Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 17 de 17
1.
J Cell Mol Med ; 28(6): e18175, 2024 Mar.
Article En | MEDLINE | ID: mdl-38451044

The study aimed to identify the biomarkers for predicting coronary atherosclerotic lesions progression in patients with inflammatory bowel disease (IBD). Related transcriptome datasets were seized from Gene Expression Omnibus database. IBD-related modules were identified via Weighted Gene Co-expression Network Analysis. The 'Limma' was applied to screen differentially expressed genes between stable coronary artery disease (CAD) and acute myocardial infarction (AMI). Subsequently, we employed protein-protein interaction (PPI) network and three machine-learning strategies to further screen for candidate hub genes. Application of the receiver operating characteristics curve to quantitatively evaluate candidates to determine key diagnostic biomarkers, followed by a nomogram construction. Ultimately, we performed immune landscape analysis, single-gene GSEA and prediction of target-drugs. 3227 IBD-related module genes and 570 DEGs accounting for AMI were recognized. Intersection yielded 85 shared genes and mostly enriched in immune and inflammatory pathways. After filtering through PPI network and multi-machine learning algorithms, five candidate genes generated. Upon validation, CTSD, CEBPD, CYP27A1 were identified as key diagnostic biomarkers with a superior sensitivity and specificity (AUC > 0.8). Furthermore, all three genes were negatively correlated with CD4+ T cells and positively correlated with neutrophils. Single-gene GSEA highlighted the importance of pathogen invasion, metabolism, immune and inflammation responses during the pathogenesis of AMI. Ten target-drugs were predicted. The discovery of three peripheral blood biomarkers capable of predicting the risk of CAD proceeding into AMI in IBD patients. These identified biomarkers were negatively correlated with CD4+ T cells and positively correlated with neutrophils, indicating a latent therapeutic target.


Coronary Artery Disease , Inflammatory Bowel Diseases , Myocardial Infarction , Humans , Coronary Artery Disease/genetics , Biomarkers , Computational Biology , Inflammatory Bowel Diseases/genetics , Machine Learning
2.
Biomed Pharmacother ; 174: 116451, 2024 May.
Article En | MEDLINE | ID: mdl-38520869

The transcription factor, signal transducer, and stimulator of transcription 3 (STAT3) is a potential target in osteoarthritis (OA) treatment. Although xanthatin (XA), a biologically active substance derived from Xanthium strumarium L, specifically inhibits STAT3 phosphorylation at Tyr705, the mechanism underlying its inhibitory effect on OA progression remains unclear. In this study, our objective was to explore the therapeutic effects exerted by XA on OA and the underlying molecular mechanisms. The effects of XA treatment on mouse OA models subjected to destabilization of the medial meniscus using medial collateral ligament transection, as well as on interleukin-1ß (IL-1ß)-induced mouse chondrocytes, were examined. Histological changes in cartilage and subchondral bone (SCB), as well as changes in the expression levels of osteophytes, cartilage degeneration- and osteoclast differentiation-related factors, and the role of XA-related signaling pathways in human cartilage tissue, were studied using different techniques. XA inhibited STAT3 phosphorylation at Tyr705 and further attenuated the activity of nuclear factor-κB (NF-κB) in chondrocytes and osteoclasts. In vitro, XA administration alleviated pro-inflammatory cytokine release, extracellular matrix catabolism, and RANKL-mediated osteoclast differentiation. In vivo, intraperitoneal injection of XA exerted a protective effect on cartilage degeneration and SCB loss. Similarly, XA exerted a protective effect on human cartilage tissue by inhibiting the STAT3/NF-κB signaling pathway. Overall, our study elucidated the therapeutic potential of XA as a small-molecule inhibitor of STAT3-driven OA progression. This discovery may help enhance innovative clinical interventions against OA.


