Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
1.
Bone ; 142: 115709, 2021 01.
Article in English | MEDLINE | ID: mdl-33148508

ABSTRACT

Hypertrophy of chondrocytes is a crucial step in the endochondral bone formation process that drives bone lengthening and the transition to endochondral bone formation. Both Parathyroid hormone-related protein (PTHrP) and Histone deacetylase 4 (HDAC4) inhibit chondrocyte hypertrophy. Use of multiple mouse genetics models reveals how PTHrP and HDAC4 participate in a pathway that regulates chondrocyte hypertrophy. PTHrP/cAMP/protein kinase A (PKA) signaling pathway phosphorylates the PKA-target sites on salt-inducible kinase 3 (Sik3), which leads to inhibition of Sik3 kinase activity. Inhibition of Sik3 kinase activity decreases phosphorylation of HDAC4 by Sik3 at binding sites for 14-3-3; lower levels of HDAC4 phosphorylation then allow HDAC4 nuclear translocation. In the nucleus, the transcription factor, Myocyte Enhancer Factor 2 (Mef2), activates Runt-related transcription factor 2 (Runx2), and together these two transcription factors drive the hypertrophic process. HDAC4 binds both Mef2 and Runx2 and blocks their activities. There are genetic redundancies in this pathway. Sik1 and Sik2 also mediate PTHrP/cAMP/PKA signaling when Sik3 activity is low. HDAC5 also mediates PTHrP signaling when HDAC4 expression is low. Thus, PTHrP triggers a kinase cascade that leads to inhibition of the key transcription factors (Mef2 and Runx2) that promote chondrocyte hypertrophy.


Subject(s)
Growth Plate , Parathyroid Hormone-Related Protein , Animals , Chondrocytes , Histone Deacetylases , Hypertrophy , Mice , Protein Serine-Threonine Kinases
2.
EMBO Rep ; 9(9): 878-84, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18660753

ABSTRACT

Transforming growth factor-beta1 (TGF-beta1) has many physiological functions and inhibits the differentiation of osteoblasts. Previously, we reported that TGF-beta1 stimulation induces the degradation of p57(KIP2) in osteoblasts. p57(KIP2) proteolysis depends on the ubiquitin-proteasome pathway and SMAD-mediated transcription; however, the molecular mechanism underlying p57(KIP2) degradation has been largely unknown. Here, we show that FBL12, a new F-box protein expressed in the limb bud of developing embryos, is involved in TGF-beta1-induced degradation of p57(KIP2). FBL12 formed an SCF(FBL12) complex and directly ubiquitinated p57(KIP2) in a phosphorylation-dependent manner. Inhibition of FBL12 by RNA interference suppressed the degradation of p57(KIP2) and a dominant-negative mutant of FBL12 (FBL12DeltaF) increased the steady-state level of p57(KIP2). Furthermore, wild-type FBL12 inhibited and FBL12DeltaF promoted the differentiation of primary osteoblasts. As overexpression of p57(KIP2) promoted osteoblast differentiation, these results indicate the importance of FBL12 and the degradation of p57(KIP2) in the regulation of osteoblast cell differentiation.


Subject(s)
Cell Differentiation/physiology , Cyclin-Dependent Kinase Inhibitor p57/metabolism , Osteoblasts/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Blotting, Northern , Cell Differentiation/drug effects , Cell Line , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p57/genetics , F-Box Proteins/genetics , F-Box Proteins/metabolism , Gene Expression/drug effects , HeLa Cells , Humans , Immunoblotting , Immunoprecipitation , Mice , Oligonucleotide Array Sequence Analysis , Osteoblasts/cytology , Protein Binding/drug effects , RNA Interference , Transfection , Transforming Growth Factor beta/pharmacology
3.
Elife ; 92020 10 16.
Article in English | MEDLINE | ID: mdl-33063669

ABSTRACT

Growth plate and articular cartilage constitute a single anatomical entity early in development but later separate into two distinct structures by the secondary ossification center (SOC). The reason for such separation remains unknown. We found that evolutionarily SOC appears in animals conquering the land - amniotes. Analysis of the ossification pattern in mammals with specialized extremities (whales, bats, jerboa) revealed that SOC development correlates with the extent of mechanical loads. Mathematical modeling revealed that SOC reduces mechanical stress within the growth plate. Functional experiments revealed the high vulnerability of hypertrophic chondrocytes to mechanical stress and showed that SOC protects these cells from apoptosis caused by extensive loading. Atomic force microscopy showed that hypertrophic chondrocytes are the least mechanically stiff cells within the growth plate. Altogether, these findings suggest that SOC has evolved to protect the hypertrophic chondrocytes from the high mechanical stress encountered in the terrestrial environment.


