccharide (LPS), CD40L or TLR8 agonist significantly enhanced Arg II expression without affecting Arg I expression. Conversely, DC differentiating cytokines [IL-4 and granulocyte macrophage-colony-stimulating factor (GM-CSF)] yielded opposite effects. In addition, Arg I and Arg II were regulated differentially during Th1 and Th17 cell polarization. Arg II deficiency in mice delayed EAE onset, ameliorated clinical symptoms and reduced myelin loss, accompanied by a remarkable reduction in the EAE-induced spinal cord expression of Th17 cell markers (IL-17 and RORγt). The abundance of Th17 cells and IL-23+ cells in relevant draining lymph nodes was significantly reduced in Arg II knockout mice. In activated DCs, Arg II deficiency significantly suppressed the expression of Th17-differentiating cytokines IL-23 and IL-6. Interestingly, Arg II deficiency did not lead to any compensatory increase in Arg I expression in vivo and in vitro. In conclusion, Arg II was identified as a factor promoting EAE likely via an Arg I-independent mechanism. Arg II may promote EAE by enhancing DC production of Th17-differentiating cytokines. Specific inhibition of Arg II could be a potential therapy for multiple sclerosis.


Asunto(s)
Arginasa/genética , Encefalomielitis Autoinmune Experimental/genética , Animales , Arginasa/inmunología , Células Dendríticas/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Perfilación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Reacción en Cadena en Tiempo Real de la Polimerasa
7.
J Cell Biochem ; 119(12): 10351-10357, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30136304

RESUMEN

Mice with disruption of Lrrk1 and patients with nonfunctional mutant Lrrk1 exhibit severe osteopetrosis phenotypes because of osteoclast cytoskeletal dysfunction. To understand how Lrrk1 regulates osteoclast function by modulating cytoskeleton rearrangement, we examined the proteins that are differentially phosphorylated in wild-type mice and Lrrk1-deficient osteoclasts by metal affinity purification coupled liquid chromatography/mass spectrometry (LC/MS) analyses. One of the candidates that we identified by LC/MS is L-plastin, an actin bundling protein. We found that phosphorylation of L-plastin at serine (Ser) residues 5 was present in wild-type osteoclasts but not in Lrrk1-deficient cells. Western blot analyses with antibodies specific for Ser5 phosphorylated L-plastin confirmed the reduced L-plastin Ser5 phosphorylation in Lrrk1 knockout (KO) osteoclasts. micro computed tomography (Micro-CT) analyses revealed that the trabecular bone volume of the distal femur was increased by 27% in the 16 to 21-week-old L-plastin KO females as compared with the wild-type control mice. The ratio of bone volume to tissue volume and connectivity density were increased by 44% and 47% (both P < 0.05), respectively, in L-plastin KO mice. Our data suggest that targeted disruption of L-plastin increases trabecular bone volume, and phosphorylation of Ser5 in L-plastin in the Lrrk1 signaling pathway may in part contribute to actin assembly in mature osteoclasts.


Asunto(s)
Actinas/genética , Glicoproteínas de Membrana/genética , Proteínas de Microfilamentos/genética , Osteopetrosis/genética , Proteínas Serina-Treonina Quinasas/genética , Animales , Hueso Esponjoso/crecimiento & desarrollo , Hueso Esponjoso/metabolismo , Citoesqueleto/genética , Humanos , Ratones , Ratones Noqueados , Osteoclastos/metabolismo , Osteoclastos/patología , Osteopetrosis/patología , Fosforilación/genética , Unión Proteica , Proteínas Serina-Treonina Quinasas/deficiencia , Serina/genética , Transducción de Señal/genética
8.
BMC Nephrol ; 19(1): 24, 2018 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-29394885