Chondrocytes , Disease Progression , Furans , Mice, Inbred C57BL , NF-kappa B , Osteoarthritis , STAT3 Transcription Factor , Signal Transduction , Animals , STAT3 Transcription Factor/metabolism , Osteoarthritis/drug therapy , Osteoarthritis/pathology , Osteoarthritis/metabolism , Signal Transduction/drug effects , NF-kappa B/metabolism , Humans , Mice , Chondrocytes/drug effects , Chondrocytes/metabolism , Chondrocytes/pathology , Male , Phosphorylation/drug effects , Disease Models, Animal , Osteoclasts/drug effects , Osteoclasts/metabolism
3.
Front Endocrinol (Lausanne) ; 13: 926934, 2022.
Article En | MEDLINE | ID: mdl-35937813

Osteoarthritis (OA) is a common degenerative joint condition associated with inflammation and characterized by progressive degradation of the articular cartilage and subchondral bone loss in the early stages. Inflammation is closely associated with these two major pathophysiological changes in OA. Velutin, a flavonoid family member, reportedly exerts anti-inflammatory effects. However, the therapeutic effects of velutin in OA have not yet been characterized. In this study, we explore the effects of velutin in an OA mouse model. Histological staining and micro-CT revealed that velutin had a protective effect against cartilage degradation and subchondral bone loss in an OA mouse model generated by surgical destabilization of the medial meniscus (DMM). Additionally, velutin rescued IL-1ß-induced inflammation in chondrocytes and inhibited RANKL-induced osteoclast formation and bone resorption in vitro. Mechanistically, the p38 signaling pathway was found to be implicated in the inhibitory effects of velutin. Our study reveals the dual protective effects of velutin against cartilage degradation and subchondral bone loss by inhibiting the p38 signaling pathway, thereby highlighting velutin as an alternative treatment for OA.


Cartilage, Articular , Osteoarthritis , Animals , Disease Models, Animal , Flavones , Inflammation/pathology , Mice , Osteoarthritis/metabolism , Signal Transduction
4.
Bone Res ; 9(1): 33, 2021 Jul 12.
Article En | MEDLINE | ID: mdl-34253712

Osteoporosis is an osteolytic disorder commonly associated with excessive osteoclast formation. Transcriptional coactivator with PDZ-binding motif (TAZ) is a key downstream effector of the Hippo signaling pathway; it was suggested to be involved in the regulation of bone homeostasis. However, the exact role of TAZ in osteoclasts has not yet been established. In this study, we demonstrated that global knockout and osteoclast-specific knockout of TAZ led to a low-bone mass phenotype due to elevated osteoclast formation, which was further evidenced by in vitro osteoclast formation assays. Moreover, the overexpression of TAZ inhibited RANKL-induced osteoclast formation, whereas silencing of TAZ reduced it. Mechanistically, TAZ bound to TGF-activated kinase 1 (TAK1) and reciprocally inhibited NF-κB signaling, suppressing osteoclast differentiation. Collectively, our findings highlight an essential role of TAZ in the regulation of osteoclastogenesis in osteoporosis and its underlying mechanism.

5.
J Cell Mol Med ; 25(11): 4988-5000, 2021 06.
Article En | MEDLINE | ID: mdl-33939240

Osteoclasts play a critical role in osteoporosis; thus, inhibiting osteoclastogenesis is a therapeutic strategy for osteoporosis. Galangin, a natural bioflavonoid extracted from a traditional Chinese herb, possesses a variety of biological activities, including anti-inflammation and anti-oxidation. However, its effects on osteoporosis have not been elucidated. In this study, we found that galangin treatment dose-dependently decreased osteoclastogenesis in bone marrow-derived macrophages (BMMs). Moreover, during osteoclastogenesis, osteoclast-specific genes, such as tartrate-resistant acid phosphatase (TRAP), cathepsin K (CtsK), ATPase, H + transporting, lysosomal V0 subunit D2 (V-ATPase d2) and dendritic cell-specific transmembrane protein (DC-STAMP), were down-regulated by galangin treatment. Furthermore, the results of the pit formation assay and F-actin ring staining revealed impaired osteoclastic bone resorption in the galangin-treated group compared with that in the control group. Additionally, galangin treatment also inhibited the phosphorylation of p38 and ERK of MAPK signalling pathway, as well as downstream factors of NFATc1, C-Jun and C-Fos. Consistent with our in vitro results, galangin suppressed lipopolysaccharide (LPS)-induced bone resorption via inhibition of osteoclastogenesis. Taken together, our findings provide evidence that galangin is a promising natural compound for the treatment of osteoporosis and may be associated with the inhibition of MAPK and NF-κB signalling pathways.


