Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 991
Filter
1.
Bioengineered ; 13(2): 4201-4211, 2022 02.
Article in English | MEDLINE | ID: mdl-35176943

ABSTRACT

Osteoporosis is a metabolic bone disease that significantly affects the quality of life and can even lead to death. In this study, we aimed to investigate the role of RAD51 recombinase (RAD51) in osteoblast and osteoclast differentiation. We analyzed differentially expressed genes using microarray analysis. The osteogenic differentiation capability was analyzed by alkaline phosphatase (ALP) staining and alizarin red staining assays. Osteogenesis and osteoclast related genes expression was detected using quantitative real-time PCR (qPCR) and Western blotting. The phosphorylation of Ataxia-telangiectasia mutated (ATM) and ATR serine/threonine kinase (ATR) was tested using Western blotting. The effect of RAD51 on osteoporosis was also explored in vivo. The results showed that RAD51 was downregulated in osteoporosis, but upregulated in differentiated osteoblasts. Overexpression of RAD51 enhanced the differentiation of osteoblasts and suppressed the formation of osteoclasts. Furthermore, p-ATM and p-ATR levels were upregulated in osteoblasts and downregulated in osteoclasts. RAD51 expression was reduced by the ATM/ATR pathway inhibitor AZ20. AZ20 treatment inhibited osteoblastogenesis and promoted osteoclastogenesis, whereas RAD51 reversed the effects induced by AZ20. Moreover, RAD51 improved bone microarchitecture in vivo. Taken together, ATM/ATR signaling-mediated RAD51 promoted osteogenic differentiation and suppressed osteoclastogenesis. These findings reveal a critical role for RAD51 in osteoporosis.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Osteoclasts/cytology , Osteogenesis , Osteoporosis/metabolism , Rad51 Recombinase/metabolism , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins/genetics , Humans , Male , Mice , NIH 3T3 Cells , Osteoblasts/cytology , Osteoblasts/enzymology , Osteoclasts/enzymology , Osteoporosis/genetics , Osteoporosis/physiopathology , Rad51 Recombinase/genetics , Rats , Rats, Sprague-Dawley
2.
J Biol Chem ; 298(3): 101639, 2022 03.
Article in English | MEDLINE | ID: mdl-35090892

ABSTRACT

Phosphatidylinositol-4-phosphate 5-kinase type-1 gamma (Pip5k1c) is a lipid kinase that plays a pivotal role in the regulation of receptor-mediated calcium signaling in multiple tissues; however, its role in the skeleton is not clear. Here, we show that while deleting Pip5k1c expression in the mesenchymal stem cells using Prx1-Cre transgenic mice does not impair the intramembranous and endochondral ossification during skeletal development, it does cause osteopenia in adult mice, but not rapidly growing young mice. We found Pip5k1c loss dramatically decreases osteoblast formation and osteoid and mineral deposition, leading to reduced bone formation. Furthermore, Pip5k1c loss inhibits osteoblastic, but promotes adipogenic, differentiation of bone marrow stromal cells. Pip5k1c deficiency also impairs cytoplasmic calcium influx and inactivates the calcium/calmodulin-dependent protein kinase, which regulates levels of transcription factor Runx2 by modulating its stability and subsequent osteoblast and bone formation. In addition, Pip5k1c loss reduces levels of the receptor activator of nuclear factor-κB ligand, but not that of osteoprotegerin, its decoy receptor, in osteoblasts in bone and in sera. Finally, we found Pip5k1c loss impairs the ability of bone marrow stromal cells to support osteoclast formation of bone marrow monocytes and reduces the osteoclast precursor population in bone marrow, resulting in reduced osteoclast formation and bone resorption. We conclude Pip5k1c deficiency causes a low-turnover osteopenia in mice, with impairment of bone formation being greater than that of bone resorption. Collectively, we uncover a novel function and mechanism of Pip5k1c in the control of bone mass and identify a potential therapeutic target for osteoporosis.


Subject(s)
Bone Diseases, Metabolic , Bone Resorption , Mesenchymal Stem Cells , Phosphotransferases (Alcohol Group Acceptor) , Animals , Bone Diseases, Metabolic/genetics , Bone Diseases, Metabolic/metabolism , Bone Remodeling/physiology , Bone Resorption/enzymology , Bone Resorption/metabolism , Calcium/metabolism , Cell Differentiation/physiology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/enzymology , Mesenchymal Stem Cells/metabolism , Mice , Osteoblasts/cytology , Osteoblasts/enzymology , Osteoblasts/metabolism , Osteoclasts/cytology , Osteoclasts/enzymology , Osteoclasts/metabolism , Osteogenesis , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Phosphotransferases (Alcohol Group Acceptor)/metabolism , RANK Ligand/metabolism
3.
J Cell Biochem ; 123(2): 275-288, 2022 02.
Article in English | MEDLINE | ID: mdl-34668232