Subject(s)
Cell Differentiation , Cell Proliferation , Chondrocytes/metabolism , Growth Plate/growth & development , Osteogenesis , Animals , Biomechanical Phenomena , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Stress, Mechanical
4.
JCI Insight ; 4(5)2019 03 07.
Article in English | MEDLINE | ID: mdl-30843886

ABSTRACT

During endochondral bone formation, chondrocyte hypertrophy represents a crucial turning point from chondrocyte differentiation to bone formation. Both parathyroid hormone-related protein (PTHrP) and histone deacetylase 4 (HDAC4) inhibit chondrocyte hypertrophy. Using multiple mouse genetics models, we demonstrate in vivo that HDAC4 is required for the effects of PTHrP on chondrocyte differentiation. We further show in vivo that PTHrP leads to reduced HDAC4 phosphorylation at the 14-3-3-binding sites and subsequent HDAC4 nuclear translocation. The Hdac4-KO mouse shares a similar but milder phenotype with the Pthrp-KO mouse, indicating the possible existence of other mediators of PTHrP action. We identify HDAC5 as an additional mediator of PTHrP signaling. While the Hdac5-KO mouse has no growth plate phenotype at birth, the KO of Hdac5 in addition to the KO of Hdac4 is required to block fully PTHrP action on chondrocyte differentiation at birth in vivo. Finally, we show that PTHrP suppresses myocyte enhancer factor 2 (Mef2) action that allows runt-related transcription factor 2 (Runx2) mRNA expression needed for chondrocyte hypertrophy. Our results demonstrate that PTHrP inhibits chondrocyte hypertrophy and subsequent bone formation in vivo by allowing HDAC4 and HDAC5 to block the Mef2/Runx2 signaling cascade. These results explain the phenotypes of several genetic abnormalities in humans.


Subject(s)
Chondrocytes/metabolism , Histone Deacetylases/metabolism , Hypertrophy/metabolism , Parathyroid Hormone-Related Protein/metabolism , Animals , Cartilage/pathology , Cell Proliferation , Chondrocytes/pathology , Core Binding Factor Alpha 1 Subunit/metabolism , Disease Models, Animal , Gene Expression Regulation , Histone Deacetylases/genetics , Humans , Hypertrophy/genetics , MEF2 Transcription Factors/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteogenesis/genetics , Osteogenesis/physiology , Parathyroid Hormone-Related Protein/genetics , Phenotype , Phosphorylation , RNA, Messenger/metabolism , Ribs/pathology , Signal Transduction , Transcriptome
5.
J Clin Invest ; 129(12): 5187-5203, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31430259

ABSTRACT

The parathyroid hormone 1 receptor (PTH1R) mediates the biologic actions of parathyroid hormone (PTH) and parathyroid hormone-related protein (PTHrP). Here, we showed that salt-inducible kinases (SIKs) are key kinases that control the skeletal actions downstream of PTH1R and that this GPCR, when activated, inhibited cellular SIK activity. Sik gene deletion led to phenotypic changes that were remarkably similar to models of increased PTH1R signaling. In growth plate chondrocytes, PTHrP inhibited SIK3, and ablation of this kinase in proliferating chondrocytes rescued perinatal lethality of PTHrP-null mice. Combined deletion of Sik2 and Sik3 in osteoblasts and osteocytes led to a dramatic increase in bone mass that closely resembled the skeletal and molecular phenotypes observed when these bone cells express a constitutively active PTH1R that causes Jansen's metaphyseal chondrodysplasia. Finally, genetic evidence demonstrated that class IIa histone deacetylases were key PTH1R-regulated SIK substrates in both chondrocytes and osteocytes. Taken together, our findings establish that SIK inhibition is central to PTH1R action in bone development and remodeling. Furthermore, this work highlights the key role of cAMP-regulated SIKs downstream of GPCR action.