Flavonoids/pharmacology , Gene Expression Regulation, Neoplastic , Mitogen-Activated Protein Kinases/antagonists & inhibitors , NF-kappa B/antagonists & inhibitors , Osteoclasts/drug effects , Osteogenesis , Osteoporosis/drug therapy , RANK Ligand/metabolism , Animals , Apoptosis , Cell Proliferation , Cells, Cultured , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice , Osteoclasts/cytology , Osteoclasts/metabolism , Osteoporosis/metabolism , Osteoporosis/pathology , Phosphorylation , RANK Ligand/genetics
6.
Drug Des Devel Ther ; 14: 4189-4203, 2020.
Article En | MEDLINE | ID: mdl-33116407

INTRODUCTION: Osteoporosis is a metabolic bone disease characterized by reduced bone quantity and microstructure, typically owing to increased osteoclastogenesis and/or enhanced osteoclastic bone resorption, resulting in uncontrolled bone loss, which primarily affects postmenopausal women. In consideration of the severe side effects of current drugs for osteoporosis, new safe and effective medications are necessary. Pristimerin (Pri), a quinone methide triterpene extracted from Celastraceae and Hippocrateaceae members, exhibits potent antineoplastic and anti-inflammatory effects. However, its effect on osteoclasts remains unknown. MATERIALS AND METHODS: We evaluated the anti-osteoclastogenic and anti-resorptive effect of Pri on bone marrow-derived osteoclasts and its underlying mechanism in vitro. In addition, the protective effect of Pri on ovariectomy model was also explored in vivo. RESULTS: In vitro, Pri inhibited osteoclast differentiation and mature osteoclastic bone resorption in a time- and dose-dependent manner. Further, Pri suppressed the expression of osteoclast-related genes and the activation of key proteins. Pri also inhibited the early activation of ERK, JNK MAPK, and AKT signaling pathways in bone marrow-derived macrophages (BMMs), ultimately inhibiting the induction and activation of the crucial osteoclast transcriptional factor nuclear factor of activated T-cell cytoplasmic 1 (NFATc1). In vivo, consistent with our in vitro data, Pri clearly prevented ovariectomy-induced bone loss. CONCLUSION: Our data showed that Pri inhibits the differentiation and activation of osteoclasts in vitro and in vivo, and could be a promising candidate for treating osteoporosis.


Bone Density Conservation Agents/pharmacology , Bone Resorption/prevention & control , Cell Differentiation/drug effects , Osteoclasts/drug effects , Osteoporosis, Postmenopausal/prevention & control , Ovariectomy , Pentacyclic Triterpenes/pharmacology , Animals , Bone Resorption/diagnostic imaging , Dose-Response Relationship, Drug , Female , Humans , MAP Kinase Signaling System/drug effects , Macrophages/drug effects , Mice , Mice, Inbred C57BL , NFATC Transcription Factors/drug effects , Osteogenesis/drug effects , Signal Transduction/drug effects , X-Ray Microtomography
7.
Biomed Pharmacother ; 131: 110650, 2020 Nov.
Article En | MEDLINE | ID: mdl-32882584

Osteoarthritis (OA) is a common degenerative disease with a series of changes occurring in aging cartilage, such as increased oxidative stress, decreased markers of healthy cartilage and alterations in the autophagy pathway. And increasing evidence indicates that osteoarthritis affects the whole joint, including both cartilage and subchondral bone. The agents that can effectively suppress chondrocyte degradation and subchondral bone deterioration are crucial for the prevention and treatment of OA. Ruboxistaurin (RU), an orally active protein kinase C inhibitor, can reduce macrophage adhesion to endothelial cells and relieve the local inflammation when applicating in diabetes and kinds of aging-related vasculopathy, which were realized by its effects on decreasing inflammatory cytokines' expression and increasing cell anti-oxidative stress ability. However, whether ruboxistaurin protects against OA remains unknown. In this study, we investigated the therapeutic effects of ruboxistaurin in an anterior cruciate ligament transection (ACLT)-induced OA model by preventing the bone mass loss of subchondral bone. We found that ruboxistaurin can effectively alleviate ACLT-induced osteoarthritis, as demonstrated by the phenomenon of correcting pathological bone loss caused by osteoclasts overactivated in the early stage of osteoarthritis and protecting against articular cartilage degeneration. Moreover, we found that ruboxistaurin inhibited osteoclast formation and resorption activity by suppressing the expressions of osteoclast-related genes and (PKCδ/MAPKs) signaling cascade. Taken together, these results show that ruboxistaurin may be a potential therapeutic agent for rescuing abnormal subchondral bone deterioration and cartilage degradation in OA and reverses the vicious cycle related to osteoarthritis.


Bone Density/drug effects , Bone Remodeling/drug effects , Bone Resorption/prevention & control , Indoles/therapeutic use , Maleimides/therapeutic use , Osteoarthritis/prevention & control , Osteoclasts/drug effects , Animals , Bone Density/physiology , Bone Remodeling/physiology , Bone Resorption/metabolism , Bone Resorption/pathology , Cells, Cultured , Disease Progression , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Female , Femur/drug effects , Femur/metabolism , Femur/pathology , Indoles/pharmacology , Male , Maleimides/pharmacology , Mice , Mice, Inbred C57BL , Osteoarthritis/metabolism , Osteoarthritis/pathology , Osteoclasts/metabolism , Osteoclasts/pathology , Osteogenesis/drug effects , Osteogenesis/physiology
8.
J Cell Mol Med ; 24(15): 8452-8465, 2020 08.
Article En | MEDLINE | ID: mdl-32652826

Osteoarthritis (OA) is a common degenerative disease that affects the musculoskeletal structure of the whole joint, which is characterized by progressive destruction of both articular cartilage and subchondral bone. Treatment of the bone pathologies, particularly osteoclast-mediated subchondral bone loss in the early stages of OA, could prevent subsequent cartilage degeneration and progression of OA. In the present study, the PKC inhibitor, Sotrastaurin, was found to inhibit RANKL-induced osteoclast formation in vitro in a dose- and time-dependent manner. In particular, SO exerted its anti-osteoclastic effect predominantly at the early stages of RANKL stimulation, suggesting inhibitory effects on precursor cell fusion. Using mature osteoclasts cultured on bovine bone discs, we showed that SO also exerts anti-resorptive effects on mature osteoclasts bone resorptive function. Mechanistically, SO attenuates the early activation of the p38, ERK and JNK signalling pathways, leeding to impaired induction of crucial osteoclast transcription factors c-Jun, c-Fos and NFATc1. We also showed that SO treatment significantly inhibited the phosphorylation of PKCδ and MARCKS, an upstream regulator of cathepsin K secretion. Finally, in animal studies, SO significantly alleviates the osteochondral pathologies of subchondral bone destruction as well as articular cartilage degeneration following DMM-induced OA, markedly improving OARSI scores. The reduced subchondral bone loss was associated with marked reductions in TRAP(+) osteoclasts in the subchondral bone tissue. Collectively, our data provide evidence for the protective effects of SO against OA by preventing aberrant subchondral bone and articular cartilage changes. Thus, SO demonstrates potential for further development as an alternative therapeutic option against OA.


Bone Resorption/drug therapy , Bone and Bones/drug effects , Osteoarthritis/drug therapy , Osteoclasts/drug effects , Protein Kinase C/antagonists & inhibitors , Pyrroles/pharmacology , Quinazolines/pharmacology , RANK Ligand/metabolism , Animals , Bone Resorption/metabolism , Bone and Bones/metabolism , Cartilage Diseases/drug therapy , Cartilage Diseases/metabolism , Cartilage, Articular/drug effects , Cartilage, Articular/metabolism , Cattle , Cell Differentiation/drug effects , Female , Male , Mice , Mice, Inbred C57BL , Osteoarthritis/metabolism , Osteoclasts/metabolism , Signal Transduction/drug effects
9.
FASEB J ; 33(8): 9100-9115, 2019 08.
Article En | MEDLINE | ID: mdl-31050917

Postmenopausal osteoporosis is the most common form of primary osteoporosis, and the incidence of the condition is rapidly increasing. In consideration of the limitations of current therapeutic options for the treatment of postmenopausal osteoporosis, there is an urgent need to develop safer alternatives. Anacardic acid, a natural phenolic acid compound extracted from cashew nut shell, possesses potent antitumor and anti-inflammatory effects and inhibits NF-κB signaling. However, its effect on osteoclasts remains unknown. This study reports the first evidence for the antiosteoclastogenic and antiresorptive effects of anacardic acid on bone marrow-derived macrophage-derived osteoclasts. Mechanistically, anacardic acid disrupts the phosphorylation of TGF-ß activated kinase 1 and subsequently suppresses multiple receptor activator of NF-κB ligand-induced signaling cascades, ultimately inhibiting the induction and activation of the crucial osteoclast transcriptional factor nuclear factor of activated T-cell cytoplasmic 1. Consistent with cellular results in vitro, anacardic acid treatment improves bone density in the murine model of ovariectomy-induced bone loss. Taken together, our study provides promising evidence for the therapeutic application of anacardic acid as a new potential pharmacological treatment for osteoporosis.-Zhao, K., Jia, Y., Peng, J., Pang, C., Zhang, T., Han, W., Jiang, J., Lu, X., Zhu, J., Qian, Y. Anacardic acid inhibits RANKL-induced osteoclastogenesis in vitro and prevents ovariectomy-induced bone loss in vivo.


Anacardic Acids/pharmacology , Bone Resorption/drug therapy , Osteoclasts/drug effects , RANK Ligand/metabolism , 3T3 Cells , Active Transport, Cell Nucleus , Animals , Bone Resorption/etiology , Bone Resorption/metabolism , Disease Models, Animal , Female , Humans , In Vitro Techniques , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Osteoclasts/metabolism , Osteoclasts/pathology , Osteogenesis/drug effects , Osteoporosis, Postmenopausal/drug therapy , Osteoporosis, Postmenopausal/metabolism , Osteoporosis, Postmenopausal/pathology , Ovariectomy/adverse effects , RAW 264.7 Cells , Signal Transduction/drug effects , Transcription Factor RelA/metabolism
10.
J Cell Physiol ; 234(10): 17549-17560, 2019 08.
Article En | MEDLINE | ID: mdl-30793311

Osteolytic diseases are characterized by an increase in the number and/or activity of bone-resorbing osteoclasts. Identification of natural compounds that can suppress osteoclast formation and function is crucial for the prevention and treatment of osteolytic diseases. Vitexin, a naturally-derived flavonoid extracted from various medicinal plant species, demonstrates a broad range of pharmacological properties including anticancer and anti-inflammatory effects. Here in this study, we showed that vitexin exerts antiosteoclastogenic effects by directly inhibiting receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclast formation and bone resorption in vitro and protected against lipopolysaccharide (LPS)-induced inflammatory osteolysis in vivo. Vitexin suppressed the early activation of ERK and p38 MAPK pathways in response to RANKL thereby attenuating the downstream induction of c-Fos and NFATc1, and abrogating the expression of osteoclast marker genes. Collectively, these results provide evidence for the therapeutic application of vitexin in the treatment of osteoclast-mediated bone lytic diseases.


Apigenin/pharmacology , Osteogenesis/drug effects , Osteolysis/prevention & control , RANK Ligand/antagonists & inhibitors , Actins/metabolism , Animals , Bone Remodeling/drug effects , Bone Remodeling/physiology , Cells, Cultured , In Vitro Techniques , Lipopolysaccharides/toxicity , MAP Kinase Signaling System/drug effects , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Osteoclasts/cytology , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteogenesis/physiology , Osteolysis/chemically induced , Osteolysis/metabolism , RANK Ligand/metabolism
11.
FASEB J ; 33(4): 5399-5410, 2019 04.
Article En | MEDLINE | ID: mdl-30702934

Bone fracture healing is a complex, dynamic process that involves various cell types, with osteoclasts and osteoblasts playing indispensable roles. In this study, we found that psoralen, the main active ingredient in Psoralea corylifolia L. fruit extract, enhanced bone fracture healing through activation of osteoclast and osteoblast activity via the ERK signaling pathway. In detail, psoralen promoted receptor activator of nuclear factor-κB ligand-induced osteoclastogenesis, mRNA expression of osteoclast-specific genes, and osteoclastic bone resorption in primary bone marrow-derived macrophages. Meanwhile, psoralen induced osteogenic differentiation by promoting the mRNA expression of the osteoblast differentiation markers alkaline phosphatase, runt-related transcription factor 2, osterix, and osteocalcin. At the molecular level, psoralen preferentially activated ERK1/2 but not JNK or p38 MAPKs. Further experiments revealed that psoralen-induced osteoclast and osteoblast differentiation was abrogated by a specific inhibitor of phosphorylated ERK. In addition, psoralen accelerated bone fracture healing in a rat tibial fracture model, and the numbers of osteoclasts and osteoblasts were increased in psoralen-treated fracture callus. Taken together, our findings indicate that psoralen accelerates bone fracture healing through activation of osteoclasts and osteoblasts via ERK signaling and has potential as a novel drug in the orthopedic clinic for the treatment of bone fractures.-Zhang, T., Han, W., Zhao, K., Yang, W., Lu, X., Jia, Y., Qin, A., Qian, Y. Psoralen accelerates bone fracture healing by activating both osteoclasts and osteoblasts.


Ficusin/pharmacology , Fracture Healing/drug effects , Osteoblasts/drug effects , Osteoclasts/drug effects , Osteogenesis/drug effects , Animals , Bone Marrow Cells/metabolism , Bone Resorption/drug therapy , Bone Resorption/metabolism , Cell Differentiation/drug effects , Core Binding Factor Alpha 1 Subunit/metabolism , Female , Macrophages/drug effects , Macrophages/metabolism , NF-kappa B/metabolism , NFATC Transcription Factors/metabolism , Osteoblasts/metabolism , Osteoclasts/metabolism , RANK Ligand/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Tartrate-Resistant Acid Phosphatase/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
12.
J Cell Physiol ; 234(3): 2719-2729, 2019 03.
Article En | MEDLINE | ID: mdl-30078209

Excessive bone resorption by osteoclasts (OCs) plays an important role in lytic bone diseases, such as osteoporosis. Although the pharmacological treatment of osteoporosis has been extensively developed, alternative treatments are still needed. Deguelin, a rotenoid isolated from several plant species, is a strong antitumor agent; however, its effect on OCs remains unclear. To the best of our knowledge, this is the first study to report that deguelin inhibits the receptor activator of nuclear factor-κB (NF-κB) ligand (RANKL)-induced osteoclastogenesis, messenger RNA expression of osteoclastic-specific genes, and osteoclastic bone resorption, in primary bone marrow-derived macrophages. At the molecular level, deguelin markedly blocked RANKL-induced osteoclastogenesis by attenuating the phosphorylation of NF-κB p65 and inhibiting p65 nuclear translocation. In addition, deguelin suppressed the downstream expression of nuclear factor of activated T-cell cytoplasmic 1, which is a crucial transcription factor in OC differentiation. Consistent with the in vitro results, deguelin inhibited lipopolysaccharide-induced bone resorption by suppressing osteoclastogenesis. Taken together, our findings reveal that deguelin has antiosteoclastic effects in vitro and in vivo and possesses potential as a new therapeutic option for osteolytic bone diseases.


Bone Resorption/drug therapy , Inflammation/pathology , Osteoclasts/drug effects , Osteogenesis/drug effects , Rotenone/analogs & derivatives , Animals , Bone Diseases, Metabolic/drug therapy , Bone Diseases, Metabolic/metabolism , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Resorption/pathology , Cell Differentiation/drug effects , Gene Expression Regulation/drug effects , Inflammation/drug therapy , Inflammation Mediators/metabolism , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Male , Mice, Inbred C57BL , NFATC Transcription Factors/metabolism , Osteoclasts/metabolism , Rotenone/pharmacology , Signal Transduction/drug effects
13.
J Cell Physiol ; 234(5): 7498-7509, 2019 05.
Article En | MEDLINE | ID: mdl-30471112

Osteoclasts (OCs) are multinuclear giant cells responsible for bone resorption, and an excessive bone resorption by OCs plays an important role in osteoporosis. Commonly used drugs for the treatment of osteoporosis have severe side effects. As such, identification of alternative treatments is essential. Garcinol, a polyisoprenylated benzophenone extracted from the fruit of Garcinia indica, has shown a strong antitumor effect through the nuclear factor-κB (NF-κB) and mitogen-associated protein kinases (MAPK) signaling pathways. However, the role of garcinol in the osteoclastogenesis is still unclear. Here, we demonstrated that garcinol can inhibit the receptor activator of NF-κB ligand (RANKL)-induced osteoclastogenesis, osteoclastogenesis-related gene expression, the f-actin ring, and resorption pit formation. In addition, garcinol abrogated RANKL-induced osteoclastogenesis by attenuating the degradation of the MAPK, NF-κB, and PI3K-AKT signaling pathway as well as downstream factors c-jun, c-fos, and NFATC1. In vivo, suppression of osteoclastogenesis by garcinol was evidenced by marked inhibition of lipopolysaccharide-induced bone resorption. In conclusion, our data demonstrated that garcinol inhibited the RANKL-induced osteoclastogenesis by suppressing the MAPK, NF-κB, and PI3K-AKT signaling pathways and thus has potential as a novel therapeutic option for osteolytic bone diseases.


Osteoclasts/drug effects , Osteogenesis/drug effects , RANK Ligand/metabolism , Terpenes/pharmacology , Animals , Bone Resorption/drug therapy , Bone Resorption/metabolism , Cell Line , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , NFATC Transcription Factors/metabolism , Osteoclasts/metabolism , Osteoporosis/drug therapy , Osteoporosis/metabolism , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-fos/metabolism , Signal Transduction/drug effects
14.
J Cell Mol Med ; 22(10): 5029-5039, 2018 10.
Article En | MEDLINE | ID: mdl-30010244

Osteoclasts (OC) are critical cells responsible for many bone diseases such as osteoporosis. It is of great interest to identify agents that can regulate the activity of OC to treat osteolytic bone diseases. In this study, we found that baicalin exerted a two-way regulatory effect on OC in a concentration-dependent manner in vitro and in vivo. In detail, baicalin at a low concentration (below 1 µmol/L) enhanced OC differentiation and bone resorption, but baicalin at a high concentration (above 2 µmol/L) exhibited inhibitory effects on OC. We demonstrated that baicalin at low concentrations enhanced the mitogen-activated protein kinase (MAPK) (ERK) signalling pathway and activated c-Fos and NFATc1 expression, and thus enhanced gene expression, OC differentiation and bone resorption. However, baicalin at higher levels not only suppressed ERK phosphorylation and c-fos and NFATc1 expression, but also altered the expression of apoptosis-related proteins, and therefore inhibiting OC function. This dual effect was further verified in an LPS-induced mouse calvarial osteolysis model, evidenced by enhanced osteolysis at a lower concentration but reduced bone loss at a higher concentration. Overall, our findings indicate that baicalin exerts dose-dependent effects on OC formation and function. Therefore, caution should be applied when using baicalin to treating OC-related bone diseases.


Bone Resorption/drug therapy , Flavonoids/administration & dosage , Osteoclasts/drug effects , Osteolysis/drug therapy , Animals , Bone Resorption/genetics , Bone Resorption/pathology , Cell Differentiation/drug effects , Gene Expression Regulation, Developmental/genetics , Humans , Lipopolysaccharides/toxicity , MAP Kinase Signaling System/drug effects , Mice , NFATC Transcription Factors/genetics , Osteolysis/chemically induced , Osteolysis/genetics , Osteolysis/pathology , Phosphorylation , Proto-Oncogene Proteins c-fos/genetics , RANK Ligand/genetics , RAW 264.7 Cells , Signal Transduction/drug effects , Skull/drug effects , Skull/growth & development , Skull/pathology
15.
Int J Mol Med ; 40(2): 311-318, 2017 Aug.
Article En | MEDLINE | ID: mdl-28586029

Osteosarcoma is the most common malignant bone tumor. Most patients diagnosed with osteosarcoma are less than 20 years of age. Osteosarcoma cells proliferate rapidly and invade other tissues. At present, neoadjuvant chemotherapy is the primary pharmacodynamic strategy to prevent the progression of osteosarcoma. However, adverse effects of this strategy limit its long­term application. Previous research has shown that fangchinoline exerts antitumor effects on several types of tumor cells; however, its effect on osteosarcoma cells remains unknown. The present study evaluated the effects of fangchinoline on the proliferation, apoptosis, migration and invasion of osteosarcoma cells in vitro and on their tumorigenesis in vivo and determined the possible underlying mechanism of action. Fangchinoline­treated MG63 and U20S cells showed significantly decreased proliferation and significantly increased apoptosis. Fangchinoline markedly suppressed the migration and invasion of the MG63 cells. Fangchinoline­treated MG63 cells showed significantly decreased expression of phosphoinositide 3­kinase (PI3K) and Aktp­Thr308. Moreover, fangchinoline­treated MG63 cells showed downregulated expression of cyclin D1 and matrix metalloproteinase 2 and 9, which act downstream of PI3K, and upregulated expression of caspase­3 and caspase­8. Furthermore, fangchinoline suppressed the growth of subcutaneous osteosarcoma tumors in Balb/c mice subcutaneously injected with osteosarcoma cells. These findings suggest that fangchinoline inhibits the progression of osteosarcoma by suppressing the proliferation, migration and invasion and by accelerating the apoptosis of osteosarcoma cells. In addition, our results suggest that the mechanism underlying the antitumor effects of fangchinoline involve the inhibition of PI3K and its downstream signaling pathways.


Antineoplastic Agents, Phytogenic/pharmacology , Benzylisoquinolines/pharmacology , Bone Neoplasms/drug therapy , Cell Proliferation/drug effects , Osteosarcoma/drug therapy , Phosphoinositide-3 Kinase Inhibitors , Signal Transduction/drug effects , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Benzylisoquinolines/therapeutic use , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Movement/drug effects , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Female , Humans , Mice, Inbred BALB C , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/prevention & control , Osteosarcoma/metabolism , Osteosarcoma/pathology , Phosphatidylinositol 3-Kinases/metabolism
16.
Oncol Rep ; 37(1): 435-441, 2017 Jan.
Article En | MEDLINE | ID: mdl-27840963

Osteosarcoma is the most common malignant bone tumor that frequently affects adolescents. Osteosarcoma cells tend to proliferate and invade other tissues such as those of the lungs. Currently, neoadjuvant chemotherapy is the primary strategy to prevent tumor progression. However, its adverse effects result in poor long-term outcomes. Previous research has shown that galangin exhibits antitumor properties on several types of cancer cells; however its effect on osteosarcoma cells is yet unknown. The aims of this study were to evaluate the effects of galangin on the proliferation, apoptosis, migration, and invasion of osteosarcoma cells and to explore the underlying mechanisms. We found that the proliferation of MG63 and U20S osteosarcoma cells decreased significantly, while the apoptosis of MG63 cells accelerated significantly after exposure to galangin. In addition, the migration and invasion of MG63 cells were significantly inhibited by galangin. Moreover, phosphoinositide 3-kinase (PI3K) and Aktp-Thr308 expression levels were found to be significantly lower in galangin-treated MG63 cells than in the control cells, and the protein expression levels of their downstream regulators cyclin D1 and matrix metalloproteinase 2/9 were also downregulated in galangin-treated groups, while those of p27Kip1, caspase-3, and caspase-8 were upregulated. These findings suggest that galangin suppresses osteosarcoma cells by inhibiting their proliferation and invasion and accelerating their apoptosis, and the mechanism may be associated with the inhibition of PI3K and its downstream signaling pathway.


Bone Neoplasms/prevention & control , Flavonoids/pharmacology , Osteosarcoma/prevention & control , Apoptosis/drug effects , Bone Neoplasms/pathology , Caspase 3/metabolism , Caspase 8/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin D1/metabolism , Humans , Matrix Metalloproteinase 2/metabolism , Osteosarcoma/pathology , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects
17.
Sci Rep ; 6: 36330, 2016 10 31.
Article En | MEDLINE | ID: mdl-27796345

Bone callus, generated during fracture healing, is commonly discarded during surgical procedures. The aim of this study was to investigate the osteogenic potential of bone callus and its possible use as autograft material for patients needing bone grafts. Histology, immunohistochemistry, micro-computed tomography, and biomechanics were performed to examine osteogenic cells, osteoinductive factors, and the osteoconductive structure of bone callus. Alkaline phosphatase-positive osteoblasts, osteoinductive factors (including BMP2, FGF2, TGFB1, and IGF1), and a porous structure were found in bone callus. Early-stage callus (within 3 months after fracture) presented significantly improved osteogenic properties compared to medium- (3-9 months) and late-stage (longer than 9 months) callus. The results revealed that bone callus induced new bone formation in a nude mouse model. Early-stage callus showed better performance to medium- and late-stage callus in the induction of new bone formation at both 8 and 12 weeks. These findings indicated that bone callus, especially early-stage callus, possesses osteogenic potential and can potentially serve as an alternative source of material for bone grafts.


Bone Transplantation/methods , Bony Callus/cytology , Osteogenesis , Adult , Alkaline Phosphatase/metabolism , Animals , Bony Callus/metabolism , Bony Callus/transplantation , Female , Humans , Male , Mice , Mice, Nude , Middle Aged , Osteoblasts/cytology , Osteoblasts/metabolism , X-Ray Microtomography
...