ABSTRACT

Systemic and intracellular metabolic states are critical factors affecting immune cell functions. The metabolic regulator AMP-activated protein kinase (AMPK) senses AMP levels and mediates cellular responses to energy-restrained conditions. The ubiquitously expressed AMPK participates in various biological functions in numerous cell types, including innate immune cell macrophages and osteoclasts, which are their specialized derivatives in bone tissues. Previous studies have demonstrated that the activation of AMPK promotes macrophage polarization toward anti-inflammatory M2 status. Additionally, AMPK acts as a negative regulator of osteoclastogenesis, and upregulation of AMPK disrupts the differentiation of osteoclasts. However, the regulation and roles of AMPK in differentiated osteoclasts have not been characterized. Here, we report that inflammatory stimuli-regulated-AMPK activation of differentiated and undifferentiated osteoclasts in opposite ways. Lipopolysaccharide (LPS) inhibited the phosphorylation of AMPK in macrophages and undifferentiated osteoclasts, but it activated AMPK in differentiated osteoclasts. Inactivating AMPK decreased cellular responses against the activation of toll-like receptor signaling, including the transcriptional activation of proinflammatory cytokines and the bone resorption genes TRAP, and MMP9. The elevation of bone resorption by LPS stimulation was disrupted by AMPK inhibitor, indicating the pivotal roles of AMPK in inflammation-induced activities in differentiated osteoclasts. The AMPK activator metformin did not increase proinflammatory responses, possibly because other factors are also required for this regulation. Notably, changing the activation status of AMPK did not alter the expression levels of bone resorption genes in unstimulated osteoclasts, indicating the essential roles of AMPK in cellular responses to inflammatory stimuli but not in the maintenance of basal levels. Unlike its M2-polarizing roles in macrophages, AMPK was not responsive to the M2 stimulus of interleukin-4. Our observations revealed differences in the cellular properties of macrophages and osteoclasts as well as the complexity of regulatory mechanisms for osteoclast functions.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Bone Resorption , Cell Differentiation/drug effects , Lipopolysaccharides/toxicity , Osteoclasts/enzymology , Animals , Bone Resorption/chemically induced , Bone Resorption/enzymology , Inflammation/chemically induced , Inflammation/enzymology , Mice , RAW 264.7 Cells
4.
Sci Rep ; 11(1): 22708, 2021 11 22.
Article in English | MEDLINE | ID: mdl-34811438

ABSTRACT

Osteoclasts are multinucleated, bone-resorbing cells. However, they also digest cartilage during skeletal maintenance, development and in degradative conditions including osteoarthritis, rheumatoid arthritis and primary bone sarcoma. This study explores the mechanisms behind the osteoclast-cartilage interaction. Human osteoclasts differentiated on acellular human cartilage expressed osteoclast marker genes (e.g. CTSK, MMP9) and proteins (TRAP, VNR), visibly damaged the cartilage surface and released glycosaminoglycan in a contact-dependent manner. Direct co-culture with chondrocytes during differentiation increased large osteoclast formation (p < 0.0001) except when co-cultured on dentine, when osteoclast formation was inhibited (p = 0.0002). Osteoclasts cultured on dentine inhibited basal cartilage degradation (p = 0.012). RNA-seq identified MMP8 overexpression in osteoclasts differentiated on cartilage versus dentine (8.89-fold, p = 0.0133), while MMP9 was the most highly expressed MMP. Both MMP8 and MMP9 were produced by osteoclasts in osteosarcoma tissue. This study suggests that bone-resident osteoclasts and chondrocytes exert mutually protective effects on their 'native' tissue. However, when osteoclasts contact non-native cartilage they cause degradation via MMPs. Understanding the role of osteoclasts in cartilage maintenance and degradation might identify new therapeutic approaches for pathologies characterized by cartilage degeneration.


Subject(s)
Cartilage/enzymology , Chondrocytes/enzymology , Dentin/enzymology , Joints/enzymology , Matrix Metalloproteinases/metabolism , Osteoclasts/enzymology , Cartilage/ultrastructure , Cell Differentiation , Cells, Cultured , Chondrocytes/ultrastructure , Coculture Techniques , Dentin/ultrastructure , Humans , Joints/ultrastructure , Matrix Metalloproteinase 8/genetics , Matrix Metalloproteinase 8/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinases/genetics , Osteoclasts/ultrastructure , Proteolysis
5.
Comput Math Methods Med ; 2021: 2909454, 2021.
Article in English | MEDLINE | ID: mdl-34691235

ABSTRACT

In an effort to bolster our understanding of regulation of bone formation in the context of osteoporosis, we screened out differentially expressed genes in osteoporosis patients with high and low bone mineral density by bioinformatics analysis. PIK3R1 is increasingly being nominated as a pivotal mediator in the differentiation of osteoblasts and osteoclasts that is closely related to bone formation. However, the specific mechanisms underlying the way that PIK3R1 affects bone metabolism are not fully elucidated. We intended to examine the potential mechanism by which PIK3R1 regulates osteoblast differentiation. Enrichment analysis was therefore carried out for differentially expressed genes. We noted that the estrogen signaling pathway, TNF signaling pathway, and osteoclast differentiation were markedly associated with ossification, and they displayed enrichment in PIK3R1. Based on western blot, qRT-PCR, and differentiation analysis in vitro, we found that upregulation of PIK3R1 enhanced osteoblastic differentiation, as evidenced by increased levels of investigated osteoblast-related genes as well as activities of ALP and ARS, while it notably decreased levels of investigated osteoclast-related genes. On the contrary, downregulation of PIK3R1 decreased levels of osteoblast-related genes and increased levels of osteoclast-related genes. Besides, in vitro experiments revealed that PIK3R1 facilitated proliferation and repressed apoptosis of osteoblasts but had an opposite impact on osteoclasts. In summary, PIK3R1 exhibits an osteoprotective effect via regulating osteoblast differentiation, which can be represented as a promising therapeutic target for osteoporosis.


Subject(s)
Class Ia Phosphatidylinositol 3-Kinase/genetics , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Osteoblasts/enzymology , Osteoclasts/enzymology , Osteogenesis/physiology , 3T3 Cells , Animals , Bone Density/genetics , Bone Density/physiology , Cell Differentiation/genetics , Cell Differentiation/physiology , Computational Biology , Female , Gene Expression Regulation, Enzymologic , Humans , Mice , Osteoblasts/cytology , Osteoclasts/cytology , Osteogenesis/genetics , Osteoporosis/enzymology , Osteoporosis/genetics , RAW 264.7 Cells , Signal Transduction , Up-Regulation
6.
J Laryngol Otol ; 135(10): 879-882, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34348812

ABSTRACT

BACKGROUND: Cholesteatoma-related bone destruction is the cause of many complications due to chronic otitis media. This study aimed to evaluate osteoclastic activity in cholesteatoma-related bone destruction using tartrate-resistant acid phosphatase 5b, an enzyme specific to osteoclastic activity. METHOD: Seventy-two patients diagnosed with chronic otitis media were included in this study and were divided into two groups: with and without bone destruction. The blood serum and tissue tartrate-resistant acid phosphatase 5b levels from both groups were compared. RESULTS: There were no significant differences in the level of serum enzymes between both groups. However, in tissue samples, tartrate-resistant acid phosphatase 5b levels were significantly lower in the bone destruction group than the group without bone destruction. CONCLUSION: This study determined that the level of tartrate-resistant acid phosphatase 5b, a specific enzyme for osteoclastic activity in cholesteatoma-related bone destruction, is locally decreased. This data suggests that osteoclastic activity may decrease in cholesteatoma-related bone destruction. However, further experimental and clinical studies are required to clarify this highly complex mechanism.


Subject(s)
Cholesteatoma, Middle Ear/complications , Osteoclasts/enzymology , Otitis Media/complications , Adult , Bone Resorption/etiology , Bone Resorption/metabolism , Bone Resorption/pathology , Case-Control Studies , Cholesteatoma, Middle Ear/metabolism , Cholesteatoma, Middle Ear/pathology , Chronic Disease , Female , Humans , Male , Middle Aged , Osteoclasts/pathology , Otitis Media/diagnosis , Otitis Media/metabolism , Tartrate-Resistant Acid Phosphatase/blood
7.
Biomed Res Int ; 2021: 6108999, 2021.
Article in English | MEDLINE | ID: mdl-34327232

ABSTRACT

Protocatechualdehyde (PCA), an important component of Salvia miltiorrhiza, has many activities, such as anti-inflammatory and antisepsis activities. However, the role of PCA in osteoclasts is not clear. We used RAW264.7 cells (a mouse leukemic monocyte/macrophage cell line) and bone marrow macrophages (BMMs) to probe the role of PCA in osteoclasts and the underlying mechanism. The effects of PCA on cell activity were evaluated with CCK-8 assays. TRAP staining detected mature osteoclasts. Corning Osteo Assay Surface plates were used to examine absorption. The levels of RNA and protein were analyzed, respectively, using RT-PCR and Western blotting. PCA (5 µg/ml) was not toxic to the two cell types but reduced the formation of osteoclasts and bone absorption. Furthermore, PCA restrained the expression of mRNAs encoding proteins associated with osteoclasts and reduced the phosphorylation of proteins in important signaling pathways. The results indicate that PCA inhibits osteoclast differentiation by suppressing NF-κB and MAPK activity.


Subject(s)
Benzaldehydes/pharmacology , Catechols/pharmacology , Cell Differentiation/drug effects , Macrophages/cytology , Macrophages/enzymology , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Osteoclasts/cytology , Osteoclasts/enzymology , Actins/metabolism , Animals , Apoptosis/drug effects , Biomarkers/metabolism , Gene Expression Regulation/drug effects , MAP Kinase Signaling System/drug effects , Macrophages/drug effects , Male , Mice , Mice, Inbred BALB C , Osteoclasts/drug effects , RANK Ligand/pharmacology , RAW 264.7 Cells , RNA, Messenger/genetics , RNA, Messenger/metabolism
8.
Cell Death Dis ; 12(7): 654, 2021 06 26.
Article in English | MEDLINE | ID: mdl-34175898

ABSTRACT

Bromodomain-containing protein 4 (BRD4) has emerged as a promising treatment target for bone-related disorders. (+)-JQ1, a thienotriazolodiazepine compound, has been shown to inhibit pro-osteoclastic activity in a BRD4-dependent approach and impede bone loss caused by ovariectomy (OVX) in vivo. However, clinical trials of (+)-JQ1 are limited because of its poor druggability. In this study, we synthesized a new (+)-JQ1 derivative differing in structure and chirality. One such derivative, (+)-ND, exhibited higher solubility and excellent inhibitory activity against BRD4 compared with its analogue (+)-JQ1. Interestingly, (-)-JQ1 and (-)-ND exhibited low anti-proliferative activity and had no significant inhibitory effect on RANKL-induced osteoclastogenesis as compared with (+)-JQ1 and (+)-ND, suggesting the importance of chirality in the biological activity of compounds. Among these compounds, (+)-ND displayed the most prominent inhibitory effect on RANKL-induced osteoclastogenesis. Moreover, (+)-ND could inhibit osteoclast-specific gene expression, F-actin ring generation, and bone resorption in vitro and prevent bone loss in OVX mice. Collectively, these findings indicated that (+)-ND represses RANKL-stimulated osteoclastogenesis and averts OVX-triggered osteoporosis by suppressing MAPK and NF-κB signalling cascades, suggesting that it may be a prospective candidate for osteoporosis treatment.


Subject(s)
Bone Density Conservation Agents/pharmacology , Bone Remodeling/drug effects , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Nuclear Proteins/antagonists & inhibitors , Osteoclasts/drug effects , Osteogenesis/drug effects , Osteoporosis, Postmenopausal/prevention & control , RANK Ligand/pharmacology , Transcription Factors/antagonists & inhibitors , Animals , Bone Density Conservation Agents/chemistry , Cells, Cultured , Disease Models, Animal , Female , Humans , Mice, Inbred C57BL , Molecular Structure , Nuclear Proteins/metabolism , Osteoclasts/enzymology , Osteoclasts/pathology , Osteoporosis, Postmenopausal/enzymology , Osteoporosis, Postmenopausal/pathology , Osteoporosis, Postmenopausal/physiopathology , Ovariectomy , Signal Transduction , Stereoisomerism , Structure-Activity Relationship , Transcription Factors/metabolism
9.
Biomolecules ; 11(3)2021 03 16.
Article in English | MEDLINE | ID: mdl-33809737

ABSTRACT

Bisphosphonates (BPs) are compounds resembling the pyrophosphate structure. BPs bind the mineral component of bones. During the bone resorption by osteoclasts, nitrogen-containing BPs are released and internalized, causing an inhibition of the mevalonate pathway. As a consequence, osteoclasts are unable to execute their function. Alendronate (ALN) is a bisphosphonate used to treat osteoporosis. Its administration could be associated with adverse effects. The purpose of this study is to evaluate four different ALN concentrations, ranging from 10-6 to 10-10 M, in the presence of different combinations of M-CSF and RANKL, to find out the effect of low ALN concentrations on osteoclastogenesis using rat and human peripheral blood mononuclear cells. The cytotoxic effect of ALN was evaluated based on metabolic activity and DNA concentration measurement. The alteration in osteoclastogenesis was assessed by the activity of carbonic anhydrase II (CA II), tartrate-resistant acid phosphatase staining, and actin ring formation. The ALN concentration of 10-6 M was cytotoxic. Low ALN concentrations of 10-8 and 10-10 M promoted proliferation, osteoclast-like cell formation, and CA II activity. The results indicated the induction of osteoclastogenesis with low ALN concentrations. However, when high doses of ALN were administered, their cytotoxic effect was demonstrated.


Subject(s)
Alendronate/pharmacology , Macrophage Colony-Stimulating Factor/pharmacology , Osteogenesis/drug effects , RANK Ligand/pharmacology , Actins/metabolism , Animals , Carbonic Anhydrase II/metabolism , Cell Proliferation/drug effects , DNA/metabolism , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Osteoclasts/drug effects , Osteoclasts/enzymology , Osteoclasts/metabolism , Rats , Staining and Labeling , Tartrate-Resistant Acid Phosphatase/metabolism
10.
Nat Commun ; 12(1): 1832, 2021 03 23.
Article in English | MEDLINE | ID: mdl-33758201

ABSTRACT

Synthetic glucocorticoids (GCs), one of the most effective treatments for chronic inflammatory and autoimmune conditions in children, have adverse effects on the growing skeleton. GCs inhibit angiogenesis in growing bone, but the underlying mechanisms remain unclear. Here, we show that GC treatment in young mice induces vascular endothelial cell senescence in metaphysis of long bone, and that inhibition of endothelial cell senescence improves GC-impaired bone angiogenesis with coupled osteogenesis. We identify angiogenin (ANG), a ribonuclease with pro-angiogenic activity, secreted by osteoclasts as a key factor for protecting the neighboring vascular cells against senescence. ANG maintains the proliferative activity of endothelial cells through plexin-B2 (PLXNB2)-mediated transcription of ribosomal RNA (rRNA). GC treatment inhibits ANG production by suppressing osteoclast formation in metaphysis, resulting in impaired endothelial cell rRNA transcription and subsequent cellular senescence. These findings reveal the role of metaphyseal blood vessel senescence in mediating the action of GCs on growing skeleton and establish the ANG/PLXNB2 axis as a molecular basis for the osteoclast-vascular interplay in skeletal angiogenesis.


Subject(s)
Cellular Senescence/drug effects , Endothelial Cells/metabolism , Glucocorticoids/pharmacology , Neovascularization, Physiologic/drug effects , Nerve Tissue Proteins/metabolism , Osteoclasts/metabolism , Ribonuclease, Pancreatic/metabolism , Animals , Apoptosis/drug effects , Bone Development/drug effects , Cell Proliferation/drug effects , Cellular Senescence/genetics , Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Methylprednisolone/pharmacology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neovascularization, Pathologic , Nerve Tissue Proteins/genetics , Osteoclasts/drug effects , Osteoclasts/enzymology , Osteogenesis/drug effects , RNA, Ribosomal/biosynthesis , RNA, Small Interfering , Recombinant Proteins , Ribonuclease, Pancreatic/genetics , Ribonuclease, Pancreatic/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Tomography Scanners, X-Ray Computed
11.
Int J Mol Sci ; 22(4)2021 Feb 11.
Article in English | MEDLINE | ID: mdl-33670411

ABSTRACT

Pycnodysostosis, a rare autosomal recessive skeletal dysplasia, is caused by a deficiency of cathepsin K. Patients have impaired bone resorption in the presence of normal or increased numbers of multinucleated, but dysfunctional, osteoclasts. Cathepsin K degrades collagen type I and generates N-telopeptide (NTX) and the C-telopeptide (CTX) that can be quantified. Levels of these telopeptides are increased in lactating women and are associated with increased bone resorption. Nothing is known about the consequences of cathepsin K deficiency in lactating women. Here we present for the first time normalized blood and CTX measurements in a patient with pycnodysostosis, exclusively related to the lactation period. In vitro studies using osteoclasts derived from blood monocytes during lactation and after weaning further show consistent bone resorption before and after lactation. Increased expression of cathepsins L and S in osteoclasts derived from the lactating patient suggests that other proteinases could compensate for the lack of cathepsin K during the lactation period of pycnodysostosis patients.


Subject(s)
Bone Resorption/enzymology , Cathepsin K/deficiency , Cathepsin L/metabolism , Cathepsins/metabolism , Lactation/metabolism , Osteoclasts/enzymology , Pycnodysostosis/enzymology , Adult , Bone Resorption/genetics , Bone Resorption/pathology , Cathepsin K/metabolism , Cathepsin L/genetics , Cathepsins/genetics , Female , Humans , Osteoclasts/pathology , Pycnodysostosis/genetics , Pycnodysostosis/pathology
12.
Eur J Pharmacol ; 899: 174015, 2021 May 15.
Article in English | MEDLINE | ID: mdl-33711307

ABSTRACT

Cyclic nucleotide phosphodiesterases (PDEs) are ubiquitously expressed enzymes that hydrolyze phosphodiester bond in the second messenger molecules including cAMP and cGMP. A wide range of drugs blocks one or more PDEs thereby preventing the inactivation of cAMP/cGMP. PDEs are differentially expressed in bone cells including osteoblasts, osteoclasts and chondrocytes. Intracellular increases in cAMP/cGMP levels in osteoblasts result in osteogenic response. Acting via the type 1 PTH receptor, teriparatide and abaloparatide increase intracellular cAMP and induce osteoanabolic effect, and many PDE inhibitors mimic this effect in preclinical studies. Since all osteoanabolic drugs are injectable and that oral drugs are considered to improve the treatment adherence and persistence, osteogenic PDE inhibitors could be a promising alternative to the currently available osteogenic therapies and directly assessed clinically in drug repurposing mode. Similar to teriparatide/abaloparatide, PDE inhibitors while stimulating osteoblast function also promote osteoclast function through stimulation of receptor activator of nuclear factor kappa-B ligand production from osteoblasts. In this review, we critically discussed the effects of PDE inhibitors in bone cells from cellular signalling to a variety of preclinical models that evaluated the bone formation mechanisms. We identified pentoxifylline (a non-selective PDE inhibitor) and rolipram (a PDE4 selective inhibitor) being the most studied inhibitors with osteogenic effect in preclinical models of bone loss at ≤ human equivalent doses, which suggest their potential for post-menopausal osteoporosis treatment through therapeutic repurposing. Subsequently, we treated pentoxifylline and rolipram as prototypical osteogenic PDEs to predict new chemotypes via the computer-aided design strategies for new drugs, based on the structural biology of PDEs.


Subject(s)
Bone and Bones/drug effects , Drug Repositioning , Osteogenesis/drug effects , Osteoporosis/drug therapy , Phosphodiesterase 4 Inhibitors/administration & dosage , Phosphodiesterase 5 Inhibitors/administration & dosage , Administration, Oral , Animals , Bone Density/drug effects , Bone Remodeling/drug effects , Bone and Bones/enzymology , Bone and Bones/pathology , Bone and Bones/physiopathology , Drug Design , Humans , Molecular Structure , Osteoblasts/drug effects , Osteoblasts/enzymology , Osteoblasts/pathology , Osteoclasts/drug effects , Osteoclasts/enzymology , Osteoclasts/pathology , Osteoporosis/enzymology , Osteoporosis/pathology , Osteoporosis/physiopathology , Phosphodiesterase 4 Inhibitors/adverse effects , Phosphodiesterase 5 Inhibitors/adverse effects , Signal Transduction , Structure-Activity Relationship
13.
J Biochem ; 169(4): 459-466, 2021 Apr 29.
Article in English | MEDLINE | ID: mdl-33135054

ABSTRACT

In osteoclasts, the a3 isoform of the proton-pumping V-ATPase plays essential roles in anterograde trafficking of secretory lysosomes and extracellular acidification required for bone resorption. This study examined functional complementation of the a isoforms by exogenously expressing the a1, a2 and a3 isoforms in a3-knockout (KO) osteoclasts. The expression levels of a1 and a2 in a3KO osteoclasts were similar, but lower than that of a3. a1 significantly localized to lysosomes, whereas a2 slightly did. On the other hand, a2 interacted with Rab7, a regulator of secretory lysosome trafficking in osteoclasts, more efficiently than a1. a1 partly complemented the functions of a3 in secretory lysosome trafficking and calcium phosphate resorption, while a2 partly complemented the former but not the latter function.


Subject(s)
Lysosomes/enzymology , Osteoclasts/enzymology , Protein Subunits , Vacuolar Proton-Translocating ATPases/metabolism , Animals , Isoenzymes/metabolism , Lysosomes/genetics , Mice , Mice, Knockout , Vacuolar Proton-Translocating ATPases/genetics , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins
14.
Biomed Pharmacother ; 133: 111089, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33378983

ABSTRACT

Osteoarthritis is a chronic, systemic and inflammatory disease. However, the pathogenesis and understanding of RA are still limited. Ubiquitin-specific protease 13 (USP13) belongs to the deubiquitinating enzyme (DUB) superfamily, and has been implicated in various cellular events. Nevertheless, its potential on RA progression has little to be investigated. In the present study, we found that USP13 expression was markedly up-regulated in synovial tissue samples from patients with RA, and was down-regulated in human fibroblast-like synoviocytes (H-FLSs) stimulated by interleukin-1ß (IL-1ß), tumor necrosis factor alpha (TNF-α) or lipopolysaccharide (LPS). We then showed that over-expressing USP13 markedly suppressed inflammatory response, oxidative stress and apoptosis in H-FLSs upon IL-1ß or TNF-α challenge, whereas USP13 knockdown exhibited detrimental effects. In addition, USP13-induced protective effects were associated with the improvement of nuclear factor erythroid 2-related factor 2 (Nrf-2) and the repression of Casapse-3. Furthermore, phosphatase and tensin homolog (PTEN) expression was greatly improved by USP13 in H-FLSs upon IL-1ß or TNF-α treatment, whereas phosphorylated AKT expression was diminished. In response to IL-1ß or TNF-α exposure, nuclear transcription factor κB (NF-κB) signaling pathway was activated, whereas being significantly restrained in H-FLSs over-expressing USP13. Mechanistically, USP13 directly interacted with PTEN. Of note, we found that USP13-regulated cellular processes including inflammation, oxidative stress and apoptotic cell death were partly dependent on AKT activation. Furthermore, USP13 over-expression effectively inhibited osteoclastogenesis and osteoclast-associated gene expression. The in vivo experiments finally confirmed that USP13 dramatically repressed synovial hyperplasia, inflammatory cell infiltration, cartilage damage and bone loss in collagen-induced arthritis (CIA) mice via the same molecular mechanisms detected in vitro. Taken together, these findings suggested that targeting USP13 may provide feasible therapies for RA.


Subject(s)
Apoptosis , Arthritis, Experimental/prevention & control , Bone Remodeling , Endopeptidases/metabolism , Joints/enzymology , Osteoarthritis/prevention & control , Oxidative Stress , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Ubiquitin-Specific Proteases/metabolism , Aged , Animals , Arthritis, Experimental/enzymology , Arthritis, Experimental/genetics , Arthritis, Experimental/pathology , Cells, Cultured , Collagen Type II , Endopeptidases/genetics , Humans , Hyperplasia , Joints/pathology , Male , Mice, Inbred C57BL , Mice, Inbred DBA , Middle Aged , Osteoarthritis/enzymology , Osteoarthritis/genetics , Osteoarthritis/pathology , Osteoclasts/enzymology , Osteoclasts/pathology , Osteogenesis , PTEN Phosphohydrolase/genetics , Signal Transduction , Synoviocytes/enzymology , Synoviocytes/pathology , Ubiquitin-Specific Proteases/genetics
15.
Biomed Pharmacother ; 129: 110341, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32554249

ABSTRACT

Osteoclasts are the only cells in the body with a bone-resorption function. The identification of anti-osteoclastogenic agents is important in managing bone loss diseases. The dried vines of Trachelospermum jasminoides (Lindl.) Lem. have been used as a herbal medicine to treat musculoskeletal soreness in East Asia for hundreds of years. In the present study, we focused on the effect of Trachelospermum jasminoides (Lindl.) Lem. extract (TJE) on osteoclast differentiation. As indicated by tartrate-resistant acid phosphatase (TRAP) staining, TJE inhibited osteoclastogenesis induced by receptor activator of nuclear factor-κB ligand from bone marrow-derived monocytes/macrophages without showing any cytotoxicity. In addition, TJE effectively suppressed F-actin ring formation and the bone-resorption function of osteoclasts. The subsequent studies such as network pharmacology and molecular investigation, revealed that TJE inhibited osteoclastogenesis-related genes in a dose- and time-dependent manner through NF-κB, MAPK and AKT-mediated mechanism followed by the nuclear factor of activated T cells, cytoplasmic 1 (NFATc1)/c-Fos pathway. Our study could potentially explain the underlying molecular pharmacology of TJE in osteoclast-related diseases. What's more, it suggested that network pharmacology could help the modernization of traditional Chinese medicine.


Subject(s)
Apocynaceae , Cell Differentiation/drug effects , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Osteoclasts/drug effects , Osteogenesis/drug effects , Plant Extracts/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , 3T3 Cells , Animals , Apocynaceae/chemistry , Cell Differentiation/genetics , Coculture Techniques , Databases, Protein , Gene Expression Regulation , Gene Regulatory Networks , Humans , Male , Mice , Mice, Inbred C57BL , Osteoclasts/enzymology , Osteogenesis/genetics , Plant Extracts/isolation & purification , Protein Interaction Maps , Signal Transduction
16.
Biosci Rep ; 40(6)2020 06 26.
Article in English | MEDLINE | ID: mdl-32478376

ABSTRACT

Bone metastasis of colorectal cancer (CRC) cells leads to osteolysis. Aberrant activation of osteoclasts is responsible for bone resorption in tumor. In general, bone marrow-derived monocytes (BMMs) differentiate into osteoclasts, however, how CRC cells interact with BMMs and how to regulate the differentiation is elusive. We here report that CRC cells promote bone resorption in bone metastasis. Transcriptomic profiling revealed CCL3 up-regulated in MC-38 conditional medium treated BMMs. Further investigation demonstrated that CCL3 produced by BMMs facilitated cell infusion and thus promoted the osteoclastogenesis. In addition, CRC cells derived EGF stimulated the production of CCL3 in BMMs through activation of ERK/CREB pathway. Blockage of EGF or CCL3 can efficiently attenuate the osteolysis in bone metastasis of CRC.


Subject(s)
Bone Neoplasms/enzymology , Chemokine CCL3/metabolism , Colorectal Neoplasms/enzymology , Epidermal Growth Factor/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Osteoclasts/enzymology , Osteogenesis , Osteolysis/enzymology , Tibia/enzymology , Animals , Bone Neoplasms/genetics , Bone Neoplasms/secondary , Cell Communication , Cell Line, Tumor , Chemokine CCL3/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Cyclic AMP Response Element-Binding Protein/genetics , Cyclic AMP Response Element-Binding Protein/metabolism , Male , Mice, Inbred C57BL , Osteoclasts/pathology , Osteolysis/genetics , Osteolysis/pathology , Signal Transduction , Tibia/pathology
17.
J Ethnopharmacol ; 257: 112873, 2020 Jul 15.
Article in English | MEDLINE | ID: mdl-32298753

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Litsea cubeba (Lour.) Pers. has been traditionally used as a folk prescription for treating rheumatic diseases in China. AIM OF THE STUDY: This study aimed to investigate the effects and underlying mechanism of LCA, a new type of dibenzyl butane lignin compound extracted from L. cubeba, on macrophage colony stimulating factor (M-CSF) plus receptor activator of NF-κB ligand (RANKL)-induced osteoclast differentiation in mouse-derived bone marrow macrophages (BMMs). MATERIAL AND METHODS: TRAP staining, TRAP enzyme activity assay and actin ring staining were applied to identify the effects of LCA on osteoclast differentiation. Protein expression of NFATc1, c-Fos and MMP-9, and phosphorylation of p65, Akt, JNK, ERK and p38 in RANKL-induced osteoclasts was determined using western blotting to investigate the underlying mechanism. RESULTS: LCA significantly suppressed RANKL-induced osteoclast differentiation by inhibiting TRAP activity, decreasing the number of TRAP+ multinuclear osteoclasts and reducing the formation of F-actin ring without obvious cytotoxicity in BMMs. Moreover, LCA treatment strongly reduced protein expression of NFATc1, c-Fos and MMP-9, and attenuated the phosphorylation of p65, Akt, JNK, ERK and p38 in RANKL-stimulated BMMs. CONCLUSIONS: LCA ameliorated RANKL-induced osteoclast differentiation via inhibition of Akt and MAPK signalings in BMMs, and may serve as a potential pro-drug for bone destruction prevention.


Subject(s)
Cell Transdifferentiation/drug effects , Lignin/pharmacology , Litsea , Macrophages/drug effects , Mitogen-Activated Protein Kinases/metabolism , Osteoclasts/drug effects , Osteogenesis/drug effects , Plant Extracts/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , RANK Ligand/pharmacology , Animals , Cells, Cultured , Femur/cytology , Lignin/isolation & purification , Litsea/chemistry , Macrophages/enzymology , Male , Mice, Inbred C57BL , Osteoclasts/enzymology , Plant Extracts/isolation & purification , Signal Transduction , Tibia/cytology
18.
Med Sci Monit ; 26: e918370, 2020 Jan 08.
Article in English | MEDLINE | ID: mdl-31914120

ABSTRACT

BACKGROUND Although extracellular signal-regulated kinase 5 (ERK5) is known to be critical for osteoclast differentiation, there are few studies on how fluid shear stress (FSS) regulates osteoclast differentiation through the ERK5 signaling pathway. We examined the expression of nuclear factor of activated T cells c1 (NFATc1) in RAW264.7 cells and its downstream factors, including cathepsin K (CTSK), tartrate-resistant acid phosphatase (TRAP), matrix metalloproteinases-9 (MMP-9) and their relationship with ERK5. MATERIAL AND METHODS RAW264.7 cells were treated with RANKL, XMD8-92 (ERK5 inhibitor), and then loaded onto 12 dyn/cm² FSS for 4 days. Endpoints measured were osteoclast differentiation, bone resorption, and TRAP activity. Cell viability was detected by using the Cell Counting Kit-8 (CCK-8) assay. Western blot was used to analyze protein expression of phosphorylated-ERK5 (p-ERK5), NFATc1, CTSK, TRAP, and MMP-9. RESULTS FSS inhibited osteoclast differentiation and expression of NFATc1, CTSK, TRAP, and MMP-9; cell viability was not affected. ERK5 expression increased by FSS but not by RANKL, and it was blocked by XMD8-92. Furthermore, FSS suppressed osteoclast differentiation in RAW264.7 cells through ERK5 pathway. CONCLUSIONS Our findings demonstrated that FSS inhibited osteoclast differentiation in RAW264.7 cells via the ERK5 pathway through reduced NFATc1 expression and its downstream factors MMP-9, CTSK, and TRAP.


Subject(s)
Mitogen-Activated Protein Kinase 7/metabolism , Osteoclasts/cytology , Osteoclasts/enzymology , Animals , Biomechanical Phenomena , Bone Resorption , Cathepsin K/metabolism , Cell Differentiation/physiology , Cell Line , Cell Survival/physiology , Hematopoiesis , Matrix Metalloproteinase 9/metabolism , Mice , NFATC Transcription Factors/metabolism , Phosphorylation , RAW 264.7 Cells , Stress, Mechanical , Tartrate-Resistant Acid Phosphatase/metabolism
19.
Mol Cancer Ther ; 19(2): 650-660, 2020 02.
Article in English | MEDLINE | ID: mdl-31694887

ABSTRACT

Bone metastasis is a severe complication associated with various carcinomas. It causes debilitating pain and pathologic fractures and dramatically impairs patients' quality of life. Drugs aimed at osteoclast formation significantly reduce the incidence of skeletal complications and are currently the standard treatment for patients with bone metastases. Here, we reported that serum- and glucocorticoid-inducible kinase 1 (SGK1) plays a pivotal role in the formation and function of osteoclasts by regulating the Ca2+ release-activated Ca2+ channel Orai1. We showed that SGK1 inhibition represses osteoclastogenesis in vitro and prevents bone loss in vivo Furthermore, we validated the effect of SGK1 on bone metastasis by using an intracardiac injection model in mice. Inhibition of SGK1 resulted in a significant reduction in bone metastasis. Subsequently, the Oncomine and the OncoLnc database were employed to verify the differential expression and the association with clinical outcome of SGK1 gene in patients with breast cancer. Our data mechanistically demonstrated the regulation of the SGK1 in the process of osteoclastogenesis and revealed SGK1 as a valuable target for curing bone metastasis diseases.


Subject(s)
Bone Neoplasms/secondary , Breast Neoplasms/pathology , Immediate-Early Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Benzoates/pharmacology , Bone Neoplasms/enzymology , Bone Neoplasms/genetics , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line, Tumor , Disease Models, Animal , Female , Heterografts , Humans , Immediate-Early Proteins/antagonists & inhibitors , Male , Mice , Mice, Inbred C57BL , Mice, Nude , NF-kappa B/metabolism , NFATC Transcription Factors/metabolism , ORAI1 Protein/metabolism , Osteoclasts/enzymology , Osteoclasts/pathology , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-fos/biosynthesis , Proto-Oncogene Proteins c-fos/genetics , RANK Ligand/antagonists & inhibitors , RANK Ligand/metabolism , Signal Transduction , Transfection
20.
Int J Mol Sci ; 20(19)2019 Sep 21.
Article in English | MEDLINE | ID: mdl-31546645

ABSTRACT

Abdominal aortic aneurysm (AAA) is among the top 20 causes of death in the United States. Surgical repair is the gold standard for AAA treatment, therefore, there is a need for non-invasive therapeutic interventions. Aneurysms are more closely associated with the osteoclast-like catabolic degradation of the artery, rather than the osteoblast-like anabolic processes of arterial calcification. We have reported the presence of osteoclast-like cells (OLCs) in human and mouse aneurysmal tissues. The aim of this study was to examine OLCs from aneurysmal tissues as a source of degenerative proteases. Aneurysmal and control tissues from humans, and from the mouse CaPO4 and angiotensin II (AngII) disease models, were analyzed via flow cytometry and immunofluorescence for the expression of osteoclast markers. We found higher expression of the osteoclast markers tartrate-resistant acid phosphatase (TRAP), matrix metalloproteinase-9 (MMP-9), and cathepsin K, and the signaling molecule, hypoxia-inducible factor-1α (HIF-1α), in aneurysmal tissue compared to controls. Aneurysmal tissues also contained more OLCs than controls. Additionally, more OLCs from aneurysms express HIF-1α, and produce more MMP-9 and cathepsin K, than myeloid cells from the same tissue. These data indicate that OLCs are a significant source of proteases known to be involved in aortic degradation, in which the HIF-1α signaling pathway may play an important role. Our findings suggest that OLCs may be an attractive target for non-surgical suppression of aneurysm formation due to their expression of degradative proteases.


Subject(s)
Aortic Aneurysm, Abdominal/enzymology , Osteoclasts/enzymology , Animals , Aortic Aneurysm, Abdominal/metabolism , Cathepsin K/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Macrophages/enzymology , Macrophages/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Inbred C57BL , Myeloid Cells/metabolism , Osteoclasts/metabolism , Proteolysis , RAW 264.7 Cells , Tartrate-Resistant Acid Phosphatase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...