Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 710: 149860, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38604070

ABSTRACT

Schizophyllan (SPG), a ß-glucan from Schizophyllum commune, is recognized for its antioxidant, immunoregulatory, and anticancer activities. In this study, its effects on bone cells, particularly osteoclasts and osteoblasts, were examined. We demonstrated that SPG dose-dependently inhibited osteoclastogenesis and reduced gene expression associated with osteoclast differentiation. SPG also decreased bone resorption and F-actin ring formation. This inhibition could have been due to the downregulation of transcription factors c-Fos and nuclear factor of activated T cells 1 (NFATc1) via the MAPKs (JNK and p38), IκBα, and PGC1ß/PPARγ pathways. In coculture, SPG lowered osteoclastogenic activity in calvaria-derived osteoblasts by reducing macrophage colony-stimulating factor (M-CSF) and receptor activator of nuclear factor-κB ligand (RANKL) expression. In addition, SPG slightly enhanced osteoblast differentiation, as evidenced by increased differentiation marker gene expression and alizarin red staining. It also exhibited antiresorptive effects in a lipopolysaccharide-induced calvarial bone loss model. These results indicated a dual role of SPG in bone cell regulation by suppressing osteoclastogenesis and promoting osteoblast differentiation. Thus, SPG could be a therapeutic agent for bone resorption-related diseases such as osteoporosis, rheumatoid arthritis, and periodontitis.


Subject(s)
Bone Resorption , Sizofiran , Humans , Osteoclasts/metabolism , Sizofiran/metabolism , Sizofiran/pharmacology , NFATC Transcription Factors/metabolism , Osteoblasts/metabolism , Cell Differentiation , Bone Resorption/drug therapy , Bone Resorption/metabolism , Osteogenesis , RANK Ligand/metabolism
2.
Biochem Biophys Res Commun ; 646: 1-7, 2023 02 26.
Article in English | MEDLINE | ID: mdl-36689911

ABSTRACT

After peripheral nerve injury, demyelinating Schwann cells discharge myelin debris and macrophages execute myelin degradation, leading to demyelination of degenerating axons, which is essential for efficient nerve regeneration. In this study, we show that vacuolar-type proton ATPase subunit d2 (Atp6v0d2) is among the most highly upregulated genes in degenerating mouse sciatic nerves after nerve injury using microarray analysis. ATP6V0D2 is mostly expressed in macrophages of injured nerves. Atp6v0d2 knockout mice display delayed peripheral nerve demyelination and significantly attenuated myelin lipid digestion after nerve injury. However, macrophage recruitment and Schwann cell dedifferentiation are unaffected by loss of Atp6v0d2 expression. Taken together, these data demonstrate that ATP6V0D2 in macrophages is specifically required for demyelination during Wallerian degeneration.


Subject(s)
Demyelinating Diseases , Peripheral Nerve Injuries , Vacuolar Proton-Translocating ATPases , Mice , Animals , Peripheral Nerve Injuries/metabolism , Adenosine Triphosphatases/metabolism , Myelin Sheath/metabolism , Schwann Cells/metabolism , Wallerian Degeneration , Sciatic Nerve/metabolism , Mice, Knockout , Demyelinating Diseases/metabolism , Nerve Regeneration , Vacuolar Proton-Translocating ATPases/genetics , Vacuolar Proton-Translocating ATPases/metabolism
3.
Nat Commun ; 12(1): 2258, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33859201

ABSTRACT

Selenoproteins containing selenium in the form of selenocysteine are critical for bone remodeling. However, their underlying mechanism of action is not fully understood. Herein, we report the identification of selenoprotein W (SELENOW) through large-scale mRNA profiling of receptor activator of nuclear factor (NF)-κΒ ligand (RANKL)-induced osteoclast differentiation, as a protein that is downregulated via RANKL/RANK/tumour necrosis factor receptor-associated factor 6/p38 signaling. RNA-sequencing analysis revealed that SELENOW regulates osteoclastogenic genes. SELENOW overexpression enhances osteoclastogenesis in vitro via nuclear translocation of NF-κB and nuclear factor of activated T-cells cytoplasmic 1 mediated by 14-3-3γ, whereas its deficiency suppresses osteoclast formation. SELENOW-deficient and SELENOW-overexpressing mice exhibit high bone mass phenotype and osteoporosis, respectively. Ectopic SELENOW expression stimulates cell-cell fusion critical for osteoclast maturation as well as bone resorption. Thus, RANKL-dependent repression of SELENOW regulates osteoclast differentiation and blocks osteoporosis caused by overactive osteoclasts. These findings demonstrate a biological link between selenium and bone metabolism.