Subject(s)
Bone Development , Bone Remodeling , Parathyroid Hormone/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptor, Parathyroid Hormone, Type 1/metabolism , Animals , Animals, Newborn , Cell Proliferation , Chondrocytes/metabolism , Extracellular Matrix Proteins/metabolism , Gene Deletion , Hypertrophy , Male , Mice , Mice, Knockout , Mutation , Osteoblasts/metabolism , Osteocytes/metabolism , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Transcriptome
6.
Nat Commun ; 7: 11277, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-27068606

ABSTRACT

Dental root formation is a dynamic process in which mesenchymal cells migrate toward the site of the future root, differentiate and secrete dentin and cementum. However, the identities of dental mesenchymal progenitors are largely unknown. Here we show that cells expressing osterix are mesenchymal progenitors contributing to all relevant cell types during morphogenesis. The majority of cells expressing parathyroid hormone-related peptide (PTHrP) are in the dental follicle and on the root surface, and deletion of its receptor (PPR) in these progenitors leads to failure of eruption and significantly truncated roots lacking periodontal ligaments. The PPR-deficient progenitors exhibit accelerated cementoblast differentiation with upregulation of nuclear factor I/C (Nfic). Deletion of histone deacetylase-4 (HDAC4) partially recapitulates the PPR deletion root phenotype. These findings indicate that PPR signalling in dental mesenchymal progenitors is essential for tooth root formation, underscoring importance of the PTHrP-PPR system during root morphogenesis and tooth eruption.


Subject(s)
Mesenchymal Stem Cells/metabolism , Organogenesis , Receptor, Parathyroid Hormone, Type 1/metabolism , Signal Transduction , Tooth Root/growth & development , Tooth Root/metabolism , Transcription Factors/metabolism , Animals , Cell Adhesion Molecules/metabolism , Cell Differentiation , Cell Lineage , Dental Cementum/cytology , Dental Sac/cytology , Epistasis, Genetic , Gene Deletion , Histone Deacetylases/metabolism , Mice , Models, Biological , Mutation/genetics , Parathyroid Hormone-Related Protein/metabolism , Phenotype , Sp7 Transcription Factor , Tooth Eruption , Tooth Root/cytology
7.
Nat Commun ; 7: 13176, 2016 10 19.
Article in English | MEDLINE | ID: mdl-27759007

ABSTRACT

Parathyroid hormone (PTH) activates receptors on osteocytes to orchestrate bone formation and resorption. Here we show that PTH inhibition of SOST (sclerostin), a WNT antagonist, requires HDAC4 and HDAC5, whereas PTH stimulation of RANKL, a stimulator of bone resorption, requires CRTC2. Salt inducible kinases (SIKs) control subcellular localization of HDAC4/5 and CRTC2. PTH regulates both HDAC4/5 and CRTC2 localization via phosphorylation and inhibition of SIK2. Like PTH, new small molecule SIK inhibitors cause decreased phosphorylation and increased nuclear translocation of HDAC4/5 and CRTC2. SIK inhibition mimics many of the effects of PTH in osteocytes as assessed by RNA-seq in cultured osteocytes and following in vivo administration. Once daily treatment with the small molecule SIK inhibitor YKL-05-099 increases bone formation and bone mass. Therefore, a major arm of PTH signalling in osteocytes involves SIK inhibition, and small molecule SIK inhibitors may be applied therapeutically to mimic skeletal effects of PTH.


Subject(s)
Bone and Bones/drug effects , Osteocytes/drug effects , Osteogenesis/drug effects , Parathyroid Hormone/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/genetics , Active Transport, Cell Nucleus/drug effects , Adaptor Proteins, Signal Transducing , Animals , Bone and Bones/cytology , Bone and Bones/metabolism , Gene Expression Regulation , Glycoproteins/antagonists & inhibitors , Glycoproteins/genetics , Glycoproteins/metabolism , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Humans , Intercellular Signaling Peptides and Proteins , Mice , Mice, Knockout , Osteocytes/cytology , Osteocytes/metabolism , Osteogenesis/genetics , Parathyroid Hormone/metabolism , Phosphorylation/drug effects , Primary Cell Culture , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , RANK Ligand/antagonists & inhibitors , RANK Ligand/genetics , RANK Ligand/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism
8.
Endocrinology ; 156(8): 2774-80, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26052897

ABSTRACT

The blood calcium concentration during fetal life is tightly regulated within a narrow range by highly interactive homeostatic mechanisms that include transport of calcium across the placenta and fluxes in and out of bone; the mechanisms of this regulation are poorly understood. Our findings that endochondral bone-specific PTH/PTHrP receptor (PPR) knockout (KO) mice showed significant reduction of fetal blood calcium concentration compared with that of control littermates at embryonic day 18.5 led us to focus on bone as a possibly major determinant of fetal calcium homeostasis. We found that the fetal calcium concentration of Runx2 KO mice was significantly higher than that of control littermates, suggesting that calcium flux into bone had a considerable influence on the circulating calcium concentration. Moreover, Runx2:PTH double mutant fetuses showed calcium levels similar to those of Runx2 KO mice, suggesting that part of the fetal hypocalcemia in PTH KO mice was caused by the increment of the mineralized bone mass allowed by the formation of osteoblasts. Finally, Rank:PTH double mutant mice had a blood calcium concentration even lower than that of the either Rank KO or PTH KO mice alone at embryonic day 18.5. These observations in our genetic models suggest that PTH/PTHrP receptor signaling in bones has a significant role of the regulation of fetal blood calcium concentration and that both placental transport and osteoclast activation contribute to PTH's hypercalcemic action. They also show that PTH-independent deposition of calcium in bone is the major controller of fetal blood calcium level.


Subject(s)
Bone and Bones/metabolism , Calcium/blood , Fetal Blood/metabolism , Homeostasis , Parathyroid Hormone-Related Protein/physiology , Parathyroid Hormone/physiology , Animals , Embryo, Mammalian , Female , Homeostasis/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Parathyroid Hormone/genetics , Parathyroid Hormone-Related Protein/genetics , Pregnancy , Signal Transduction/physiology
9.
J Bone Miner Res ; 30(6): 1044-52, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25529628

ABSTRACT

Growth plate chondrocytes go through multiple differentiation steps and eventually become hypertrophic chondrocytes. The parathyroid hormone (PTH)-related peptide (PTHrP) signaling pathway plays a central role in regulation of hypertrophic differentiation, at least in part, through enhancing activity of histone deacetylase 4 (HDAC4), a negative regulator of MEF2 transcription factors that drive hypertrophy. We have previously shown that loss of the chondrocyte-specific microRNA (miRNA), miR-140, alters chondrocyte differentiation including mild acceleration of hypertrophic differentiation. Here, we provide evidence that miR-140 interacts with the PTHrP-HDAC4 pathway to control chondrocyte differentiation. Heterozygosity of PTHrP or HDAC4 substantially impaired animal growth in miR-140 deficiency, whereas these mutations had no effect in the presence of miR-140. miR-140-deficient chondrocytes showed increased MEF2C expression with normal levels of total and phosphorylated HDAC4, indicating that the miR-140 pathway merges with the PTHrP-HDAC4 pathway at the level of MEF2C. miR-140 negatively regulated p38 mitogen-activated protein kinase (MAPK) signaling, and inhibition of p38 MAPK signaling reduced MEF2C expression. These results demonstrate that miR-140 ensures the robustness of the PTHrP/HDAC4 regulatory system by suppressing MEF2C-inducing stimuli. © 2014 American Society for Bone and Mineral Research © 2015 American Society for Bone and Mineral Research.


Subject(s)
Chondrocytes/metabolism , Histone Deacetylases/metabolism , MicroRNAs/metabolism , Parathyroid Hormone-Related Protein/metabolism , Signal Transduction , Animals , Chondrocytes/pathology , Histone Deacetylases/genetics , Hypertrophy/genetics , Hypertrophy/metabolism , Hypertrophy/pathology , MEF2 Transcription Factors/biosynthesis , MEF2 Transcription Factors/genetics , Mice , Mice, Knockout , MicroRNAs/genetics , Parathyroid Hormone-Related Protein/genetics
10.
J Bone Miner Res ; 30(3): 400-11, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25271055

ABSTRACT

Osteocytes secrete paracrine factors that regulate the balance between bone formation and destruction. Among these molecules, sclerostin (encoded by the gene SOST) inhibits osteoblastic bone formation and is an osteoporosis drug target. The molecular mechanisms underlying SOST expression remain largely unexplored. Here, we report that histone deacetylase 5 (HDAC5) negatively regulates sclerostin levels in osteocytes in vitro and in vivo. HDAC5 shRNA increases, whereas HDAC5 overexpression decreases SOST expression in the novel murine Ocy454 osteocytic cell line. HDAC5 knockout mice show increased levels of SOST mRNA, more sclerostin-positive osteocytes, decreased Wnt activity, low trabecular bone density, and reduced bone formation by osteoblasts. In osteocytes, HDAC5 binds and inhibits the function of MEF2C, a crucial transcription factor for SOST expression. Using chromatin immunoprecipitation, we have mapped endogenous MEF2C binding in the SOST gene to a distal intergenic enhancer 45 kB downstream from the transcription start site. HDAC5 deficiency increases SOST enhancer MEF2C chromatin association and H3K27 acetylation and decreases recruitment of corepressors NCoR and HDAC3. HDAC5 associates with and regulates the transcriptional activity of this enhancer, suggesting direct regulation of SOST gene expression by HDAC5 in osteocytes. Finally, increased sclerostin production achieved by HDAC5 shRNA is abrogated by simultaneous knockdown of MEF2C, indicating that MEF2C is a major target of HDAC5 in osteocytes.


Subject(s)
Glycoproteins/metabolism , Histone Deacetylases/physiology , Osteocytes/metabolism , Adaptor Proteins, Signal Transducing , Animals , Histone Deacetylases/genetics , Intercellular Signaling Peptides and Proteins , MEF2 Transcription Factors/metabolism , Mice , Mice, Knockout
12.
Biochem Biophys Res Commun ; 304(4): 632-7, 2003 May 16.
Article in English | MEDLINE | ID: mdl-12727200

ABSTRACT

The mechanism of transcriptional repression by nuclear hormone receptors, especially in the presence of the ligands, is largely unknown. We previously reported that 1,25-dihydroxyvitamin D(3) (1,25 vitamin D3) inhibited expression of the parathyroid hormone-related polypeptide (PTHrP) gene through the interaction between the liganded monomeric vitamin D receptor (VDR) and the negative DNA element in the PTHrP gene (nVDRE(RP)). In this study, we employed chromatin immunoprecipitation (ChIP) assay and confirmed that 1,25 vitamin D3 recruited DNA-dependent protein kinase (DNA-PKcs) to the chromatinized nVDRE(RP). Conversely, the regulatory subunits of DNA-PK were associated with the nVDRE(RP) sequences only when 1,25 vitamin D3 was absent. VDR was constitutively associated with these chromatinized nVDRE(RP) sequences. Furthermore, DNA-PKcs could phosphorylate VDR in vitro. We raise a possibility that a conformational change of VDR through its phosphorylation mediated by DNA-PKcs underlies the mechanism of gene repression by 1,25 vitamin D3-bound VDR.


Subject(s)
Calcitriol/metabolism , Chromatin/metabolism , DNA-Binding Proteins , Gene Expression Regulation , Peptide Hormones/genetics , Protein Serine-Threonine Kinases/metabolism , Vitamin D Response Element , Androstadienes/metabolism , Animals , Cell Line , DNA-Activated Protein Kinase , Enzyme Inhibitors/metabolism , Genes, Reporter , Humans , Nuclear Proteins , Parathyroid Hormone-Related Protein , Phosphorylation , Protein Conformation , Rats , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Wortmannin
13.
J Biol Chem ; 278(52): 52919-23, 2003 Dec 26.
Article in English | MEDLINE | ID: mdl-14530263

ABSTRACT

p57Kip2 is the only cyclin-dependent kinase (Cdk) inhibitor shown to be essential for mouse embryogenesis. The fact suggests that p57 has a specific role that cannot be compensated by other Cdk inhibitors. LIM-kinase 1 (LIMK-1) is a downstream effector of the Rho family of GTPases that phosphorylates and inactivates an actin depolymerization factor, cofilin, to induce the formation of actin fiber. Here we demonstrate that p57 regulates actin dynamics by binding and translocating LIMK-1 from the cytoplasm into the nucleus, which in turn results in a reorganization of actin fiber. The central region of p57, a unique feature among the Cdk inhibitors, and the N-terminal region of LIMK-1, which contains the LIM domains were essential for the interaction. Expression of p57, but not p27Kip1 or a p57 mutant, with a deletion in the central region was shown to induce marked reorganization of actin filament and a translocation of LIMK-1. Our findings indicate p57 may act as a key regulator in embryogenesis by bearing two distinct functions, the regulation of cell cycle through binding to Cdks and the regulation of actin dynamics through binding to LIMK-1, both of which should be important in developmental procedure.


Subject(s)
Actins/metabolism , Cell Nucleus/metabolism , Nuclear Proteins/physiology , Protein Kinases/metabolism , Actins/chemistry , Active Transport, Cell Nucleus , Animals , COS Cells , Cell Cycle , Cell Cycle Proteins/metabolism , Cell Line , Cells , Cyclin-Dependent Kinase Inhibitor p27 , Cyclin-Dependent Kinase Inhibitor p57 , Cytoplasm/metabolism , DNA, Complementary/metabolism , Enzyme Inhibitors/pharmacology , Gene Library , Humans , Immunoblotting , Immunohistochemistry , Lim Kinases , Mice , Models, Biological , Mutation , Nuclear Proteins/metabolism , Oligonucleotide Array Sequence Analysis , Osteoblasts/metabolism , Phosphorylation , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Protein Transport , Transfection , Tumor Suppressor Proteins/metabolism , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL