Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
PLoS One ; 14(8): e0220653, 2019.
Article in English | MEDLINE | ID: mdl-31390373

ABSTRACT

Vitamin C (ascorbic acid, AA) is a well-known regulator of bone and cartilage metabolism. However, the mechanisms of AA's action in these tissues are only partly understood. In this study, we confirmed that AA contributes to bone and cartilage metabolism by showing decreased articular cartilage and trabecular bone in AA-deficient spontaneous fracture (sfx) mutant mice. In vitro, we found that AA exerts differential effects on chondrocyte and osteoblast differentiation. Since AA is known to increase levels of 5-hydroxymethylcytosine (5-hmC) and induce DNA demethylation via the ten-eleven translocases (TETs), and since prolyl hydroxylase domain-containing protein 2 (PHD2), a known mediator of AA's effects in these tissues, is part of the same enzyme family as the TETs, we next investigated whether increases in 5-hmC might mediate some of these effects. All TETs and PHDs are expressed in chondrocytes and osteoblasts, and PHD2 is localized in both the cytoplasm and nucleus of the cell, lending plausibility to the hypothesis of altered 5-hmC content in these cells. We found that AA treatment increased levels of 5-hmC in both cell types globally, notably including promoter regions of osteoblast differentiation genes. Furthermore, inhibition of PHD2 decreased 5-hmC levels in chondrocyte differentiation gene promoters, and knockdown of Phd2 in chondrocytes reduced global 5-hmC levels, suggesting for the first time that PHD2 may itself directly mediate increases in 5-hmC in chondrocyte and osteoblast genes. Further investigation of this mechanism could lead to novel therapeutic approaches to treat debilitating diseases such as osteoarthritis and osteoporosis.


Subject(s)
5-Methylcytosine/analogs & derivatives , Ascorbic Acid/pharmacology , Chondrocytes/metabolism , Gene Expression/drug effects , Osteoblasts/metabolism , 5-Methylcytosine/metabolism , Animals , Cell Differentiation/genetics , Cells, Cultured , Hypoxia-Inducible Factor-Proline Dioxygenases/pharmacology , Mice
2.
Sci Rep ; 7(1): 10432, 2017 09 05.
Article in English | MEDLINE | ID: mdl-28874841

ABSTRACT

Endochondral ossification in the diaphysis of long bones has been studied in-depth during fetal development but not postnatally in the epiphysis. Immunohistochemical studies revealed that Sox9 and Col2 expressing immature chondrocytes in the epiphysis transition into prehypertrophic and hypetrophic chondrocytes and finally into osteoblasts expressing Col1 and BSP during postnatal day 7-10, when serum levels of thyroid hormone (TH) rise. Lineage tracing using Rosa-td tomato Col2-Cre-ERT2 mice treated with tamoxifen indicated that the same Col2 expressing chondrocytes expressed prehypertrophic, hypertrophic, and subsequently bone formation markers in a sequential manner in euthyroid but not hypothyroid mice, thus providing evidence that chondrocyte to osteoblast transdifferentiation is TH-dependent. Vascular invasion was apparent at the time of bone formation but not earlier. In vitro studies revealed that TH acting via TRα1 promoted expression of SHH while TRß1 activation increased IHH but inhibited SHH expression. SHH promoted expression of markers of immature chondrocytes but inhibited chondrocyte hypertrophy while IHH promoted chondrocyte hypertrophy. Based on our data, we propose a model in which TH acting through TRα1 and TRß1, respectively, fine tune levels of SHH and IHH and, thereby control the transit of proliferating immature chondrocytes into mature hypertrophic chondrocytes to become osteoblasts at the epiphysis.


Subject(s)
Cell Transdifferentiation , Chondrocytes/cytology , Chondrocytes/metabolism , Epiphyses/physiology , Osteoblasts/cytology , Osteoblasts/metabolism , Osteogenesis , Thyroid Hormones/metabolism , Animals , Biomarkers , Fluorescent Antibody Technique , Gene Expression , Immunohistochemistry , Mice , Mice, Transgenic , Signal Transduction
3.
Development ; 144(9): 1698-1711, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28356311

ABSTRACT

Regulated retinal ganglion cell (RGC) differentiation and axonal guidance is required for a functional visual system. Homeodomain and basic helix-loop-helix transcription factors are required for retinogenesis, as well as patterning, differentiation and maintenance of specific retinal cell types. We hypothesized that Dlx1, Dlx2 and Brn3b homeobox genes function in parallel intrinsic pathways to determine RGC fate and therefore generated Dlx1/Dlx2/Brn3b triple-knockout mice. A more severe retinal phenotype was found in the Dlx1/Dlx2/Brn3b-null retinas than was predicted by combining features of the Brn3b single- and Dlx1/Dlx2 double-knockout retinas, including near total RGC loss with a marked increase in amacrine cells in the ganglion cell layer. Furthermore, we discovered that DLX1 and DLX2 function as direct transcriptional activators of Brn3b expression. Knockdown of Dlx2 expression in primary embryonic retinal cultures and Dlx2 gain of function in utero strongly support that DLX2 is both necessary and sufficient for Brn3b expression in vivo We suggest that ATOH7 specifies RGC-committed progenitors and that Dlx1 and Dlx2 function both downstream of ATOH7 and in parallel, but cooperative, pathways that involve regulation of Brn3b expression to determine RGC fate.


Subject(s)
Cell Differentiation , Homeodomain Proteins/metabolism , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , Transcription Factor Brn-3B/metabolism , Transcription Factors/metabolism , Vertebrates/metabolism , Amacrine Cells/cytology , Amacrine Cells/metabolism , Animals , Apoptosis/genetics , Base Sequence , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Count , Cell Division/genetics , Cell Lineage/genetics , Cell Proliferation , Cells, Cultured , Cholinergic Neurons/cytology , Cholinergic Neurons/metabolism , Electroporation , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Gene Deletion , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Mice, Knockout , Models, Biological , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Promoter Regions, Genetic , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcription Factor Brn-3B/deficiency , Transcription Factors/deficiency
4.
Sci Rep ; 7: 45408, 2017 03 28.
Article in English | MEDLINE | ID: mdl-28349987

ABSTRACT

Based on our findings that PHD2 is a negative regulator of chondrocyte differentiation and that hypoxia signaling is implicated in the pathogenesis of osteoarthritis, we investigated the consequence of disruption of the Phd2 gene in chondrocytes on the articular cartilage phenotype in mice. Immunohistochemistry detected high expression of PHD2 in the superficial zone (SZ), while PHD3 and HIF-1α (target of PHD2) are mainly expressed in the middle-deep zone (MDZ). Conditional deletion of the Phd2 gene (cKO) in chondrocytes accelerated the transition of progenitors to hypertrophic (differentiating) chondrocytes as revealed by reduced SZ thickness, and increased MDZ thickness, as well as increased chondrocyte hypertrophy. Immunohistochemistry further revealed decreased levels of progenitor markers but increased levels of hypertrophy markers in the articular cartilage of the cKO mice. Treatment of primary articular chondrocytes, in vitro, with IOX2, a specific inhibitor of PHD2, promoted articular chondrocyte differentiation. Knockdown of Hif-1α expression in primary articular chondrocytes using lentiviral vectors containing Hif-1α shRNA resulted in reduced expression levels of Vegf, Glut1, Pgk1, and Col10 compared to control shRNA. We conclude that Phd2 is a key regulator of articular cartilage development that acts by inhibiting the differentiation of articular cartilage progenitors via modulating HIF-1α signaling.


Subject(s)
Cartilage, Articular/growth & development , Cell Differentiation/genetics , Chondrocytes/cytology , Chondrogenesis/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Animals , Cartilage, Articular/cytology , Cells, Cultured , Collagen Type X/biosynthesis , Glucose Transporter Type 1/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphoglycerate Kinase/biosynthesis , RNA Interference , RNA, Small Interfering/genetics , Signal Transduction/genetics , Vascular Endothelial Growth Factor A/biosynthesis
5.
Sci Rep ; 6: 35748, 2016 10 24.
Article in English | MEDLINE | ID: mdl-27775044

ABSTRACT

Endochondral ossification plays an important role in the formation of the primary ossification centers (POCs) and secondary ossification centers (SOCs) of mammalian long bones. However, the molecular mechanisms that regulate POC and SOC formation are different. We recently demonstrated that Prolyl Hydroxylase Domain-containing Protein 2 (Phd2) is a key mediator of vitamin C effects on bone. We investigated the role of Phd2 on endochondral ossification of the epiphyses by conditionally deleting the Phd2 gene in osteoblasts and chondrocytes. We found that the deletion of Phd2 in osteoblasts did not cause changes in bone parameters in the proximal tibial epiphyses in 5 week old mice. In contrast, deletion of Phd2 in chondrocytes resulted in increased bone mass and bone formation rate (normalized to tissue volume) in long bone epiphyses, indicating that Phd2 expressed in chondrocytes, but not osteoblasts, negatively regulates secondary ossification of epiphyses. Phd2 deletion in chondrocytes elevated mRNA expression of hypoxia-inducible factor (HIF) signaling molecules including Hif-1α, Hif-2α, Vegfa, Vegfb, and Epo, as well as markers for chondrocyte hypertrophy and mineralization such as Col10, osterix, alkaline phosphatase, and bone sialoprotein. These data suggest that Phd2 expressed in chondrocytes inhibits endochondral ossification at the epiphysis by suppressing HIF signaling pathways.


Subject(s)
Cell Differentiation/physiology , Chondrocytes/metabolism , Chondrocytes/physiology , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Animals , Biomarkers/metabolism , Chondrogenesis/physiology , Erythropoietin/metabolism , Growth Plate/metabolism , Growth Plate/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Osteoblasts/metabolism , Osteoblasts/physiology , Osteogenesis/physiology , RNA, Messenger/metabolism , Signal Transduction , Tibia/metabolism , Tibia/physiology , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor B/metabolism
6.
Calcif Tissue Int ; 99(4): 435-42, 2016 10.
Article in English | MEDLINE | ID: mdl-27312083

ABSTRACT

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.


Subject(s)
Cell Differentiation , Osteoblasts/cytology , Thyroxine/blood , Triiodothyronine/blood , 3T3 Cells , Alkaline Phosphatase/metabolism , Animals , Bone and Bones/metabolism , Cell Line , Disease Models, Animal , Homeostasis , Mice , Osteoblasts/drug effects , Osteocalcin/blood , Osteogenesis , Signal Transduction
7.
Am J Physiol Endocrinol Metab ; 310(10): E846-54, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27026086

ABSTRACT

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.


Subject(s)
Epiphyses/metabolism , Hedgehog Proteins/genetics , Osteogenesis/genetics , RNA, Messenger/metabolism , Thyroid Hormone Receptors beta/genetics , Tibia/metabolism , Animals , Bone Development/genetics , Female , Gene Expression Regulation, Developmental , Hedgehog Proteins/drug effects , Male , Mice , Mice, Knockout , RNA, Messenger/drug effects , Real-Time Polymerase Chain Reaction , Receptors, Thyrotropin/genetics , Signal Transduction , Thyroid Hormone Receptors alpha/genetics , Thyroid Hormone Receptors beta/agonists , Thyroxine/pharmacology , Triiodothyronine/pharmacology , Up-Regulation
8.
Endocrinology ; 157(1): 127-40, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26562260

ABSTRACT

The hypoxic growth plate cartilage requires hypoxia-inducible factor (HIF)-mediated pathways to maintain chondrocyte survival and differentiation. HIF proteins are tightly regulated by prolyl hydroxylase domain-containing protein 2 (Phd2)-mediated proteosomal degradation. We conditionally disrupted the Phd2 gene in chondrocytes by crossing Phd2 floxed mice with type 2 collagen-α1-Cre transgenic mice and found massive increases (>50%) in the trabecular bone mass of long bones and lumbar vertebra of the Phd2 conditional knockout (cKO) mice caused by significant increases in trabecular number and thickness and reductions in trabecular separation. Cortical thickness and tissue mineral density at the femoral middiaphysis of the cKO mice were also significantly increased. Dynamic histomorphometric analyses revealed increased longitudinal length and osteoid surface per bone surface in the primary spongiosa of the cKO mice, suggesting elevated conversion rate from hypertrophic chondrocytes to mineralized bone matrix as well as increased bone formation in the primary spongiosa. In the secondary spongiosa, bone formation measured by mineralizing surface per bone surface and mineral apposition rate were not changed, but resorption was slightly reduced. Increases in the mRNA levels of SRY (sex determining region Y)-box 9, osterix (Osx), type 2 collagen, aggrecan, alkaline phosphatase, bone sialoprotein, vascular endothelial growth factor, erythropoietin, and glycolytic enzymes in the growth plate of cKO mice were detected by quantitative RT-PCR. Immunohistochemistry revealed an increased HIF-1α protein level in the hypertrophic chondrocytes of cKO mice. Infection of chondrocytes isolated from Phd2 floxed mice with adenoviral Cre resulted in similar gene expression patterns as observed in the cKO growth plate chondrocytes. Our findings indicate that Phd2 suppresses endochondral bone formation, in part, via HIF-dependent mechanisms in mice.


Subject(s)
Chondrocytes/metabolism , Chondrogenesis , Gene Expression Regulation, Developmental , Growth Plate/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Osteogenesis , Absorptiometry, Photon , Animals , Bone Density , Cells, Cultured , Chondrocytes/cytology , Crosses, Genetic , Female , Femur/cytology , Femur/diagnostic imaging , Femur/growth & development , Femur/metabolism , Growth Plate/cytology , Growth Plate/diagnostic imaging , Growth Plate/growth & development , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Lumbar Vertebrae/cytology , Lumbar Vertebrae/diagnostic imaging , Lumbar Vertebrae/growth & development , Lumbar Vertebrae/metabolism , Male , Mice, Knockout , Mice, Transgenic , Organ Specificity , Tibia/cytology , Tibia/diagnostic imaging , Tibia/growth & development , Tibia/metabolism , X-Ray Microtomography
9.
ScientificWorldJournal ; 2015: 989536, 2015.
Article in English | MEDLINE | ID: mdl-26167539

ABSTRACT

Dynamic behaviour of cable networks is highly dependent on the installation location, stiffness, and damping of cross-ties. Thus, these are the important design parameters for a cable network. While the effects of the former two on the network response have been investigated to some extent in the past, the impact of cross-tie damping has rarely been addressed. To comprehend our knowledge of mechanics associated with cable networks, in the current study, an analytical model of a cable network will be proposed by taking into account both cross-tie stiffness and damping. In addition, the damping property of main cables in the network will also be considered in the formulation. This would allow exploring not only the effectiveness of a cross-tie design on enhancing the in-plane stiffness of a constituted cable network, but also its energy dissipation capacity. The proposed analytical model will be applied to networks with different configurations. The influence of cross-tie stiffness and damping on the modal response of various types of networks will be investigated by using the corresponding undamped rigid cross-tie network as a reference base. Results will provide valuable information on the selection of cross-tie properties to achieve more effective cable vibration control.

10.
J Cell Biochem ; 116(9): 1932-8, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25727884

ABSTRACT

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.


Subject(s)
Osteoblasts/physiology , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/metabolism , 3T3 Cells , Adaptor Proteins, Signal Transducing/metabolism , Animals , Ascorbic Acid/pharmacology , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Differentiation/drug effects , Cells, Cultured , Gene Expression Regulation, Developmental/drug effects , Membrane Proteins/metabolism , Mice , Osteoblasts/cytology , Osteoblasts/drug effects , Osteogenesis , Skull/cytology , Skull/metabolism , Sp7 Transcription Factor , Stromal Cells/metabolism , Transcription Factors/genetics
11.
J Bone Miner Res ; 29(10): 2262-75, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24753031

ABSTRACT

Thyroid hormones (THs) are known to regulate endochondral ossification during skeletal development via acting directly in chondrocytes and osteoblasts. In this study, we focused on TH effects on the secondary ossification center (SOC) because the time of appearance of SOCs in several species coincides with the time when peak levels of TH are attained. Accordingly, micro-computed tomography (µCT) evaluation of femurs and tibias at day 21 in TH-deficient and control mice revealed that endochondral ossification of SOCs is severely compromised owing to TH deficiency and that TH treatment for 10 days completely rescued this phenotype. Staining of cartilage and bone in the epiphysis revealed that whereas all of the cartilage is converted into bone in the prepubertal control mice, this conversion failed to occur in the TH-deficient mice. Immunohistochemistry studies revealed that TH treatment of thyroid stimulating hormone receptor mutant (Tshr(-/-) ) mice induced expression of Indian hedgehog (Ihh) and Osx in type 2 collagen (Col2)-expressing chondrocytes in the SOC at day 7, which subsequently differentiate into type 10 collagen (Col10)/osteocalcin-expressing chondro/osteoblasts at day 10. Consistent with these data, treatment of tibia cultures from 3-day-old mice with 10 ng/mL TH increased expression of Osx, Col10, alkaline phosphatase (ALP), and osteocalcin in the epiphysis by sixfold to 60-fold. Furthermore, knockdown of the TH-induced increase in Osx expression using lentiviral small hairpin RNA (shRNA) significantly blocked TH-induced ALP and osteocalcin expression in chondrocytes. Treatment of chondrogenic cells with an Ihh inhibitor abolished chondro/osteoblast differentiation and SOC formation. Our findings indicate that TH regulates the SOC initiation and progression via differentiating chondrocytes into bone matrix-producing osteoblasts by stimulating Ihh and Osx expression in chondrocytes.


Subject(s)
Chondrocytes/metabolism , Epiphyses/cytology , Hedgehog Proteins/genetics , Osteogenesis/drug effects , Signal Transduction/genetics , Thyroid Hormones/pharmacology , Transcription Factors/genetics , Animals , Biomarkers/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Chondrocytes/drug effects , Collagen Type II/genetics , Femur/diagnostic imaging , Femur/drug effects , Hedgehog Proteins/metabolism , Mice , Mice, Mutant Strains , Models, Biological , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteocalcin/metabolism , Osteogenesis/genetics , Radiography , Receptors, Thyrotropin/deficiency , Receptors, Thyrotropin/metabolism , Signal Transduction/drug effects , Sp7 Transcription Factor , Thyroid Hormones/deficiency , Tibia/diagnostic imaging , Tibia/drug effects , Transcription Factors/metabolism
12.
J Bone Miner Res ; 29(10): 2276-86, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24753072

ABSTRACT

We have previously shown that the increase in osterix (Osx) expression during osteoblast maturation is dependent on the activity of the prolyl hydroxylase domain-containing protein 2 (Phd2), a key regulator of protein levels of the hypoxia-inducible factor family proteins in many tissues. In this study, we generated conditional Phd2 knockout mice (cKO) in osteoblast lineage cells by crossing floxed Phd2 mice with a Col1α2-iCre line to investigate the function of Phd2 in vivo. The cKO mice developed short stature and premature death at 12 to 14 weeks of age. Bone mineral content, bone area, and bone mineral density were decreased in femurs and tibias, but not vertebrae of the cKO mice compared to WT mice. The total volume (TV), bone volume (BV), and bone volume fraction (BV/TV) in the femoral trabecular bones of cKO mice were significantly decreased. Cross-sectional area of the femoral mid-diaphysis was also reduced in the cKO mice. The reduced bone size and trabecular bone volume in the cKO mice were a result of impaired bone formation but not bone resorption as revealed by dynamic histomorphometric analyses. Bone marrow stromal cells derived from cKO mice formed fewer and smaller nodules when cultured with mineralization medium. Quantitative RT-PCR and immunohistochemistry detected reduced expression of Osx, osteocalcin, and bone sialoprotein in cKO bone cells. These data indicate that Phd2 plays an important role in regulating bone formation in part by modulating expression of Osx and bone formation marker genes.


Subject(s)
Gene Deletion , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Osteogenesis/genetics , Absorptiometry, Photon , Animals , Biomarkers/metabolism , Blood Cell Count , Bone Resorption/blood , Bone Resorption/diagnostic imaging , Bone Resorption/pathology , Calcification, Physiologic , Cell Differentiation , Collagen Type I/metabolism , Femur/diagnostic imaging , Gene Expression Regulation, Developmental , Hematocrit , Hypoxia-Inducible Factor-Proline Dioxygenases/deficiency , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Mice, Knockout , Osteoblasts/pathology , Spine/diagnostic imaging , X-Ray Microtomography
13.
Physiol Genomics ; 45(19): 917-23, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23943855

ABSTRACT

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.


Subject(s)
Bone Development/genetics , Chondrocytes/metabolism , Chondrocytes/pathology , Haploinsufficiency/genetics , Transcription Factors/genetics , Absorptiometry, Photon , Animals , Animals, Newborn , Biomarkers/metabolism , Calcification, Physiologic/genetics , Femur/diagnostic imaging , Femur/pathology , Gene Deletion , Hypertrophy , Mice , Mice, Knockout , Organ Specificity/genetics , Sp7 Transcription Factor
14.
PLoS One ; 8(7): e69051, 2013.
Article in English | MEDLINE | ID: mdl-23874863

ABSTRACT

To test if ephrin B1 overexpression enhances bone mass, we generated transgenic mice overexpressing ephrin B1 under the control of a 3.6 kb rat collagen 1A1 promoter (Col3.6-Tg (efnb1) ). Col3.6-Tg (efnb1) mice express 6-, 12- and 14-fold greater levels of full-length ephrin B1 protein in bone marrow stromal cells, calvarial osteoblasts, and osteoclasts, respectively. The long bones of both genders of Col3.6-Tg (efnb1) mice have increased trabecular bone volume, trabecular number, and trabecular thickness and decreased trabecular separation. Enhanced bone formation and decreased bone resorption contributed to this increase in trabecular bone mass in Col3.6-Tg (efnb1) mice. Consistent with these findings, our in vitro studies showed that overexpression of ephrin B1 increased osteoblast differentiation and mineralization, osterix and collagen 1A1 expression in bone marrow stromal cells. Interaction of ephrin B1 with soluble clustered EphB2-Fc decreased osteoclast precursor differentiation into multinucleated cells. Furthermore, we demonstrated that the mechanical loading-induced increase in EphB2 expression and newly formed bone were significantly greater in the Col3.6-Tg (efnb1) mice than in WT littermate controls. Our findings that overexpression of ephrin B1 in bone cells enhances bone mass and promotes a skeletal anabolic response to mechanical loading suggest that manipulation of ephrin B1 actions in bone may provide a means to sensitize the skeleton to mechanical strain to stimulate new bone formation.


Subject(s)
Bone and Bones/physiology , Ephrin-B1/genetics , Ephrin-B1/metabolism , Osteogenesis/physiology , Animals , Bone Density/genetics , Bone Density/physiology , Bone and Bones/metabolism , Cell Differentiation/genetics , Cell Differentiation/physiology , Collagen/genetics , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/physiology , Mice , Mice, Transgenic , Osteoblasts/metabolism , Osteoblasts/physiology , Osteoclasts/metabolism , Osteoclasts/physiology , Osteogenesis/genetics , Promoter Regions, Genetic , Rats , Sp7 Transcription Factor , Transcription Factors/genetics , Transcription Factors/metabolism
15.
J Bone Miner Res ; 28(9): 1962-74, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23526378

ABSTRACT

To assess the roles of Lrrk1 and Lrrk2, we examined skeletal phenotypes in Lrrk1 and Lrrk2 knockout (KO) mice. Lrrk1 KO mice exhibit severe osteopetrosis caused by dysfunction of multinucleated osteoclasts, reduced bone resorption in endocortical and trabecular regions, and increased bone mineralization. Lrrk1 KO mice have lifelong accumulation of bone and respond normally to the anabolic actions of teriparatide treatment, but are resistant to ovariectomy-induced bone boss. Precursors derived from Lrrk1 KO mice differentiate into multinucleated cells in response to macrophage colony-stimulating factor (M-CSF)/receptor activator of NF-κB ligand (RANKL) treatment, but these cells fail to form peripheral sealing zones and ruffled borders, and fail to resorb bone. The phosphorylation of cellular Rous sarcoma oncogene (c-Src) at Tyr-527 is significantly elevated whereas at Tyr-416 is decreased in Lrrk1-deficient osteoclasts. The defective osteoclast function is partially rescued by overexpression of the constitutively active form of Y527F c-Src. Immunoprecipitation assays in osteoclasts detected a physical interaction of Lrrk1 with C-terminal Src kinase (Csk). Lrrk2 KO mice do not show obvious bone phenotypes. Precursors derived from Lrrk2 KO mice differentiate into functional multinucleated osteoclasts. Our finding of osteopetrosis in Lrrk1 KO mice provides convincing evidence that Lrrk1 plays a critical role in negative regulation of bone mass in part through modulating the c-Src signaling pathway in mice.


Subject(s)
Gene Targeting , Osteopetrosis/enzymology , Osteopetrosis/pathology , Protein Serine-Threonine Kinases/metabolism , Acid Phosphatase/blood , Alkaline Phosphatase/blood , Animals , Bone Density , Bone Resorption/enzymology , Bone Resorption/pathology , Bone Resorption/physiopathology , Bone and Bones/diagnostic imaging , Bone and Bones/enzymology , Bone and Bones/pathology , CSK Tyrosine-Protein Kinase , Female , Isoenzymes/blood , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Mice , Mice, Knockout , Mutant Proteins/metabolism , Osteoclasts/enzymology , Osteoclasts/pathology , Osteogenesis , Osteopetrosis/diagnostic imaging , Osteopetrosis/physiopathology , Ovariectomy , Peptide Fragments/blood , Phenotype , Phosphorylation , Procollagen/blood , Protein Binding , Protein Serine-Threonine Kinases/deficiency , Proto-Oncogene Proteins pp60(c-src)/metabolism , Tartrate-Resistant Acid Phosphatase , X-Ray Microtomography , src-Family Kinases/metabolism
16.
PLoS One ; 7(3): e32887, 2012.
Article in English | MEDLINE | ID: mdl-22403721

ABSTRACT

Disruption of ephrin B1 in collagen I producing cells in mice results in severe skull defects and reduced bone formation. Because ephrin B1 is also expressed during osteoclast differentiation and because little is known on the role of ephrin B1 reverse signaling in bone resorption, we examined the bone phenotypes in ephrin B1 conditional knockout mice, and studied the function of ephrin B1 reverse signaling on osteoclast differentiation and resorptive activity. Targeted deletion of ephrin B1 gene in myeloid lineage cells resulted in reduced trabecular bone volume, trabecular number and trabecular thickness caused by increased TRAP positive osteoclasts and bone resorption. Histomorphometric analyses found bone formation parameters were not changed in ephrin B1 knockout mice. Treatment of wild-type precursors with clustered soluble EphB2-Fc inhibited RANKL induced formation of multinucleated osteoclasts, and bone resorption pits. The same treatment of ephrin B1 deficient precursors had little effect on osteoclast differentiation and pit formation. Similarly, activation of ephrin B1 reverse signaling by EphB2-Fc treatment led to inhibition of TRAP, cathepsin K and NFATc1 mRNA expression in osteoclasts derived from wild-type mice but not conditional knockout mice. Immunoprecipitation with NHERF1 antibody revealed ephrin B1 interacted with NHERF1 in differentiated osteoclasts. Treatment of osteoclasts with exogenous EphB2-Fc resulted in reduced phosphorylation of ezrin/radixin/moesin. We conclude that myeloid lineage produced ephrin B1 is a negative regulator of bone resorption in vivo, and that activation of ephrin B1 reverse signaling inhibits osteoclast differentiation in vitro in part via a mechanism that involves inhibition of NFATc1 expression and modulation of phosphorylation status of ezrin/radixin/moesin.


Subject(s)
Bone Resorption/genetics , Cell Differentiation/genetics , Ephrin-B1/deficiency , Ephrin-B1/genetics , Gene Knockout Techniques , Myeloid Cells/metabolism , Osteoclasts/cytology , Animals , Bone Density/genetics , Bone Resorption/pathology , Cytoskeletal Proteins/metabolism , Ephrin-B1/metabolism , Gene Expression Regulation/genetics , Humans , Macrophage Colony-Stimulating Factor/metabolism , Membrane Proteins/metabolism , Mice , Microfilament Proteins/metabolism , Myeloid Cells/cytology , Osteoclasts/metabolism , RANK Ligand/metabolism , Signal Transduction/genetics
17.
J Biol Chem ; 279(35): 36993-7003, 2004 Aug 27.
Article in English | MEDLINE | ID: mdl-15213236

ABSTRACT

Gap junctions are composed of connexin (Cx) proteins and mediate intercellular communication required for many developmental and physiological processes. Here we describe the isolation and characterization of Cx48.5, a zebrafish connexin with the highest sequence identity to mammalian Cx46. Expression analysis showed that Cx48.5 is expressed in the adult and embryonic lens and heart, adult testis, and transiently in the embryonic otic vesicles. Injection of Cx48.5 cRNA into Xenopus oocytes elicited intercellular electrical coupling with voltage sensitivity similar to mammalian Cx46. In single oocytes, Cx48.5 also induced large outward currents on depolarization, consistent with gap-junctional hemichannels. Disruption of Cx48.5 expression in embryos with antisense morpholino oligos (morpholinos) revealed that Cx48.5 has an essential role in the maintenance of lens homeostasis. The morpholino-treated embryos also developed small lenses and eyes as well as severe cardiovascular abnormalities.


Subject(s)
Connexins/physiology , Lens, Crystalline/embryology , Zebrafish Proteins/physiology , Amino Acid Sequence , Animals , Cardiovascular System/embryology , Chickens , Connexins/metabolism , Gene Library , Immunohistochemistry , In Situ Hybridization , Lens, Crystalline/metabolism , Male , Mice , Molecular Sequence Data , Oligonucleotides/chemistry , Oocytes/metabolism , RNA, Complementary/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Testis/metabolism , Time Factors , Xenopus , Zebrafish , Zebrafish Proteins/metabolism
18.
Dev Dyn ; 228(4): 709-15, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14648847

ABSTRACT

Connexins (Cx), the protein units of gap junctions, play important roles in lens development and homeostasis. Here, we report the mRNA expression patterns of zebrafish Cx48.5, Cx44.1, Cx43 during lens development. The expression of all three connexins in the adult lens was first confirmed by reverse transcriptase-polymerase chain reaction. By whole-mount in situ hybridization, we detected Cx48.5 expression throughout the lens, except the lateral lens epithelium, at 36 hours postfertilization (hpf). The pattern remained the same at 2 days postfertilization (dpf). By 3 and 4 dpf, Cx48.5 expression was restricted to the differentiating lens fibers in the equatorial and medial regions. Cx44.1 was expressed in a similar manner as Cx48.5 from 36 hpf to 4 dpf. However, Cx44.1 expression was also detected in the lens at 24 hpf. Cx43 expression was detected throughout the lens at 24 and 36 hpf but became restricted to the lateral epithelium at later stages.


Subject(s)
Connexin 43/biosynthesis , Connexins/biosynthesis , Gene Expression Regulation, Developmental , Lens, Crystalline/metabolism , Zebrafish Proteins/biosynthesis , Animals , Chickens , DNA Primers/pharmacology , Gap Junctions , In Situ Hybridization , Mice , Phylogeny , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Tissue Distribution , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL
...