Subject(s)
Bone Remodeling/genetics , Osteoclasts/physiology , Osteogenesis/genetics , Osteoporosis/genetics , Selenoprotein W/metabolism , 14-3-3 Proteins/metabolism , Animals , Cell Differentiation/genetics , Disease Models, Animal , Gene Expression Regulation/physiology , Humans , Male , Mice , Mice, Knockout , NFATC Transcription Factors/metabolism , Osteoporosis/pathology , RANK Ligand/metabolism , RNA-Seq , Selenoprotein W/genetics , Signal Transduction/physiology , TNF Receptor-Associated Factor 6/metabolism
4.
J Bone Metab ; 27(1): 53-63, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32190609

ABSTRACT

BACKGROUND: Rosae Multiflorae fructus (RMF), known to have anti-inflammatory and antioxidant properties, has been used as a traditional remedy for inflammatory diseases such as arthritis in Eastern Asia. However, its effect on osteoclasts, which play a crucial role in resorptive inflammatory bone diseases, is yet to be elucidated. METHODS: The effect of extract of RMF (RMF-E) on receptor activator of nuclear factor-κB ligand (RANKL)-mediated osteoclastogenesis was examined by tartrate-resistant acid phosphatase (TRAP) staining, real-time polymerase chain reaction and western blot analysis. In addition, RANKL-induced Ca2+-oscillation was also investigated. RESULTS: RMF-E remarkably inhibited TRAP+-osteoclast and resorptive pit formation in a dose-dependent manner. In addition, the expression of c-Fos and nuclear factor of activated T-cells cytoplasmic, known as pivotal transcription factors for osteoclast formation in vitro and in vivo, and that of the osteoclast differentiation markers such as Acp5, Oscar, CtsK, Atp6v0d2, Tm7sf4, and Nfatc1 were significantly decreased by RMF-E treatment during osteoclastogenesis. The inhibitory effect of RMF-E on RANKL-induced osteoclastogenesis was caused by the suppression of p38 mitogen-activated protein kinase activation, and RANKL-induced Ca2+-oscillation removal via inactivation of Bruton's tyrosine kinase (BTK), and subsequently phospholipase C-γ2. CONCLUSIONS: RMF-E negatively regulates osteoclast differentiation and formation. These findings suggest the possibility of RMF-E as a traditional therapeutic agent against osteoclast-related bone disorders such as osteoporosis, rheumatoid arthritis, and periodontitis.

5.
Photodiagnosis Photodyn Ther ; 30: 101688, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32087294

ABSTRACT

BACKGROUND: To determine the safety and efficacy of antimicrobial photodynamic therapy (aPDT) combination of 0.33 mM Toluidine Blue O (TBO) with 60 mW/cm2 LED irradiation for 5 min that we had established, this study investigated the cytotoxic effect of aPDT combination on mammalian oral cells (gingival fibroblast and periodontal ligament cells) and compared the antimicrobial efficacy of antibiotics (the combination of amoxicillin (AMX) and metronidazole (MTZ)) against representative periodontitis pathogenic bacteria (Porphyromonas gingivalis, Fusobacterium nucleatum, and Aggregatibacter actinomycetemcomitans) versus our aPDT combination. RESULT: aPDT combination did not show any detectable effect on the viability of Streptococcus sanguinis or Streptococcus mitis, the most common resident species in the oral flora. However, it significantly reduced CFU values of P. gingivalis, F. nucleatum, and A. actinomycetemcomitans. The cytotoxicity of the present aPDT combination to mammalian oral cells was comparable to that of standard antiseptics used in oral cavity. In antimicrobial efficacy test, the present aPDT combination showed equivalent bactericidal rate compared to the combination of AMX + MTZ, the most widely used antibiotics in the periodontitis treatment. The bactericidal ability of the AMX + MTZ combination was effective against all five bacteria tested regardless of the bacterial species, whereas the bactericidal ability of the aPDT combination was effective only against P. gingivalis, F. nucleatum, and A. actinomycetemcomitans, the representative periodontitis pathogenic bacterial species. CONCLUSION: The present study demonstrated the safety and efficacy of the present aPDT combination in periodontitis treatment. TBO-mediated aPDT with LED irradiation has the potential to serve as a safe single or adjunctive antimicrobial procedure for nonsurgical periodontal treatment without damaging adjacent normal oral tissue or resident flora.


Subject(s)
Anti-Infective Agents , Periodontitis , Photochemotherapy , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Anti-Infective Agents/therapeutic use , Periodontitis/drug therapy , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Porphyromonas gingivalis
6.
BMB Rep ; 52(8): 514-519, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31383250

ABSTRACT

Osteoclasts are multinucleated giant cells derived from myeloid progenitors. Excessive bone resorption by osteoclasts can result in serious clinical outcomes for which better treatment options are needed. Here, we identified fibronectin leucine-rich transmembrane protein 2 (Flrt2), a ligand of the Unc5 receptor family for neurons, as a novel target associated with the late/maturation stage of osteoclast differentiation. Flrt2 expression is induced by stimulation with receptor activator of nuclear factor-kB ligand (RANKL). Flrt2 deficiency in osteoclasts results in reduced hyper-multinucleation, which could be restored by RNAi-mediated knockdown of Unc5b. Treatment with Netrin1, another ligand of Unc5b which negatively controls osteoclast multinucleation through down regulation of RANKL-induced Rac1 activation, showed no inhibitory effects on Flrt2-deficient cells. In addition, RANKL-induced Rac1 activation was attenuated in Flrt2-deficient cells. Taken together, these results suggest that Flrt2 regulates osteoclast multinucleation by interfering with Netrin 1-Unc5b interaction and may be a suitable therapeutic target for diseases associated with bone remodeling. [BMB Reports 2019; 52(8): 514-519].


Subject(s)
Membrane Glycoproteins/metabolism , Osteoclasts/metabolism , Animals , Membrane Glycoproteins/deficiency , Mice , Mice, Knockout , Osteoclasts/cytology
7.
Sci Rep ; 9(1): 8248, 2019 06 03.
Article in English | MEDLINE | ID: mdl-31160615

ABSTRACT

Periodontitis is initiated by causative bacteria in the gingival sulcus. However, as the lesion is often deep and out of circulation system and biofilm is frequently formed on the bacteria cluster, use of antibacterial agents has been limited and the invasive method such as curettage is thought as an only treatment. Here we designed non-invasive photodynamic therapy (PDT), with the ointment which leads a photosensitizer deliverable into gingival sulcus. We assessed whether 650 nm light-emitting-diode (LED) penetrates the 3-mm soft tissue and effectively activates a photosensitizer toluidine-blue-O (TBO) through the thickness to remove Porphyromonas gingivalis and Fusobacterium nucleatum species. The oral ointment formulation was optimized to efficiently deliver the photosensitizer into gingival sulcus and its efficacy of PDT was evaluated in in vitro and in vivo models. Four weeks of TBO-formulation mediated-PDT treatment significantly attenuated periodontitis-induced alveolar bone loss and inflammatory cytokines production in rats. These results confirm that a 650 nm LED indeed penetrates the gingiva and activates our TBO formulation which is sufficiently delivered to, and retained within, the gingival sulcus; thus, it effectively kills the bacteria that reside around the gingival sulcus. Collectively, TBO-mediated PDT using LED irradiation has potential as a safe adjunctive procedure for periodontitis treatment.


Subject(s)
Fusobacterium nucleatum/drug effects , Periodontitis/drug therapy , Periodontitis/microbiology , Photochemotherapy , Porphyromonas gingivalis/drug effects , Animals , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Biofilms/drug effects , Bone Resorption/pathology , Drug Liberation , Inflammation/pathology , Male , Microbial Sensitivity Tests , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Rats, Wistar , Tolonium Chloride/pharmacology , Tolonium Chloride/therapeutic use , Viscosity
8.
BMB Rep ; 51(7): 356-361, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29921413

ABSTRACT

Actin-binding LIM protein 1 (ABLIM1), a member of the LIM-domain protein family, mediates interactions between actin filaments and cytoplasmic targets. However, the role of ABLIM1 in osteoclast and bone metabolism has not been reported. In the present study, we investigated the role of ABLIM1 in the receptor activator of NF-κB ligand (RANKL)- mediated osteoclastogenesis. ABLIM1 expression was induced by RANKL treatment and knockdown of ABLIM1 by retrovirus infection containing Ablim1-specific short hairpin RNA (shAblim1) decreased mature osteoclast formation and bone resorption activity in a RANKL-dose dependent manner. Coincident with the downregulated expression of osteoclast differentiation marker genes, the expression levels of c-Fos and the nuclear factor of activated T-cells cytoplasmic 1 (NFATc1), critical transcription factors of osteoclastogenesis, were also decreased in shAblim1-infected osteoclasts during RANKLmediated osteoclast differentiation. In addition, the motility of preosteoclast was reduced by ABLIM1 knockdown via modulation of the phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/Akt/Rac1 signaling pathway, suggesting another regulatory mechanism of ABLIM1 in osteoclast formation. These data demonstrated that ABLIM1 is a positive regulator of RANKLmediated osteoclast formation via the modulation of the differentiation and PI3K/Akt/Rac1-dependent motility. [BMB Reports 2018; 51(7): 356-361].


Subject(s)
Cell Differentiation/drug effects , LIM Domain Proteins/metabolism , Microfilament Proteins/metabolism , RANK Ligand/pharmacology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Movement/drug effects , Cells, Cultured , LIM Domain Proteins/antagonists & inhibitors , LIM Domain Proteins/genetics , Mice , Mice, Inbred C57BL , Microfilament Proteins/antagonists & inhibitors , Microfilament Proteins/genetics , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Neuropeptides/metabolism , Osteoclasts/cytology , Osteoclasts/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , rac1 GTP-Binding Protein/metabolism
10.
Am J Chin Med ; 45(8): 1725-1744, 2017.
Article in English | MEDLINE | ID: mdl-29121799

ABSTRACT

Puerariae radix, the dried root of Pueraria lobate Ohwi, is known to prevent bone loss in ovariectomized mice; however, the precise molecular mechanisms are not understood. In this study, we investigated the effects and underlying mechanisms of action of Puerariae radix extract (PRE) on receptor activator of NF-κB ligand (RANKL)-induced osteoclastogenesis. PRE dose-dependently inhibited osteoclast differentiation and formation, decreased the bone-resorbing activity of osteoclasts, and downregulated the expression of osteoclast differentiation marker genes. The expression of osteoclastogenic factors produced by PRE-treated osteoblasts such as RANKL, macrophage colony-stimulating factor (M-CSF), and osteoprotegerin (OPG) was comparable to that of untreated (control) cells. However, the formation of osteoclasts via bone marrow cell and calvaria-derived osteoblast co-cultures was suppressed by PRE treatment. Therefore, the inhibitory effects of PRE on osteoclastogenesis clearly targeted osteoclasts, but not osteoblasts. PRE treatment considerably reduced RANKL-induced mitogen-activated protein kinases (MAPKs) activity, especially c-Jun N-terminal kinase, in osteoclast precursor cells. In addition, PRE markedly suppressed cAMP response element-binding protein (CREB) activation and the induction of peroxisome proliferator-activated receptor gamma coactivator 1ß (PGC1ß), which stimulate osteoclastogenesis - an effect that was not observed for puerarin and 17-ß estradiol. Finally, PRE treatment significantly repressed the expression of c-Fos and the nuclear factor of activated T-cells cytoplasmic 1 (NFATc1), which is a master transcription factor for osteoclastogenesis in vitro and in vivo. Overall, these results strongly suggest that PRE is an effective inhibitor of RANKL-induced osteoclastogenesis and may be a potent therapeutic agent for bone-related diseases such as osteoporosis, rheumatoid arthritis, and periodontitis.


Subject(s)
Down-Regulation/drug effects , Gene Expression/drug effects , Osteogenesis/drug effects , Plant Extracts/pharmacology , Pueraria/chemistry , RANK Ligand/adverse effects , Signal Transduction/drug effects , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Cytokines/drug effects , Dose-Response Relationship, Drug , Female , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Mice, Inbred C57BL , NFATC Transcription Factors/genetics , NFATC Transcription Factors/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Osteoclasts/cytology , Osteoclasts/drug effects , Osteoporosis/drug therapy , Osteoprotegerin/genetics , Osteoprotegerin/metabolism , Phytotherapy , Plant Extracts/isolation & purification , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Transcription Factors/genetics , Transcription Factors/metabolism
12.
BMC Complement Altern Med ; 17(1): 164, 2017 Mar 23.
Article in English | MEDLINE | ID: mdl-28335757

ABSTRACT

BACKGROUND: Sophorae Flos (SF) is a composite of flowers and buds of Styphnolobium japonicum (L.) Schott and has been used in traditional Korean and Chinese medicine for the treatment of hemostasis and inflammation. Previous studies reported that SF possesses anti-obesity properties, as well as anti-allergic, anti-proliferative, and anti-inflammatory activities. However, the effect of SF in bone resorption has not been studies. In this study, we examined the potential of SF extract (SFE) to inhibit receptor activator of NF-κB ligand (RANKL) -induced osteoclast differentiation in cultured mouse-derived bone marrow macrophages (BMMs). METHODS: BMMs, that act as osteoclast precursors, were cultured with M-CSF (50 ng/ml) and RANKL (100 ng/ml) for 4 days to generate osteoclasts. Osteoclast differentiation was measured by tartrate-resistant acidic phosphatase (TRAP) staining and the TRAP solution assay. Osteoclast differentiation marker genes were analyzed by the quantitative real-time polymerase chain reaction analysis. RANKLs signaling pathways were confirmed through western blotting. RESULTS: SFE significantly decreased osteoclast differentiation in a dose-dependent manner. SFE inhibited RANKL-induced osteoclastogenesis by suppressing NF-κB activation. By contrast, SFE did not affect phospholipase C gamma 2 or subsequent cAMP response element binding activation. SFE inhibited the RANKL-induced expression of nuclear factor of activated T cells c1 (NFATc1). CONCLUSIONS: SFE attenuated the RANKL-mediated induction of NF-κB through inhibition of IκBα phosphorylation, which contributed to inhibiting of RANKL-induced osteoclast differentiation through downregulation of NFATc1.


Subject(s)
Bone Marrow Cells/drug effects , NFATC Transcription Factors/metabolism , Osteoclasts/drug effects , Osteogenesis/drug effects , Plant Extracts/pharmacology , RANK Ligand/metabolism , Sophora/chemistry , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cells, Cultured , Down-Regulation/drug effects , Flowers/chemistry , Mice , Mice, Inbred BALB C , NF-kappa B/genetics , NF-kappa B/metabolism , NFATC Transcription Factors/genetics , Osteoclasts/cytology , Osteoclasts/metabolism , Signal Transduction/drug effects
13.
Biochem Biophys Res Commun ; 482(4): 1073-1079, 2017 Jan 22.
Article in English | MEDLINE | ID: mdl-27913299

ABSTRACT

ß-lapachone (ß-L) is a substrate of reduced nicotinamide adenine dinucleotide (NADH): quinone oxidoreductase 1 (NQO1). NQO1 reduces quinones to hydroquinones using NADH as an electron donor and consequently increases the intracellular NAD+/NADH ratio. The activation of NQO1 by ß-L has beneficial effects on several metabolic syndromes, such as obesity, hypertension, and renal injury. However, the effect of ß-L on bone metabolism remains unclear. Here, we show that ß-L might be a potent inhibitor of receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclastogenesis. ß-L inhibited osteoclast formation in a dose-dependent manner and also reduced the expression of osteoclast differentiation marker genes, such as tartrate-resistant acid phosphatase (Acp5 or TRAP), cathepsin K (CtsK), the d2 isoform of vacuolar ATPase V0 domain (Atp6v0d2), osteoclast-associated receptor (Oscar), and dendritic cell-specific transmembrane protein (Dc-stamp). ß-L treatment of RANKL-induced osteoclastogenesis significantly increased the cellular NAD+/NADH ratio and resulted in the activation of 5' AMP-activated protein kinase (AMPK), a negative regulator of osteoclast differentiation. In addition, ß-L treatment led to significant suppression of the expression of peroxisome proliferator-activated receptor gamma (PPARγ) and peroxisome proliferator-activated receptor gamma coactivator 1ß (PGC1ß), which can stimulate osteoclastogenesis. ß-L treatment downregulated c-Fos and nuclear factor of activated T-cells 1 (NFATc1), which are master transcription factors for osteoclastogenesis. Taken together, the results demonstrated that ß-L inhibits RANKL-induced osteoclastogenesis and could be considered a potent inhibitor of RANKL-mediated bone diseases, such as postmenopausal osteoporosis, rheumatoid arthritis, and periodontitis.


Subject(s)
Naphthoquinones/chemistry , Osteoclasts/cytology , RANK Ligand/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Bone Diseases/metabolism , Cell Differentiation , Cell Survival , Gene Expression Profiling , Mice , Mice, Inbred C57BL , NAD/chemistry , NAD(P)H Dehydrogenase (Quinone)/metabolism , NFATC Transcription Factors/metabolism , Osteoclasts/metabolism , Osteogenesis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Real-Time Polymerase Chain Reaction
14.
J Bone Miner Res ; 32(2): 385-396, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27589205

ABSTRACT

Lysosomal Ca2+ emerges as a critical component of receptor-evoked Ca2+ signaling and plays a crucial role in many lysosomal and physiological functions. Lysosomal Ca2+ release is mediated by the transient receptor potential (TRP) family member TRPML1, mutations that cause the lysosomal storage disease mucolipidosis type 4. Lysosomes play a key role in osteoclast function. However, nothing is known about the role of lysosomal Ca2+ signaling in osteoclastogenesis and bone metabolism. In this study, we addressed this knowledge gap by studying the role of lysosomal Ca2+ signaling in osteoclastogenesis, osteoclast and osteoblast functions, and bone homeostasis in vivo. We manipulated lysosomal Ca2+ signaling by acute knockdown of TRPML1, deletion of TRPML1 in mice, pharmacological inhibition of lysosomal Ca2+ influx, and depletion of lysosomal Ca2+ storage using the TRPML agonist ML-SA1. We found that knockdown and deletion of TRPML1, although it did not have an apparent effect on osteoblast differentiation and bone formation, markedly attenuated osteoclast function, RANKL-induced cytosolic Ca2+ oscillations, inhibited activation of NFATc1 and osteoclastogenesis-controlling genes, suppressed the formation of tartrate-resistant acid phosphatase (TRAP)-positive multinucleated cells (MNCs), and markedly reduced the differentiation of bone marrow-derived macrophages into osteoclasts. Moreover, deletion of TRPML1 resulted in enlarged lysosomes, inhibition of lysosomal secretion, and attenuated the resorptive activity of mature osteoclasts. Notably, depletion of lysosomal Ca2+ with ML-SA1 similarly abrogated RANKL-induced Ca2+ oscillations and MNC formation. Deletion of TRPML1 in mice reduced the TRAP-positive bone surfaces and impaired bone remodeling, resulting in prominent osteopetrosis. These findings demonstrate the essential role of lysosomal Ca2+ signaling in osteoclast differentiation and mature osteoclast function, which play key roles in bone homeostasis. © 2016 American Society for Bone and Mineral Research.


Subject(s)
Bone Remodeling , Calcium Signaling , Lysosomes/metabolism , Osteoclasts/metabolism , Osteogenesis , Animals , Bone Remodeling/drug effects , Bone Resorption/pathology , Calcium Signaling/drug effects , Cell Size , Gene Deletion , Lysosomes/drug effects , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred C57BL , Osteoclasts/drug effects , Osteogenesis/drug effects , RANK Ligand/pharmacology , Tartrate-Resistant Acid Phosphatase/metabolism , Transient Receptor Potential Channels/deficiency , Transient Receptor Potential Channels/metabolism
15.
Biochem Biophys Res Commun ; 475(1): 125-32, 2016 06 17.
Article in English | MEDLINE | ID: mdl-27179783

ABSTRACT

Cytosolic malate dehydrogenase (malate dehydrogenase 1, MDH1) plays pivotal roles in the malate/aspartate shuttle that might modulate metabolism between the cytosol and mitochondria. In this study, we investigated the role of MDH1 in osteoclast differentiation and formation. MDH1 expression was induced by receptor activator of nuclear factor kappa-B ligand (RANKL) treatment. Knockdown of MDH1 by infection with retrovirus containing MDH1-specific shRNA (shMDH1) reduced mature osteoclast formation and bone resorption activity. Moreover, the expression of marker genes associated with osteoclast differentiation was downregulated by shMDH1 treatment, suggesting a role of MDH1 in osteoclast differentiation. In addition, intracellular ATP production was reduced following the activation of adenosine 5' monophosphate-activated protein kinase (AMPK), a cellular energy sensor and negative regulator of RANKL-induced osteoclast differentiation, in shMDH1-infected osteoclasts compared to control cells. In addition, the expression of c-Fos and nuclear factor of activated T-cells, cytoplasmic 1 (NFATc1), a critical transcription factor of osteoclastogenesis, was decreased with MDH1 knockdown during RANKL-mediated osteoclast differentiation. These findings provide strong evidence that MDH1 plays a critical role in osteoclast differentiation and function via modulation of the intracellular energy status, which might affect AMPK activity and NFATc1 expression.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Malate Dehydrogenase/metabolism , NFATC Transcription Factors/metabolism , Osteoclasts/metabolism , Proto-Oncogene Proteins c-fos/metabolism , RANK Ligand/metabolism , Signal Transduction , Animals , Cell Differentiation , Mice, Inbred C57BL , Osteoclasts/cytology
16.
PLoS One ; 11(4): e0153886, 2016.
Article in English | MEDLINE | ID: mdl-27077737

ABSTRACT

Osteoclasts seem to be metabolic active during their differentiation and bone-resorptive activation. However, the functional role of lactate dehydrogenase (LDH), a tetrameric enzyme consisting of an A and/or B subunit that catalyzes interconversion of pyruvate to lactate, in RANKL-induced osteoclast differentiation is not known. In this study, RANKL treatment induced gradual gene expression and activation of the LDH A2B2 isotype during osteoclast differentiation as well as the LDH A1B3 and B4 isotypes during osteoclast maturation after pre-osteoclast formation. Glucose consumption and lactate production in growth media were accelerated during osteoclast differentiation, together with enhanced expression of H+-lactate co-transporter and increased extracellular acidification, demonstrating that glycolytic metabolism was stimulated during differentiation. Further, oxygen consumption via mitochondria was stimulated during osteoclast differentiation. On the contrary, depletion of LDH-A or LDH-B subunit suppressed both glycolytic and mitochondrial metabolism, resulting in reduced mature osteoclast formation via decreased osteoclast precursor fusion and down-regulation of the osteoclastogenic critical transcription factor NFATc1 and its target genes. Collectively, our findings suggest that RANKL-induced LDH activation stimulates glycolytic and mitochondrial respiratory metabolism, facilitating mature osteoclast formation via osteoclast precursor fusion and NFATc1 signaling.


Subject(s)
Cell Differentiation/genetics , L-Lactate Dehydrogenase/genetics , NFATC Transcription Factors/genetics , Osteoclasts/metabolism , Signal Transduction/genetics , Animals , Cells, Cultured , Gene Expression/drug effects , Glucose/metabolism , Glycolysis/drug effects , Glycolysis/genetics , Immunoblotting , Isoenzymes/genetics , Isoenzymes/metabolism , L-Lactate Dehydrogenase/metabolism , Lactates/metabolism , Macrophage Colony-Stimulating Factor/pharmacology , Male , Mice, Inbred C57BL , NFATC Transcription Factors/metabolism , Osteoclasts/cytology , Osteoclasts/drug effects , Oxygen Consumption/genetics , RANK Ligand/pharmacology , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction
17.
Sci Rep ; 6: 22511, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26928655

ABSTRACT

The mitochondrial sirtuin 3 (SIRT3) is involved in suppressing the onset of multiple pathologies, including cardiovascular disease, fatty liver, age-related hearing loss, and breast cancer. But a physiological role of SIRT3 in bone metabolism is not known. Here we show that SIRT3 is a key regulatory molecule to maintain bone homeostasis. Mice deficient in SIRT3 exhibited severe osteopenia owing to increased numbers of osteoclasts. Osteoclast precursors from Sirt3-/- mice underwent increased osteoclastogenesis in response to receptor activator of nuclear factor-κB ligand (RANKL), an essential cytokine for osteoclast differentiation. SIRT3 expression from RANKL induction depended on the transcription coactivator PGC-1ß (peroxisome proliferator-activated receptor-γ co-activator-1ß) and the nuclear receptor ERRα (estrogen receptor-related receptor α), and that SIRT3 inhibited the differentiation by interfering with the RANKL-induced expression of PGC-1ß. Thus an auto-regulatory feedback mechanism operates to induce its own inhibitor SIRT3 by PGC-1ß. Moreover, Sirt3-/- osteoclast precursors reduced AMP-activated protein kinase (AMPK) phosphorylation through down-regulating the expression of AMPK. Our results suggest that a mitochondrial SIRT3 is an intrinsic inhibitor for RANKL-mediated osteoclastogenesis.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Osteoclasts/cytology , Osteogenesis/physiology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Receptors, Estrogen/metabolism , Sirtuin 3/metabolism , Animals , Bone Diseases, Metabolic/genetics , Bone Remodeling/genetics , Bone Remodeling/physiology , Cells, Cultured , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteogenesis/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , RANK Ligand/metabolism , RNA Interference , RNA, Small Interfering/genetics , Sirtuin 3/genetics , ERRalpha Estrogen-Related Receptor
18.
J Immunol ; 196(3): 1123-31, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-26695370

ABSTRACT

Rac1, a member of small GTPases, is a key regulator of osteoclast differentiation and function. The Crk family adaptor proteins, consisting of Src homology (SH) 2 and SH3 protein-binding domains, regulate cell proliferation, migration, and invasion through Rac1 activation. In this study, we examined the role of CrkII in osteoclast differentiation and function. Retroviral overexpression of CrkII in osteoclast precursors enhanced osteoclast differentiation and resorptive function through Rac1 activation. The knockdown of CrkII in osteoclast precursors using small interfering RNA inhibited osteoclast differentiation and its resorption activity. Unlike wild-type CrkII, overexpression of the three SH domains in mutant forms of CrkII did not enhance either osteoclast differentiation or function. Phosphorylation of p130 Crk-associated substrate (p130Cas) by osteoclastogenic cytokines in preosteoclasts increased the interaction between p130Cas and CrkII, which is known to be involved in Rac1 activation. Furthermore, transgenic mice overexpressing CrkII under control of a tartrate-resistant acid phosphatase promoter exhibited a low bone mass phenotype, associated with increased resorptive function of osteoclasts in vivo. Taken together, our data suggest that the p130Cas/CrkII/Rac1 signaling pathway plays an important role in osteoclast differentiation and function, both in vitro and in vivo.


Subject(s)
Cell Differentiation/physiology , Osteoclasts/physiology , Proto-Oncogene Proteins c-crk/metabolism , Signal Transduction/physiology , Animals , Blotting, Western , Crk-Associated Substrate Protein/metabolism , Gene Knockdown Techniques , Mice , Mice, Transgenic , Osteoclasts/cytology , RANK Ligand/metabolism , RNA, Small Interfering , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transfection , X-Ray Microtomography
19.
Int J Mol Sci ; 16(12): 29305-14, 2015 Dec 09.
Article in English | MEDLINE | ID: mdl-26690137

ABSTRACT

Small G-protein adenosine diphosphate (ADP)-ribosylation factors (ARFs) regulate a variety of cellular functions, including actin cytoskeleton remodeling, plasma membrane reorganization, and vesicular transport. Here, we propose the functional roles of ARF1 in multiple stages of osteoclast differentiation. ARF1 was upregulated during receptor activator of nuclear factor kappa-B ligand (RANKL)-induced osteoclast differentiation and transiently activated in an initial stage of their differentiation. Differentiation of ARF1-deficient osteoclast precursors into mature osteoclasts temporarily increased in pre-maturation stage of osteoclasts followed by reduced formation of mature osteoclasts, indicating that ARF1 regulates the osteoclastogenic process. ARF1 deficiency resulted in reduced osteoclast precursor proliferation and migration as well as increasing cell-cell fusion. In addition, ARF1 silencing downregulated c-Jun N-terminal kinase (JNK), Akt, osteopontin, and macrophage colony-stimulating factor (M-CSF)-receptor c-Fms as well as upregulating several fusion-related genes including CD44, CD47, E-cadherin, and meltrin-α. Collectively, we showed that ARF1 stimulated proliferation and migration of osteoclast precursors while suppressing their fusion, suggesting that ARF1 may be a plausible inter-player that mediates the transition to osteoclast fusion at multiple steps during osteoclast differentiation.


Subject(s)
ADP-Ribosylation Factor 1/physiology , Cell Differentiation , Cell Movement , Cell Proliferation , Osteoclasts/physiology , Animals , Cell Fusion , Cells, Cultured , Male , Mice, Inbred C57BL , Signal Transduction
20.
Bone ; 78: 165-73, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25979161

ABSTRACT

Age-related osteoporosis is associated with a reciprocal decrease in bone formation and an increase in adiposity in the bone marrow niche. We previously reported Transmembrane protein 64 (Tmem64) to be an important regulator of osteoclast function; however, its precise role in osteoblasts has not yet been established. Here, we showed that ablation of the Tmem64 gene in mice resulted in markedly increased osteoblast and reduced adipocyte differentiation from bone marrow-derived stromal cells (BMSCs). Conversely, Tmem64 overexpression inhibited osteogenesis and accelerated adipogenesis. Furthermore, BMSCs isolated from Tmem64 knockout mice formed a greater number of colony-forming unit-osteoblasts and a lower number of colony-forming unit-adipocytes than the wild type controls. Mechanistically, the expression level of ß-catenin, the key Wnt signaling molecule, increased significantly, and its nuclear translocation was enhanced in Tmem64-deficient cells. Introduction of Tmem64 significantly suppressed ß-catenin-mediated transcriptional activity in an in vitro co-transfection experiment as well as during an in vivo experiment involving BAT-Gal reporter mice. These results demonstrate that Tmem64 plays an important role in the regulation of mesenchymal lineage allocation by modulating Wnt/ß-catenin signaling.


Subject(s)
Adipocytes/cytology , Membrane Proteins/genetics , Membrane Proteins/physiology , Osteoblasts/cytology , Wnt Signaling Pathway , Adipogenesis , Animals , CCAAT-Enhancer-Binding Protein-alpha/metabolism , Cell Differentiation , Cell Lineage , Cell Nucleus/metabolism , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Osteogenesis , PPAR gamma/metabolism , Signal Transduction , Stem Cells , Wnt Proteins/metabolism , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...