RESUMEN

BACKGROUND: Changes in mineral metabolism and bone structure develop early in the course of chronic kidney disease and at end-stage are associated with increased risk of fragility fractures. The disruption of phosphorus homeostasis leads to secondary hyperparathyroidism, a common complication of chronic kidney disease. However, the molecular pathways by which high phosphorus influences bone metabolism in the early stages of the disease are not completely understood. We investigated the effects of a high phosphorus diet on bone and mineral metabolism using a 5/6 nephrectomy model of chronic kidney disease. METHODS: Four-week old rats were randomly assigned into groups: 1) Control with standard diet, 2) Nephrectomy with standard rodent diet, and 3) Nephrectomy with high phosphorus diet. Rats underwent in vivo imaging at baseline, day 14, and day 28, followed by ex vivo imaging. RESULTS: Cortical bone density at the femoral mid-diaphysis was reduced in nephrectomy-control and nephrectomy-high phosphorus compared to control rats. In contrast, trabecular bone mass was reduced at both the lumbar vertebrae and the femoral secondary spongiosa in nephrectomy-high phosphorus but not in nephrectomy-control. Reduced trabecular bone volume adjusted for tissue volume was caused by changes in trabecular number and separation at day 35. Histomorphometry revealed increased bone resorption in tibial secondary spongiosa in nephrectomy-control. High phosphorus diet-induced changes in bone microstructure were accompanied by increased serum parathyroid hormone and fibroblast growth factor 23 levels. CONCLUSION: Our study demonstrates that changes in mineral metabolism and hormonal dysfunction contribute to trabecular and cortical bone changes in this model of early chronic kidney disease.


Asunto(s)
Hueso Esponjoso/patología , Hueso Cortical/patología , Hiperparatiroidismo Secundario/patología , Insuficiencia Renal/patología , Animales , Hueso Esponjoso/metabolismo , Hueso Cortical/metabolismo , Fémur/metabolismo , Fémur/patología , Hiperparatiroidismo Secundario/metabolismo , Vértebras Lumbares/metabolismo , Vértebras Lumbares/patología , Masculino , Ratas , Ratas Sprague-Dawley , Insuficiencia Renal/metabolismo
9.
J Med Genet ; 53(8): 568-74, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27055475

RESUMEN

BACKGROUND: Osteosclerotic metaphyseal dysplasia (OSMD) is a unique form of osteopetrosis characterised by severe osteosclerosis localised to the bone ends. The mode of inheritance is autosomal recessive. Its genetic basis is not known. OBJECTIVE: To identify the disease gene for OSMD. METHODS AND RESULTS: By whole exome sequencing in a boy with OSMD, we identified a homozygous 7 bp deletion (c.5938_5944delGAGTGGT) in the LRRK1 gene. His skeletal phenotype recapitulated that seen in the Lrrk1-deficient mouse. The shared skeletal hallmarks included severe sclerosis in the undermodelled metaphyses and epiphyseal margins of the tubular bones, costal ends, vertebral endplates and margins of the flat bones. The deletion is predicted to result in an elongated LRRK1 protein (p.E1980Afs*66) that lacks a part of its WD40 domains. In vitro functional studies using osteoclasts from Lrrk1-deficient mice showed that the deletion was a loss of function mutation. Genetic analysis of LRRK1 in two unrelated patients with OSMD suggested that OSMD is a genetically heterogeneous condition. CONCLUSIONS: This is the first study to identify the causative gene of OSMD. Our study provides evidence that LRRK1 plays a critical role in the regulation of bone mass in humans.


Asunto(s)
Mutación/genética , Osteocondrodisplasias/genética , Osteosclerosis/genética , Proteínas Serina-Treonina Quinasas/genética , Animales , Huesos/patología , Preescolar , Homocigoto , Humanos , Masculino , Ratones , Osteoclastos/patología , Osteopetrosis/genética
10.
Am J Physiol Endocrinol Metab ; 311(4): E772-E780, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27600824

RESUMEN

Leucine-rich repeat kinase-1 (Lrrk1) consists of ankyrin repeats (ANK), leucine-rich repeats (LRR), a GTPase-like domain of Roc (ROC), a COR domain, a serine/threonine kinase domain (KD), and WD40 repeats (WD40). Previous studies have revealed that knockout (KO) of Lrrk1 in mice causes severe osteopetrosis, and a human mutation of Lrrk1 leads to osteosclerotic metaphysial dysplasia. The molecular mechanism by which Lrrk1 regulates osteoclast function is unknown. In this study, we generated a series of Lrrk1 mutants and evaluated their ability to rescue defective bone resorption in Lrrk1-deficient osteoclasts by use of pit formation assays. Overexpression of Lrrk1 or LRR-truncated Lrrk1, but not ANK-truncated Lrrk1, WD40-truncated Lrrk1, Lrrk1-KD, or K651A mutant Lrrk1, rescued bone resorption function of Lrrk1 KO osteoclasts. We next examined whether RAC1/Cdc42 small GTPases are direct substrates of Lrrk1 in osteoclasts. Western blot and pull-down assays revealed that Lrrk1 deficiency in osteoclasts resulted in reduced phosphorylation and activation of RAC1/Cdc42. In vitro kinase assays confirmed that recombinant Lrrk1 phosphorylated RAC1-GST protein, and immunoprecipitation showed that the interaction of Lrrk1 with RAC1 occurred within 10 min after RANKL treatment. Overexpression of constitutively active Q61L RAC1 partially rescued the resorptive function of Lrrk1-deficient osteoclasts. Furthermore, lack of Lrrk1 in osteoclasts led to reduced autophosphorylation of p21 protein-activated kinase-1 at Ser144, catalyzed by RAC1/Cdc42 binding and activation. Our data indicate that Lrrk1 regulates osteoclast function by directly modulating phosphorylation and activation of small GTPase RAC1/Cdc42 and that its function depends on ANK, ROC, WD40, and kinase domains.


Asunto(s)
Proteínas Activadoras de GTPasa/metabolismo , Neuropéptidos/metabolismo , Osteoclastos/fisiología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína de Unión al GTP rac1/metabolismo , Animales , Repetición de Anquirina , Resorción Ósea/genética , Masculino , Ratones , Ratones Noqueados , Mutación/genética , Fosforilación , Proteínas Serina-Treonina Quinasas/deficiencia , Ligando RANK/farmacología , Células RAW 264.7
11.
Am J Physiol Endocrinol Metab ; 310(10): E846-54, 2016 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-27026086

RESUMEN

Thyroid hormone (TH) action is mediated through two nuclear TH receptors, THRα and THRß. Although the role of THRα is well established in bone, less is known about the relevance of THRß-mediated signaling in bone development. On ther basis of our recent finding that TH signaling is essential for initiation and formation of secondary ossification center, we evaluated the role of THRs in mediating TH effects on epiphysial bone formation. Two-day treatment of TH-deficient Tshr(-/-) mice with TH increased THRß1 mRNA level 3.4-fold at day 7 but had no effect on THRα1 mRNA level at the proximal tibia epiphysis. Treatment of serum-free cultures of tibias from 3-day-old mice with T3 increased THRß1 expression 2.1- and 13-fold, respectively, at 24 and 72 h. Ten-day treatment of Tshr(-/-) newborns (days 5-14) with THRß1 agonist GC1 at 0.2 or 2.0 µg/day increased BV/TV at day 21 by 225 and 263%, respectively, compared with vehicle treatment. Two-day treatment with GC1 (0.2 µg/day) increased expression levels of Indian hedgehog (Ihh) 100-fold, osterix 15-fold, and osteocalcin 59-fold compared with vehicle at day 7 in the proximal tibia epiphysis. Gel mobility shift assay demonstrated that a putative TH response element in the distal promoter of mouse Ihh gene interacted with THRß1. GC1 treatment (1 nM) increased Ihh distal promoter activity 20-fold after 48 h in chondroctyes. Our data suggest a novel role for THRß1 in secondary ossification at the epiphysis that involves transcriptional upregulation of Ihh gene.


Asunto(s)
Epífisis/metabolismo , Proteínas Hedgehog/genética , Osteogénesis/genética , ARN Mensajero/metabolismo , Receptores beta de Hormona Tiroidea/genética , Tibia/metabolismo , Animales , Desarrollo Óseo/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , ARN Mensajero/efectos de los fármacos , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Tirotropina/genética , Transducción de Señal , Receptores alfa de Hormona Tiroidea/genética , Receptores beta de Hormona Tiroidea/agonistas , Tiroxina/farmacología , Triyodotironina/farmacología , Regulación hacia Arriba
12.
Calcif Tissue Int ; 99(4): 435-42, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27312083

RESUMEN

Studies involving human genetic mutations and mutant mouse models have provided irrevocable evidence for a key role for thyroid hormones (THs) in the regulation of skeletal growth. While T3 binds to TH receptors with higher affinity than T4, T4 occupied TH receptors have also been reported in the nucleus under euthyroid conditions raising the possibility that T4 bound nuclear receptors may be biologically relevant in thyroid syndromes with elevated free T4 and reduced T3 levels. We, therefore, evaluated the direct effects of T4, T3, and their metabolites (rT3 and T2) in stimulating osteoblast differentiation using MC3T3-E1 preosteoblasts which do not produce detectable levels of deiodinases. Under serum-free conditions, a 24-h treatment of MC3T3-E1 cells with THs and their metabolites caused a dose-dependent increase in the expression of osteoblast differentiation markers, osterix, and osteocalcin. Circulating concentrations of T3 (~1 ng/ml) and T4 (~30 ng/ml) showed similar potency in stimulating osteoblast differentiation marker expression, while rT3 and T2 were less potent than T3 and T4. Moreover, T3 and T4 treatments elevated the IGF-1 mRNA level suggesting the involvement of IGF-1 signaling in the TH regulation of osteoblast differentiation. We conclude that an elevated T4 level in the absence of T3 may exert stimulatory effects on osteoblast differentiation. The establishment of cell-specific effects of T4 on osteoblasts may provide a strategy to generate T4 mimics that exert skeletal specific effects without the confounding T3 effects on other tissues.


Asunto(s)
Diferenciación Celular , Osteoblastos/citología , Tiroxina/sangre , Triyodotironina/sangre , Células 3T3 , Fosfatasa Alcalina/metabolismo , Animales , Huesos/metabolismo , Línea Celular , Modelos Animales de Enfermedad , Homeostasis , Ratones , Osteoblastos/efectos de los fármacos , Osteocalcina/sangre , Osteogénesis , Transducción de Señal
13.
Physiol Genomics ; 47(2): 33-43, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25492928

RESUMEN

In this study, we evaluated the role of the microRNA (miR)17-92 cluster in osteoblast lineage cells using a Cre-loxP approach in which Cre expression is driven by the entire regulatory region of the type I collagen α2 gene. Conditional knockout (cKO) mice showed a 13-34% reduction in total body bone mineral content and area with little or no change in bone mineral density (BMD) by DXA at 2, 4, and 8 wk in both sexes. Micro-CT analyses of the femur revealed an 8% reduction in length and 25-27% reduction in total volume at the diaphyseal and metaphyseal sites. Neither cortical nor trabecular volumetric BMD was different in the cKO mice. Bone strength (maximum load) was reduced by 10% with no change in bone toughness. Quantitative histomorphometric analyses revealed a 28% reduction in the periosteal bone formation rate and in the mineral apposition rate but with no change in the resorbing surface. Expression levels of periostin, Elk3, Runx2 genes that are targeted by miRs from the cluster were decreased by 25-30% in the bones of cKO mice. To determine the contribution of the miR17-92 cluster to the mechanical strain effect on periosteal bone formation, we subjected cKO and control mice to 2 wk of mechanical loading by four-point bending. We found that the periosteal bone response to mechanical strain was significantly reduced in the cKO mice. We conclude that the miR17-92 cluster expressed in type I collagen-producing cells is a key regulator of periosteal bone formation in mice.


Asunto(s)
Colágeno Tipo I/metabolismo , MicroARNs/genética , Familia de Multigenes , Osteogénesis/genética , Animales , Femenino , Fémur/fisiología , Regulación de la Expresión Génica , Factor I del Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Ratones Noqueados , Ratones Transgénicos , Osteoblastos , Osteogénesis/fisiología , Condicionamiento Físico Animal , Reproducibilidad de los Resultados , Tibia/fisiología
14.
J Cell Biochem ; 116(9): 1932-8, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25727884

RESUMEN

RE1-silencing transcription factor (Rest) has been identified as a master negative regulator of neuronal differentiation. Nothing is known about Rest function in bone cells. In this study, we examined the Rest expression levels and role during osteoblast differentiation. We found that Rest is abundantly expressed in bone marrow stromal cells, calvarial osteoblasts, and MC3T3-E1 osteoblasts. Treatment of primary osteoblasts with ascorbic acid (AA) down regulated Rest mRNA expression at an early stage, but not in later stages of differentiation. Consistent with treatment of primary cultures, AA treatment of MC3T3-E1 cells significantly reduced Rest protein expression at day 3 and at day 8 after initiation of osteoblast differentiation. Treatment of bone marrow stromal cells with BMP-2 and dexamethasone, but not IGF-I for 3 days greatly decreased Rest mRNA expression. To test the function of Rest during osteoblast differentiation, Rest expression was knocked down in MC3T3-E1 cell subclones segregated on the basis of ALP activity (differentiation status) using lentivirus expressing shRNA against Rest. An 80% knockdown of Rest expression decreased Osterix (Osx) expression by 52-57% and as a result, increased both basal and AA induced ALP expression and activity in the subclone that expresses low basal level of ALP (undifferentiated). By contrast, a 98% knockdown of Rest expression in cells that express high basal levels of ALP (differentiated cells) caused a significant reduction in Osx expression, basal and AA induced ALP expression and activity. These data suggest that Rest regulates early osteoblast differentiation via modulating Rest expression that is independent of Osx expression.


Asunto(s)
Osteoblastos/fisiología , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Células 3T3 , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Ácido Ascórbico/farmacología , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Ratones , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteogénesis , Cráneo/citología , Cráneo/metabolismo , Factor de Transcripción Sp7 , Células del Estroma/metabolismo , Factores de Transcripción/genética
15.
J Bone Miner Metab ; 32(2): 124-35, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23700285

RESUMEN

Urokinase plasminogen activator (uPA) regulates a proteolytic cascade of extracellular matrix degradation that functions in tissue development and tissue repair. The development and remodeling of the skeletal extracellular matrix during wound healing suggests that uPA might regulate bone development and repair. To determine whether uPA functions regulate bone development and repair, we examined the basal skeletal phenotype and endochondral bone fracture repair in uPA-deficient mice. The skeletal phenotype of uPA knockout mice was compared with that of control mice under basal conditions by dual-energy X-ray absorptiometry and micro-CT analysis, and during femur fracture repair by micro-CT and histological examination of the fracture callus. No effects of uPA gene deficiency were observed in the basal skeletal phenotype of the whole body or the femur. However, uPA gene deficiency resulted in increased fracture callus cartilage abundance during femur fracture repair at 14 days healing. The increase in cartilage corresponded to reduced tartrate-resistant acid phosphatase (TRAP) staining for osteoclasts in the uPA knockout fracture callus at this time, consistent with impaired osteoclast-mediated remodeling of the fracture cartilage. CD31 staining was reduced in the knockout fracture tissues at this time, suggesting that angiogenesis was also reduced. Osteoclasts also colocalized with CD31 expression in the endothelial cells of the fracture tissues during callus remodeling. These results indicate that uPA promotes remodeling of the fracture cartilage by osteoclasts that are associated with angiogenesis and suggest that uPA promotes angiogenesis and remodeling of the fracture cartilage at this time of bone fracture repair.


Asunto(s)
Cartílago/metabolismo , Curación de Fractura/genética , Activador de Plasminógeno de Tipo Uroquinasa/genética , Animales , Cartílago/patología , Regulación de la Expresión Génica , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa
16.
Brain Inj ; 28(2): 244-51, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24295038

RESUMEN

INTRODUCTION: While it is well established that the brain produces hypothalamic hormones and neuropeptides that influence skeletal metabolism, the impact of traumatic brain injury (TBI) on bone is unknown. Based on the recognition from clinical studies that there is an association between TBI and long-term hypothalamic pituitary dysfunction, it was hypothesized that TBI exerts a negative impact on skeletal growth and maintenance. METHODS: To test the hypothesis, this study employed a repetitive weight drop model for TBI. Four impacts were applied for four consecutive days on 5-week old female C57BL/6 J mice. Bone measurements were taken 2 weeks after the first impact. RESULTS: Bone mineral content (BMC), bone area (B area) and bone mineral density (BMD) in the total body were reduced by 14.5%, 9.8% and 5.2%, respectively, in the impacted vs. control mice. There was a 17.1% reduction in total volumetric BMD (vBMD) and a 4.0% reduction in material vBMD in cortical bone. In trabecular bone, there was a 44.0% reduction in BV/TV. Although there was no change in the cross-sectional bone size, the tibial growth plate and the tibia itself were shortened. CONCLUSION: The repetitive animal TBI model produced an immediate, strong negative impact on bone mass acquisition in young mice.


Asunto(s)
Huesos/metabolismo , Lesiones Encefálicas/metabolismo , Osteocalcina/metabolismo , Receptores de la Hormona Hipofisaria/metabolismo , Animales , Densidad Ósea , Desarrollo Óseo , Lesiones Encefálicas/complicaciones , Lesiones Encefálicas/fisiopatología , Modelos Animales de Enfermedad , Femenino , Ratones , Ratones Endogámicos C57BL
17.
Biology (Basel) ; 13(2)2024 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-38392326

RESUMEN

Mice lacking Claudin11 (Cldn11) manifest reduced trabecular bone mass. However, the impact of Cldn11 expression in osteoblasts in vivo remains understudied. Herein, we generated osteoblast-specific transgenic (Tg) mice expressing Cldn11 and characterized their skeletal phenotype. Micro-CT analyses of the distal metaphysis of the femur showed a 50% and a 38% increase in trabecular bone mass in Tg male and female mice, respectively, due to a significant increase in trabecular number and a reduction in trabecular separation. Histomorphometry and serum biomarker studies uncovered that increased trabecular bone mass in Cldn11 Tg mice was the consequence of enhanced bone formation. Accordingly, an abundance of bone formation (Alp, Bsp), but not bone resorption (Ctsk), markers were augmented in the femurs of Cldn11 Tg mice. Since the trabecular bone density is known to inversely correlate with the amount of marrow adipose tissue (MAT), we measured the MAT in osmium-tetroxide-labeled bones by micro-CT scanning. We found 86% less MAT in the proximal tibia of the Tg males. Consistently, the expression levels of the adipogenic markers, adiponectin and leptin, were 50% lower in the femurs of the Tg males. Our data are consistent with the possibility that claudin11 exerts anabolic effects in osteoblastic lineage cells that act via promoting the differentiation of marrow stem cells towards osteoblasts at the expense of adipocytes.

18.
Endocrinology ; 165(8)2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38935021

RESUMEN

Thyroid hormone (TH) plays a crucial role in regulating the functions of both bone and adipose tissue. Given that TH exerts its cholesterol-lowering effects in hepatic tissue through the TH receptor-ß (TRß), we hypothesized that TRß agonist therapy using MGL3196 (MGL) would be effective in treating increased adiposity and bone loss in response to a 12-week high-fat diet (HFD) in adult C57BL/6J mice. Transcriptional and serum profiling revealed that HFD-induced leptin promoted weight gain in both males and females, but MGL only suppressed leptin induction and weight gain in males. In vitro studies suggest that estrogen suppresses MGL activity in adipocytes, indicating that estrogen might interfere with MGL-TRß function. Compared to systemic adiposity, HFD reduced bone mass in male but not female mice. Paradoxically, MGL treatment reversed macroscopic bone mineral density loss in appendicular bones, but micro-CT revealed that MGL exacerbated HFD-induced trabecular bone loss, and reduced bone strength. In studies on the mechanisms for HFD effects on bone, we found that HFD induced Rankl expression in male femurs that was blocked by MGL. By ex vivo assays, we found that RANKL indirectly represses osteoblast lineage allocation of osteoprogenitors by induction of inflammatory cytokines TNFα, IL-1ß, and CCL2. Finally, we found that MGL functions in both systemic adiposity and bone by nongenomic TRß signaling, as HFD-mediated phenotypes were not rescued in TRß147F knockout mice with normal genomic but defective nongenomic TRß signaling. Our findings demonstrate that the negative effects of HFD on body fat and bone phenotypes are impacted by MGL in a gender-specific manner.


Asunto(s)
Dieta Alta en Grasa , Transducción de Señal , Receptores beta de Hormona Tiroidea , Animales , Femenino , Masculino , Ratones , Adipocitos/metabolismo , Adipocitos/efectos de los fármacos , Adiposidad/efectos de los fármacos , Densidad Ósea/efectos de los fármacos , Leptina/metabolismo , Ratones Endogámicos C57BL , Osteoblastos/metabolismo , Osteoblastos/efectos de los fármacos , Ligando RANK/metabolismo , Ligando RANK/genética , Caracteres Sexuales , Factores Sexuales , Transducción de Señal/efectos de los fármacos , Receptores beta de Hormona Tiroidea/metabolismo , Receptores beta de Hormona Tiroidea/genética , Aumento de Peso/efectos de los fármacos
19.
JBMR Plus ; 8(5): ziae034, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38586475

RESUMEN

Mutations in PLEKHM1 cause osteopetrosis in humans and rats. The germline and osteoclast conditional deletions of Plekhm1 gene in mice lead to defective osteoclast bone resorption and increased trabecular bone mass without overt abnormalities in other organs. As an adaptor protein, pleckstrin homology and RUN domain containing M1 (PLEKHM1) interacts with the key lysosome regulator small GTPase RAB7 via its C-terminal RUBICON homologous (RH) domain. In this study, we have conducted a structural-functional study of the PLEKHM1 RH domain and RAB7 interaction in osteoclasts in vitro. The single mutations of the key residues in the Plekhm1 RH predicted from the crystal structure of the RUBICON RH domain and RAB7 interface failed to disrupt the Plekhm1-Rab7 binding, lysosome trafficking, and bone resorption. The compound alanine mutations at Y949-R954 and L1011-I1018 regions decreased Plekhm1 protein stability and Rab7-binding, respectively, thereby attenuated lysosome trafficking and bone resorption in osteoclasts. In contrast, the compound alanine mutations at R1060-Q1068 region were dispensable for Rab7-binding and Plekhm1 function in osteoclasts. These results indicate that the regions spanning Y949-R954 and L1011-I1018 of Plekhm1 RH domain are functionally important for Plekhm1 in osteoclasts and offer the therapeutic targets for blocking bone resorption in treatment of osteoporosis and other metabolic bone diseases.

20.
Physiol Genomics ; 45(19): 917-23, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23943855

RESUMEN

Osterix (Osx) is essential for both intramembranous or endochondral bone formation. Osteoblast-specific ablation of Osx using Col1α1-Cre resulted in osteopenia, because of impaired osteoblast differentiation in adult mice. Since Osx is also known to be expressed in chondrocytes, we evaluated the role of Osx expressed in chondrocytes by examining the skeletal phenotype of mice with conditional disruption of Osx in Col2α1-expressing chondrocytes. Surprisingly, Cre-positive mice that were homozygous for Osx floxed alleles died after birth. Alcian blue and alizarin red staining revealed that the lengths of skeleton, femur, and vertebrae were reduced by 21, 26, and 14% (P < 0.01), respectively, in the knockout (KO) compared with wild-type mice. To determine if haploid insufficiency of Osx in chondrocytes influenced postnatal skeletal growth, we compared skeletal phenotype of floxed heterozygous mice that were Cre-positive or Cre-negative. Body length was reduced by 8% (P < 0.001), and areal BMD of total body, femur, and tibia was reduced by 5, 7, and 8% (P < 0.05), respectively, in mice with conditional disruption of one allele of Osx in chondrocytes. Micro-CT showed reduced cortical volumetric bone mineral density and trabecular bone volume to total volume in the femurs of Osx(flox/+);col2α1-Cre mice. Histological analysis revealed that the impairment of longitudinal growth was associated with disrupted growth plates in the Osx(flox/+);col2α1-Cre mice. Primary chondrocytes isolated from KO embryos showed reduced expression of chondral ossification markers but elevated expression of chondrogenesis markers. Our findings indicate that Osx expressed in chondrocytes regulates bone growth in part by regulating chondrocyte hypertrophy.


Asunto(s)
Desarrollo Óseo/genética , Condrocitos/metabolismo , Condrocitos/patología , Haploinsuficiencia/genética , Factores de Transcripción/genética , Absorciometría de Fotón , Animales , Animales Recién Nacidos , Biomarcadores/metabolismo , Calcificación Fisiológica/genética , Fémur/diagnóstico por imagen , Fémur/patología , Eliminación de Gen , Hipertrofia , Ratones , Ratones Noqueados , Especificidad de Órganos/genética , Factor de Transcripción Sp7
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA