Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.540
Filter
1.
FASEB J ; 38(13): e23779, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38967255

ABSTRACT

Epigenetic modifications affect cell differentiation via transcriptional regulation. G9a/EHMT2 is an important epigenetic modifier that catalyzes the methylation of histone 3 lysine 9 (H3K9) and interacts with various nuclear proteins. In this study, we investigated the role of G9a in osteoclast differentiation. When we deleted G9a by infection of Cre-expressing adenovirus into bone marrow macrophages (BMMs) from G9afl/fl (Ehmt2fl/fl) and induced osteoclastic differentiation by the addition of macrophage colony-stimulating factor (M-CSF) and receptor activator of NF-κB ligand (RANKL), the number of TRAP-positive multinucleated osteoclasts significantly increased compared with control. Furthermore, the mRNA expression of osteoclast markers, TRAP, and cathepsin K, and to a lesser extent, NFATc1, a critical transcription factor, increased in G9a KO cells. Infection of wild-type (WT) G9a-expressing adenovirus in G9a KO cells restored the number of TRAP-positive multinucleated cells. In G9a KO cells, increased nuclear accumulation of NFATc1 protein and decreased H3K9me2 accumulation were observed. Furthermore, ChIP experiments revealed that NFATc1 binding to its target, Ctsk promoter, was enhanced by G9a deletion. For in vivo experiments, we created G9a conditional knock-out (cKO) mice by crossing G9afl/fl mice with Rank Cre/+ (Tnfrsf11aCre/+) mice, in which G9a is deleted in osteoclast lineage cells. The trabecular bone volume was significantly reduced in female G9a cKO mice. The serum concentration of the C-terminal telopeptide of type I collagen (CTX), a bone-resorbing indicator, was higher in G9a cKO mice. In addition, osteoclasts differentiated from G9a cKO BMMs exhibited greater bone-resorbing activity. Our findings suggest that G9a plays a repressive role in osteoclastogenesis by modulating NFATc1 function.


Subject(s)
Bone Resorption , Cell Differentiation , Histone-Lysine N-Methyltransferase , NFATC Transcription Factors , Osteoclasts , Osteogenesis , Animals , NFATC Transcription Factors/metabolism , NFATC Transcription Factors/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histone-Lysine N-Methyltransferase/genetics , Mice , Osteoclasts/metabolism , Bone Resorption/metabolism , Osteogenesis/physiology , Mice, Knockout , RANK Ligand/metabolism , Mice, Inbred C57BL , Cells, Cultured
2.
Biochem Biophys Res Commun ; 727: 150317, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-38959733

ABSTRACT

Abnormalities in osteoclastic generation or activity disrupt bone homeostasis and are highly involved in many pathologic bone-related diseases, including rheumatoid arthritis, osteopetrosis, and osteoporosis. Control of osteoclast-mediated bone resorption is crucial for treating these bone diseases. However, the mechanisms of control of osteoclastogenesis are incompletely understood. In this study, we identified that inosine 5'-monophosphate dehydrogenase type II (Impdh2) positively regulates bone resorption. By histomorphometric analysis, Impdh2 deletion in mouse myeloid lineage cells (Impdh2LysM-/- mice) showed a high bone mass due to the reduced osteoclast number. qPCR and western blotting results demonstrated that the expression of osteoclast marker genes, including Nfatc1, Ctsk, Calcr, Acp5, Dcstamp, and Atp6v0d2, was significantly decreased in the Impdh2LysM-/- mice. Furthermore, the Impdh inhibitor MPA treatment inhibited osteoclast differentiation and induced Impdh2-cytoophidia formation. The ability of osteoclast differentiation was recovered after MPA deprivation. Interestingly, genome-wide analysis revealed that the osteoclastic mitochondrial biogenesis and functions, such as oxidative phosphorylation, were impaired in the Impdh2LysM-/- mice. Moreover, the deletion of Impdh2 alleviated ovariectomy-induced bone loss. In conclusion, our findings revealed a previously unrecognized function of Impdh2, suggesting that Impdh2-mediated mechanisms represent therapeutic targets for osteolytic diseases.


Subject(s)
IMP Dehydrogenase , Mitochondria , Osteoclasts , Osteogenesis , Osteoporosis , Ovariectomy , Oxidative Phosphorylation , Animals , Osteoporosis/metabolism , Osteoporosis/etiology , Osteoporosis/genetics , Osteoporosis/pathology , Mice , Female , Osteoclasts/metabolism , Osteoclasts/pathology , Mitochondria/metabolism , Mitochondria/pathology , IMP Dehydrogenase/metabolism , IMP Dehydrogenase/genetics , IMP Dehydrogenase/deficiency , Mice, Knockout , Mice, Inbred C57BL , Cell Differentiation , Bone Resorption/metabolism , Bone Resorption/genetics , Bone Resorption/pathology , Bone Resorption/etiology
3.
Immunopharmacol Immunotoxicol ; 46(4): 538-549, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39013842

ABSTRACT

OBJECTIVE: Rheumatoid arthritis (RA), a chronic inflammatory disease, is characterized by joint swelling, cartilage erosion, and bone destruction. This study investigated the therapeutic efficacy of Carnosic acid (CA), a natural compound with anti-inflammatory and antioxidant properties, in an adjuvant-induced arthritis model. METHODS: Paw swelling and arthritis index were measured. Oxidative stress markers, including lipid peroxidation and antioxidant enzyme levels, were assessed. Synovial tissue was analyzed for pro-inflammatory markers using real-time Q-PCR and Western blotting. The expression of mPGES-1 was determined by Western blotting. Peripheral neuropathic pain was assessed using cold and mechanical allodynia tests. Bone loss was quantitatively assessed through microcomputed tomography (µCT) scanning of femurs and X-ray radiography. Indomethacin-induced gastric ulcers were evaluated. Molecular docking studies were conducted to analyze the binding affinity of CA to mPGES-1. RESULTS: The CA treatment not only demonstrated a significant reduction in joint inflammation and paw swelling but also mitigated oxidative stress and improved the antioxidant defence system. CA inhibited microsomal prostaglandin E synthase-1 (mPGES-1) expression and the expression of pro-inflammatory molecules such as inducible nitric oxide synthase (iNOS) and cyclooxygenases-2 (COX-2), thus attenuating the arthritis symptoms without severe gastrointestinal side effects. Additionally, it inhibited the expression of pro-inflammatory molecules such as iNOS and COX-2, contributing to the reduction of arthritis symptoms. Notably, CA treatment prevented the common side effects of traditional RA treatments like corticosteroids and non-steroidal anti-inflammatory drugs (NSAIDs), including weight loss, bone degradation, and gastric ulcers. CONCLUSIONS: These findings suggest that CA, through specific enzyme inhibition, offers a compelling alternative therapeutic approach for RA. Further research is warranted to explore the potential of CA in other arthritis models and its suitability for human RA treatment.


CA significantly reduces inflammation in FCA induced arthritis model.CA treatment inhibits key pro-inflammatory molecules, including mPGES-1 and COX-2In silico docking studies confirm the affinity of CA to mPGES-1.CA prevents bone loss and avoids side effects seen with standard treatments.Antioxidant properties of CA counteract oxidative stress related to chronic inflammation.


Subject(s)
Abietanes , Arthritis, Experimental , Cyclooxygenase 2 , Prostaglandin-E Synthases , Rats, Sprague-Dawley , Animals , Arthritis, Experimental/drug therapy , Arthritis, Experimental/pathology , Arthritis, Experimental/metabolism , Arthritis, Experimental/chemically induced , Male , Prostaglandin-E Synthases/metabolism , Prostaglandin-E Synthases/genetics , Rats , Abietanes/pharmacology , Cyclooxygenase 2/metabolism , Bone Resorption/drug therapy , Bone Resorption/metabolism , Bone Resorption/pathology , Molecular Docking Simulation , Oxidative Stress/drug effects
4.
Anticancer Res ; 44(8): 3365-3374, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39060037

ABSTRACT

BACKGROUND/AIM: Cancer-associated fibroblasts (CAFs) have recently been suggested as critical cellular components of bone invasion in oral squamous cell carcinoma (OSCC). However, the underlying molecular mechanisms and subtypes related to their bone-invasive function are unclear. This study investigated the implications of thymidine phosphorylase (TP)-positive CAFs (TP+CAFs) in OSCC bone invasion. MATERIALS AND METHODS: TP expression was determined in 116 patients with OSCC using immunohistochemistry. The influence of TP expression on the biological behavior of CAFs was investigated in vitro. The possible impact of TP+CAFs on bone invasion in OSCC was further evaluated using patient-derived xenograft (PDX) mouse models. RESULTS: In bone-invasive OSCC tissues, TP+CAFs were mainly distributed on the surface of resorbed bone tissue rather than on the tumor side. High levels of TP+CAFs were significantly associated with higher T-stage, bone invasion, and worse overall survival and recurrence-free survival in our study cohort. Recombinant human TP promoted the proliferative and invasive abilities of CAFs and increased matrix metalloproteinase-9 mRNA expression in vitro, related to bone resorption. In the PDX mouse models, TP+CAFs were found in early bone resorption on the surface of resorbed bony tissues. Bone resorption occurred more frequently in the PDX models with TP+CAFs than in those without. CONCLUSION: TP+CAFs were significantly associated with bone invasion and the prognosis of OSCC. This study provides insights into cellular and molecular targets for the early diagnosis and treatment of bone-invasive OSCC.


Subject(s)
Cancer-Associated Fibroblasts , Carcinoma, Squamous Cell , Mouth Neoplasms , Neoplasm Invasiveness , Thymidine Phosphorylase , Humans , Animals , Mouth Neoplasms/pathology , Mouth Neoplasms/metabolism , Mouth Neoplasms/genetics , Female , Male , Mice , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Thymidine Phosphorylase/metabolism , Thymidine Phosphorylase/genetics , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/genetics , Middle Aged , Bone Neoplasms/pathology , Bone Neoplasms/metabolism , Bone Neoplasms/genetics , Cell Line, Tumor , Aged , Cell Proliferation , Matrix Metalloproteinase 9/metabolism , Matrix Metalloproteinase 9/genetics , Prognosis , Xenograft Model Antitumor Assays , Bone Resorption/pathology , Bone Resorption/metabolism
5.
Chem Biol Interact ; 399: 111135, 2024 Aug 25.
Article in English | MEDLINE | ID: mdl-38971422

ABSTRACT

Iron overload is a risk factor for osteoporosis due to its oxidative toxicity. Previous studies have demonstrated that an excessive amount of iron increases osteocyte apoptosis and receptor activator of nuclear factor κ-B ligand (RANKL) production, which stimulates osteoclast differentiation in vitro. However, the effects of exogenous iron supplementation-induced iron overload on osteocytes in vivo and its role in iron overload-induced bone loss are unknown. This work aimed to develop an iron overloaded murine model of C57BL/6 mice by intraperitoneal administration of iron dextran for two months. The iron levels in various organs, bone, and serum, as well as the microstructure and strength of bone, apoptosis of osteocytes, oxidative stress in bone tissue, and bone formation and resorption, were assessed. The results showed that 2 months of exogenous iron supplementation significantly increased iron levels in the liver, spleen, kidney, bone tissue, and serum. Iron overload negatively affected bone microstructure and strength. Osteocyte apoptosis and empty lacunae rate were elevated by exogenous iron. Iron overload upregulated RANKL expression but had no significant impact on osteoprotegerin (OPG) and sclerostin levels. Static and dynamic histologic analyses and serum biochemical assay showed that iron overload increased bone resorption without significantly affecting bone formation. Exogenous iron promoted oxidative stress in osteocytes in vivo and in vitro. Additional supplementation of iron chelator (deferoxamine) or N-acetyl-l-cysteine (NAC) partially alleviated bone loss, osteocyte apoptosis, osteoclast formation, and oxidative stress due to iron overload. These findings, in line with prior in vitro studies, suggest that exogenous iron supplementation induces osteoclastogenesis and osteoporosis by promoting osteocyte apoptosis and RANKL production via oxidative stress.


Subject(s)
Apoptosis , Bone Resorption , Iron , Mice, Inbred C57BL , Osteocytes , Oxidative Stress , RANK Ligand , Animals , Osteocytes/drug effects , Osteocytes/metabolism , Oxidative Stress/drug effects , Apoptosis/drug effects , RANK Ligand/metabolism , Bone Resorption/metabolism , Bone Resorption/pathology , Mice , Iron/metabolism , Disease Models, Animal , Male , Iron Overload/metabolism , Iron Overload/pathology , Iron Overload/chemically induced , Osteoprotegerin/metabolism , Acetylcysteine/pharmacology , Adaptor Proteins, Signal Transducing
6.
Bone Res ; 12(1): 40, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38987568

ABSTRACT

Efficient cellular fusion of mononuclear precursors is the prerequisite for the generation of fully functional multinucleated bone-resorbing osteoclasts. However, the exact molecular factors and mechanisms controlling osteoclast fusion remain incompletely understood. Here we identify RANKL-mediated activation of caspase-8 as early key event during osteoclast fusion. Single cell RNA sequencing-based analyses suggested that activation of parts of the apoptotic machinery accompanied the differentiation of osteoclast precursors into mature multinucleated osteoclasts. A subsequent characterization of osteoclast precursors confirmed that RANKL-mediated activation of caspase-8 promoted the non-apoptotic cleavage and activation of downstream effector caspases that translocated to the plasma membrane where they triggered activation of the phospholipid scramblase Xkr8. Xkr8-mediated exposure of phosphatidylserine, in turn, aided cellular fusion of osteoclast precursors and thereby allowed generation of functional multinucleated osteoclast syncytia and initiation of bone resorption. Pharmacological blockage or genetic deletion of caspase-8 accordingly interfered with fusion of osteoclasts and bone resorption resulting in increased bone mass in mice carrying a conditional deletion of caspase-8 in mononuclear osteoclast precursors. These data identify a novel pathway controlling osteoclast biology and bone turnover with the potential to serve as target for therapeutic intervention during diseases characterized by pathologic osteoclast-mediated bone loss. Proposed model of osteoclast fusion regulated by caspase-8 activation and PS exposure. RANK/RANK-L interaction. Activation of procaspase-8 into caspase-8. Caspase-8 activates caspase-3. Active capase-3 cleaves Xkr8. Local PS exposure is induced. Exposed PS is recognized by the fusion partner. FUSION. PS is re-internalized.


Subject(s)
Caspase 8 , Cell Fusion , Osteoclasts , Phosphatidylserines , Phospholipid Transfer Proteins , Caspase 8/metabolism , Caspase 8/genetics , Animals , Osteoclasts/metabolism , Phosphatidylserines/metabolism , Phospholipid Transfer Proteins/metabolism , Phospholipid Transfer Proteins/genetics , Mice , Mice, Inbred C57BL , Bone Resorption/metabolism , Bone Resorption/pathology , Bone Resorption/genetics , Cell Differentiation , RANK Ligand/metabolism
7.
Cell Mol Biol Lett ; 29(1): 100, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38977961

ABSTRACT

OBJECTIVE: Osteoporosis is a global health issue characterized by decreased bone mass and microstructural degradation, leading to an increased risk of fractures. This study aims to explore the molecular mechanism by which P2X7 receptors influence osteoclast formation and bone resorption through the PI3K-Akt-GSK3ß signaling pathway. METHODS: An osteoporosis mouse model was generated through ovariectomy (OVX) in normal C57BL/6 and P2X7f/f; LysM-cre mice. Osteoclasts were isolated for transcriptomic analysis, and differentially expressed genes were selected for functional enrichment analysis. Metabolite analysis was performed using liquid chromatography-tandem mass spectrometry (LC-MS/MS), and multivariate statistical analysis and pattern recognition were used to identify differential lipid metabolism markers and their distribution. Bioinformatics analyses were conducted using the Encyclopedia of Genes and Genomes database and the MetaboAnalyst database to assess potential biomarkers and create a metabolic pathway map. Osteoclast precursor cells were used for in vitro cell experiments, evaluating cell viability and proliferation using the Cell Counting Kit 8 (CCK-8) assay. Osteoclast precursor cells were induced to differentiate into osteoclasts using macrophage colony-stimulating factor (M-CSF) and receptor activator of nuclear factor kappa-beta ligand (RANKL), and tartrate-resistant acid phosphatase (TRAP) staining was performed to compare differentiation morphology, size, and quantity between different groups. Western blot analysis was used to assess the expression of differentiation markers, fusion gene markers, and bone resorption ability markers in osteoclasts. Immunofluorescence staining was employed to examine the spatial distribution and quantity of osteoclast cell skeletons, P2X7 protein, and cell nuclei, while pit assay was used to evaluate osteoclast bone resorption ability. Finally, in vivo animal experiments, including micro computed tomography (micro-CT), hematoxylin and eosin (HE) staining, TRAP staining, and immunohistochemistry, were conducted to observe bone tissue morphology, osteoclast differentiation, and the phosphorylation level of the PI3K-Akt-GSK3ß signaling pathway. RESULTS: Transcriptomic and metabolomic data collectively reveal that the P2X7 receptor can impact the pathogenesis of osteoporosis through the PI3K-Akt-GSK3ß signaling pathway. Subsequent in vitro experiments showed that cells in the Sh-P2X7 + Recilisib group exhibited increased proliferative activity (1.15 versus 0.59), higher absorbance levels (0.68 versus 0.34), and a significant increase in resorption pit area (13.94 versus 3.50). Expression levels of osteoclast differentiation-related proteins MMP-9, CK, and NFATc1 were markedly elevated (MMP-9: 1.72 versus 0.96; CK: 2.54 versus 0.95; NFATc1: 3.05 versus 0.95), along with increased fluorescent intensity of F-actin rings. In contrast, the OE-P2X7 + LY294002 group showed decreased proliferative activity (0.64 versus 1.29), reduced absorbance (0.34 versus 0.82), and a significant decrease in resorption pit area (5.01 versus 14.96), accompanied by weakened expression of MMP-9, CK, and NFATc1 (MMP-9: 1.14 versus 1.79; CK: 1.26 versus 2.75; NFATc1: 1.17 versus 2.90) and decreased F-actin fluorescent intensity. Furthermore, in vivo animal experiments demonstrated that compared with the wild type (WT) + Sham group, mice in the WT + OVX group exhibited significantly increased levels of CTX and NTX in serum (CTX: 587.17 versus 129.33; NTX: 386.00 versus 98.83), a notable decrease in calcium deposition (19.67 versus 53.83), significant reduction in bone density, increased trabecular separation, and lowered bone mineral density (BMD). When compared with the KO + OVX group, mice in the KO + OVX + recilisib group showed a substantial increase in CTX and NTX levels in serum (CTX: 503.50 versus 209.83; NTX: 339.83 versus 127.00), further reduction in calcium deposition (29.67 versus 45.33), as well as decreased bone density, increased trabecular separation, and reduced BMD. CONCLUSION: P2X7 receptors positively regulate osteoclast formation and bone resorption by activating the PI3K-Akt-GSK3ß signaling pathway.


Subject(s)
Bone Resorption , Cell Differentiation , Glycogen Synthase Kinase 3 beta , Mice, Inbred C57BL , Osteoclasts , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Receptors, Purinergic P2X7 , Signal Transduction , Animals , Female , Mice , Bone Resorption/metabolism , Bone Resorption/genetics , Bone Resorption/pathology , Cell Differentiation/genetics , Glycogen Synthase Kinase 3 beta/metabolism , Glycogen Synthase Kinase 3 beta/genetics , Osteoclasts/metabolism , Osteoporosis/metabolism , Osteoporosis/genetics , Osteoporosis/pathology , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-akt/genetics , RANK Ligand/metabolism , RANK Ligand/genetics , Receptors, Purinergic P2X7/metabolism , Receptors, Purinergic P2X7/genetics
8.
PeerJ ; 12: e17722, 2024.
Article in English | MEDLINE | ID: mdl-39006031

ABSTRACT

Background: Osteoporosis is a disease associated with bone resorption, characterized primarily by the excessive activation of osteoclasts. Ginkgetin is a compound purified from natural ginkgo leaves which has various biological properties, including anti-inflammation, antioxidant, and anti-tumor effects. This study investigated the bone-protective effects of ginkgetin in ovariectomized (OVX) mice and explored their potential signaling pathway in inhibiting osteoclastogenesis in a mouse model of osteoporosis. Methods: Biochemical assays were performed to assess the levels of Ca, ALP, and P in the blood. Micro CT scanning was used to evaluate the impact of ginkgetin on bone loss in mice. RT-PCR was employed to detect the expression of osteoclast-related genes (ctsk, c-fos, trap) in their femoral tissue. Hematoxylin and eosin (H&E) staining was utilized to assess the histopathological changes in femoral tissue due to ginkgetin. The TRAP staining was used to evaluate the impact of ginkgetin osteoclast generation in vivo. Western blot analysis was conducted to investigate the effect of ginkgetin on the expression of p-NF-κB p65 and IκBα proteins in mice. Results: Our findings indicate that ginkgetin may increase the serum levels of ALP and P, while decreasing the serum level of Ca in OVX mice. H&E staining and micro CT scanning results suggest that ginkgetin can inhibit bone loss in OVX mice. The TRAP staining results showed ginkgetin suppresses the generation of osteoclasts in OVX mice. RT-PCR results demonstrate that ginkgetin downregulate the expression of osteoclast-related genes (ctsk, c-fos, trap) in the femoral tissue of mice, and this effect is dose-dependent. Western blot analysis results reveal that ginkgetin can inhibit the expression of p-NF-κB p65 and IκBα proteins in mice. Conclusion: Ginkgetin can impact osteoclast formation and activation in OVX mice by inhibiting the NF-κB/IκBα signaling pathway, thereby attenuating bone loss in mice.


Subject(s)
Biflavonoids , NF-kappa B , Osteoclasts , Signal Transduction , Animals , Biflavonoids/pharmacology , Biflavonoids/therapeutic use , Signal Transduction/drug effects , Mice , NF-kappa B/metabolism , Female , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoporosis/drug therapy , Osteoporosis/metabolism , Osteoporosis/pathology , Ovariectomy , Disease Models, Animal , Bone Resorption/drug therapy , Bone Resorption/metabolism , Bone Resorption/prevention & control , Bone Resorption/pathology , X-Ray Microtomography , NF-KappaB Inhibitor alpha/metabolism , Mice, Inbred C57BL
9.
Int J Mol Sci ; 25(11)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38892322

ABSTRACT

Estrogen (17ß-estradiol) deficiency post-menopause alters bone homeostasis whereby bone resorption by osteoclasts exceeds bone formation by osteoblasts, leading to osteoporosis in females. We established an in vitro model to examine the consequences of estrogen withdrawal (E2-WD) on osteoclasts derived from the mouse macrophage RAW 264.7 cell line and utilized it to investigate the mechanism behind the enhanced osteoclast activity post-menopause. We found that a greater population of osteoclasts that underwent E2-WD contained a podosome belt necessary for osteoclasts to adhere and resorb bone and possessed elevated resorptive activity compared to osteoclasts exposed to estrogen (E2) continuously. Our results show that compared to osteoclasts that received E2 continuously, those that underwent E2-WD had a faster rate of microtubule (MT) growth, reduced RhoA activation, and shorter podosome lifespan. Thus, altered podosome and MT dynamics induced by the withdrawal of estrogen supports podosome belt assembly/stability in osteoclasts, which may explain their enhanced bone resorption activity.


Subject(s)
Bone Resorption , Estrogens , Osteoclasts , Animals , Osteoclasts/metabolism , Osteoclasts/cytology , Mice , RAW 264.7 Cells , Estrogens/metabolism , Estrogens/pharmacology , Bone Resorption/metabolism , Podosomes/metabolism , Microtubules/metabolism , Female , rhoA GTP-Binding Protein/metabolism , Estradiol/pharmacology , Estradiol/metabolism , Cell Culture Techniques
10.
Int J Mol Sci ; 25(11)2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38892426

ABSTRACT

Globally, osteoporosis is the most common systemic skeletal disease. There are many factors that influence osteoporosis' development and progression. During the pathogenesis of this disease, bone turnover is imbalanced between resorption and the formation of bone tissue. A growing interest has been devoted to the role that microRNA (miRNA) plays in osteoporosis regulation. A microRNA (miRNA) is a group of small single-stranded RNA molecules involved in regulating gene expression in eukaryotic organisms. As microRNAs (miRNAs) are key regulators of gene expression and can modulate processes related to bone metabolism, they have become increasingly important for studying osteoporosis pathogenesis. The available research suggests that miRNAs play an important role in regulating processes associated with bone metabolism, especially by influencing bone resorption and synthesis. Furthermore, microRNAs can also serve as potential therapeutic targets for osteoporosis, besides being a rapid and specific biomarker.


Subject(s)
MicroRNAs , Osteoporosis , Osteoporosis/genetics , Osteoporosis/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Humans , Animals , Gene Expression Regulation , Biomarkers , Bone and Bones/metabolism , Bone Resorption/genetics , Bone Resorption/metabolism
11.
Nat Commun ; 15(1): 5094, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38877020

ABSTRACT

Interactions between osteolineage cells and myeloid cells play important roles in maintaining skeletal homeostasis. Herein, we find that osteolineage cells transfer mitochondria to myeloid cells. Impairment of the transfer of mitochondria by deleting MIRO1 in osteolineage cells leads to increased myeloid cell commitment toward osteoclastic lineage cells and promotes bone resorption. In detail, impaired mitochondrial transfer from osteolineage cells alters glutathione metabolism and protects osteoclastic lineage cells from ferroptosis, thus promoting osteoclast activities. Furthermore, mitochondrial transfer from osteolineage cells to myeloid cells is involved in the regulation of glucocorticoid-induced osteoporosis, and glutathione depletion alleviates the progression of glucocorticoid-induced osteoporosis. These findings reveal an unappreciated mechanism underlying the interaction between osteolineage cells and myeloid cells to regulate skeletal metabolic homeostasis and provide insights into glucocorticoid-induced osteoporosis progression.


Subject(s)
Bone Resorption , Ferroptosis , Mitochondria , Myeloid Cells , Osteoclasts , Osteoporosis , Animals , Mitochondria/metabolism , Bone Resorption/metabolism , Bone Resorption/pathology , Osteoclasts/metabolism , Myeloid Cells/metabolism , Osteoporosis/metabolism , Osteoporosis/pathology , Mice , Glucocorticoids/metabolism , Glutathione/metabolism , Mice, Inbred C57BL , Cell Differentiation , Mice, Knockout , Humans , Male
12.
Biochem Pharmacol ; 226: 116391, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38914317

ABSTRACT

Inhibition of excessive osteoclastic activity is an efficient therapeutic strategy for many bone diseases induced by increased bone resorption, such as osteoporosis. BMS-582949, a clinical p38α inhibitor, is a promising drug in Phase II studies for treating rheumatoid arthritis. However, its function on bone resorption is largely unknown. In this study, we find that BMS-582949 represses RANKL-induced osteoclast differentiation in a dose-dependent manner. Moreover, BMS-582949 inhibits osteoclastic F-actin ring formation and osteoclast-specific gene expression. Mechanically, BMS-582949 treatment attenuates RANKL-mediated osteoclastogenesis through mitogen-activated protein kinases (MAPKs) and protein kinase B (AKT) signaling pathways without disturbing nuclear factor-κB (NF-κB) signaling. Interestingly, BMS-582949 impairs osteoclastic mitochondrial biogenesis and functions, such as oxidative phosphorylation (OXPHOS). Furthermore, BMS-582949 administration prevents bone loss in ovariectomized mouse mode by inhibiting both bone resorption and bone formation in vivo. Taken together, these findings indicate that BMS-582949 may be a potential and effective drug for the therapy of osteolytic diseases.


Subject(s)
Mice, Inbred C57BL , Osteoclasts , Osteogenesis , Ovariectomy , p38 Mitogen-Activated Protein Kinases , Animals , Mice , Ovariectomy/adverse effects , Female , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteogenesis/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Bone Resorption/prevention & control , Bone Resorption/drug therapy , Bone Resorption/metabolism , RAW 264.7 Cells , Protein Kinase Inhibitors/pharmacology , Bone Remodeling/drug effects , RANK Ligand/metabolism , Cell Differentiation/drug effects , Dose-Response Relationship, Drug
13.
Pflugers Arch ; 476(8): 1289-1302, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38833170

ABSTRACT

Osteoclasts are multinucleated cells of hematopoietic origin, with a pivotal role in bone development and remodeling. Failure in osteoclast differentiation and activation leads to various bone disorders; thus, attention has focused on a search of molecules involved in osteoclast regulatory pathways. Caspase-8 appears to be an interesting candidate for further exploration, due to its potential function in bone development and homeostasis. Mouse bone marrow cells were differentiated into osteoclasts by RANKL stimulation. Increased activation of caspase-8 and its downstream executioner caspases (caspase-3 and caspase-6) was found during osteoclastogenesis. Subsequent inhibition of caspase-8, caspase-3, or caspase-6, respectively, during osteoclast differentiation showed distinct changes in the formation of TRAP-positive multinucleated cells and reduced expression of osteoclast markers including Acp5, Ctsk, Dcstamp, and Mmp9. Analysis of bone matrix resorption confirmed significantly reduced osteoclast function after caspase inhibition. The results clearly showed the role of caspases in the proper development of osteoclasts and contributed new knowledge about non-apoptotic function of caspases.


Subject(s)
Bone Marrow Cells , Caspase Inhibitors , Cell Differentiation , Osteoclasts , RANK Ligand , Animals , Mice , Bone Marrow Cells/metabolism , Bone Resorption/metabolism , Caspase 3/metabolism , Caspase 6/metabolism , Caspase 8/metabolism , Caspase Inhibitors/pharmacology , Cells, Cultured , Mice, Inbred C57BL , Osteoclasts/metabolism , RANK Ligand/metabolism , Tartrate-Resistant Acid Phosphatase/metabolism
14.
Bone ; 186: 117163, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38857854

ABSTRACT

Osteocytes engage in bone resorption and mineralization surrounding their expansive lacunar-canalicular system (LCS) through peri-LCS turnover. However, fundamental questions persist about where, when, and how often osteocytes engage in peri-LCS turnover and how these processes change with aging. Furthermore, whether peri-LCS turnover is associated with natural variation in cortical tissue strain remains unexplored. To address these questions, we utilized confocal scanning microscopy, immunohistochemistry, and scanning electron microscopy to characterize osteocyte peri-LCS turnover in the cortical (mid-diaphysis) and cancellous (metaphysis) regions of femurs from young adult (5 mo) and early-old-age (22 mo) female C57BL/6JN mice. LCS bone mineralization was measured by the presence of perilacunar fluorochrome labels. LCS bone resorption was measured by immunohistochemical marker of bone resorption. The dynamics of peri-LCS turnover were estimated from serial fluorochrome labeling, where each mouse was administered two labels between 2 and 16 days before euthanasia. Osteocyte participation in mineralizing their surroundings is highly abundant in both cortical and cancellous bone of young adult mice but significantly decreases with aging. LCS bone resorption also decreases with aging. Aging has a greater impact on peri-LCS turnover dynamics in cancellous bone than in cortical bone. Lacunae with recent peri-LCS turnover are larger in both age groups. While peri-LCS turnover is associated with variation in tissue strain between cortical quadrants and intracortical location for 22 mo mice, these associations were not seen for 5 mo mice. The impact of aging on decreasing peri-LCS turnover may have significant implications for bone quality and mechanosensation.


Subject(s)
Aging , Mice, Inbred C57BL , Osteocytes , Animals , Female , Osteocytes/metabolism , Aging/physiology , Mice , Bone Resorption/pathology , Bone Resorption/metabolism , Femur/metabolism , Bone Remodeling/physiology
15.
Differentiation ; 138: 100789, 2024.
Article in English | MEDLINE | ID: mdl-38896972

ABSTRACT

Osteoclast (OC) differentiation, vital for bone resorption, depends on osteoclast and precursor fusion. Osteoprotegerin (OPG) inhibits osteoclast differentiation. OPG's influence on fusion and mechanisms is unclear. Osteoclasts and precursors were treated with OPG alone or with ATP. OPG significantly reduced OC number, area and motility and ATP mitigated OPG's inhibition. However, OPG hardly affected the motility of precusors. OPG downregulated fusion-related molecules (CD44, CD47, DC-STAMP, ATP6V0D2) in osteoclasts, reducing only CD47 in precursors. OPG reduced Connexin43 phosphorylated forms (P1 and P2) in osteoclasts, affecting only P2 in precursors. OPG disrupted subcellular localization of CD44, CD47, DC-STAMP, ATP6V0D2, and Connexin43 in both cell types. Findings underscore OPG's multifaceted impact, inhibiting multinucleated osteoclast and mononuclear precursor fusion through distinct molecular mechanisms. Notably, ATP mitigates OPG's inhibitory effect, suggesting a potential regulatory role for the ATP signaling pathway. This study enhances understanding of intricate processes in osteoclast differentiation and fusion, offering insights into potential therapeutic targets for abnormal bone metabolism.


Subject(s)
Adenosine Triphosphate , Cell Differentiation , Osteoclasts , Osteoprotegerin , Osteoprotegerin/metabolism , Osteoprotegerin/genetics , Osteoclasts/metabolism , Osteoclasts/cytology , Animals , Adenosine Triphosphate/metabolism , Mice , Connexin 43/metabolism , Connexin 43/genetics , Cell Fusion , CD47 Antigen/metabolism , CD47 Antigen/genetics , Hyaluronan Receptors/metabolism , Hyaluronan Receptors/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , Bone Resorption/metabolism , Bone Resorption/genetics , Bone Resorption/pathology , Signal Transduction , Vacuolar Proton-Translocating ATPases/metabolism , Vacuolar Proton-Translocating ATPases/genetics , Nerve Tissue Proteins
16.
J Biol Chem ; 300(7): 107487, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38908751

ABSTRACT

Macrophages are essential regulators of inflammation and bone loss. Receptor activator of nuclear factor-κß ligand (RANKL), a pro-inflammatory cytokine, is responsible for macrophage differentiation to osteoclasts and bone loss. We recently showed that 14-3-3ζ-knockout (YwhazKO) rats exhibit increased bone loss in the inflammatory arthritis model. 14-3-3ζ is a cytosolic adaptor protein that actively participates in many signaling transductions. However, the role of 14-3-3ζ in RANKL signaling or bone remodeling is unknown. We investigated how 14-3-3ζ affects osteoclast activity by evaluating its role in RANKL signaling. We utilized 14-3-3ζ-deficient primary bone marrow-derived macrophages obtained from wildtype and YwhazKO animals and RAW264.7 cells generated using CRISPR-Cas9. Our results showed that 14-3-3ζ-deficient macrophages, upon RANKL stimulation, have bigger and stronger tartrate-resistant acid phosphatase-positive multinucleated cells and increased bone resorption activity. The presence of 14-3-3ζ suppressed RANKL-induced MAPK and AKT phosphorylation, transcription factors (NFATC1 and p65) nuclear translocation, and subsequently, gene induction (Rank, Acp5, and Ctsk). Mechanistically, 14-3-3ζ interacts with TRAF6, an essential component of the RANKL receptor complex. Upon RANKL stimulation, 14-3-3ζ-TRAF6 interaction was increased, while RANK-TRAF6 interaction was decreased. Importantly, 14-3-3ζ supported TRAF6 ubiquitination and degradation by the proteasomal pathway, thus dampening the downstream RANKL signaling. Together, we show that 14-3-3ζ regulates TRAF6 levels to suppress inflammatory RANKL signaling and osteoclast activity. To the best of our knowledge, this is the first report on 14-3-3ζ regulation of RANKL signaling and osteoclast activation.


Subject(s)
14-3-3 Proteins , Osteoclasts , RANK Ligand , Signal Transduction , TNF Receptor-Associated Factor 6 , Animals , Mice , 14-3-3 Proteins/metabolism , 14-3-3 Proteins/genetics , Bone Resorption/metabolism , Bone Resorption/genetics , Bone Resorption/pathology , Macrophages/metabolism , Mice, Knockout , Osteoclasts/metabolism , Osteoclasts/cytology , Protein Stability , RANK Ligand/metabolism , RANK Ligand/genetics , RAW 264.7 Cells , TNF Receptor-Associated Factor 6/metabolism , TNF Receptor-Associated Factor 6/genetics , Ubiquitination
17.
Commun Biol ; 7(1): 548, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38719881

ABSTRACT

Hyperthyroidism is a well-known trigger of high bone turnover that can lead to the development of secondary osteoporosis. Previously, we have shown that blocking bone morphogenetic protein (BMP) signaling systemically with BMPR1A-Fc can prevent bone loss in hyperthyroid mice. To distinguish between bone cell type-specific effects, conditional knockout mice lacking Bmpr1a in either osteoclast precursors (LysM-Cre) or osteoprogenitors (Osx-Cre) were rendered hyperthyroid and their bone microarchitecture, strength and turnover were analyzed. While hyperthyroidism in osteoclast precursor-specific Bmpr1a knockout mice accelerated bone resorption leading to bone loss just as in wildtype mice, osteoprogenitor-specific Bmpr1a deletion prevented an increase of bone resorption and thus osteoporosis with hyperthyroidism. In vitro, wildtype but not Bmpr1a-deficient osteoblasts responded to thyroid hormone (TH) treatment with increased differentiation and activity. Furthermore, we found an elevated Rankl/Opg ratio with TH excess in osteoblasts and bone tissue from wildtype mice, but not in Bmpr1a knockouts. In line, expression of osteoclast marker genes increased when osteoclasts were treated with supernatants from TH-stimulated wildtype osteoblasts, in contrast to Bmpr1a-deficient cells. In conclusion, we identified the osteoblastic BMP receptor BMPR1A as a main driver of osteoporosis in hyperthyroid mice promoting TH-induced osteoblast activity and potentially its coupling to high osteoclastic resorption.


Subject(s)
Bone Morphogenetic Protein Receptors, Type I , Bone Resorption , Hyperthyroidism , Osteoblasts , Animals , Male , Mice , Bone Morphogenetic Protein Receptors, Type I/genetics , Bone Morphogenetic Protein Receptors, Type I/metabolism , Bone Resorption/metabolism , Bone Resorption/genetics , Cell Differentiation , Hyperthyroidism/metabolism , Hyperthyroidism/genetics , Hyperthyroidism/complications , Mice, Knockout , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteoporosis/metabolism , Osteoporosis/genetics , Osteoporosis/etiology , Osteoporosis/pathology
18.
Biomed Pharmacother ; 175: 116732, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38739990

ABSTRACT

Osteoporosis is a systemic bone disease characterized by decreased bone mass that is tightly regulated by the coordinated actions of osteoclasts and osteoblasts. Apoptosis as a precise programmed cell death involves a cascade of gene expression events which are mechanistically linked to the regulation of bone metabolism. Nevertheless, the critical biomolecules involved in regulating cell apoptosis in osteoporosis remain unknown. To gain a deeper insight into the relationship between apoptosis and osteoporosis, this study integrated the sequencing results of human samples and using a machine learning workflow to overcome the limitations of a single study. Among all immune cell populations, we assessed the apoptotic level and portrayed the distinct subtypes and lineage differentiation of monocytic cells in osteoporotic tissues. Osteoclasts expressed a higher level of Spermidine/spermine-N1-Acetyltransferase1 (SAT1) during osteoclastogenesis which prevented osteoclasts apoptosis and facilitate osteoporosis progression. In addition, Berenil, one potent SAT1 inhibitor, increased osteoclast apoptosis and reversed the bone loss in the femurs of a murine ovariectomy model. In summary, Berenil promotes osteoclast apoptosis, inhibits the bone resorption and improves the abnormal bone structure in vitro and in vivo models by targeting SAT1, demonstrating its potential as a precise therapeutic strategy for clinical osteoporosis treatment.


Subject(s)
Acetyltransferases , Apoptosis , Osteoclasts , Osteoporosis , Apoptosis/drug effects , Animals , Osteoclasts/metabolism , Osteoclasts/pathology , Osteoclasts/drug effects , Osteoporosis/pathology , Osteoporosis/prevention & control , Osteoporosis/metabolism , Humans , Female , Mice , Acetyltransferases/metabolism , Acetyltransferases/genetics , Mice, Inbred C57BL , Bone Resorption/metabolism , Bone Resorption/pathology , Bone Resorption/prevention & control , Ovariectomy , Osteogenesis/drug effects , Cell Differentiation , Disease Models, Animal
19.
JCI Insight ; 9(10)2024 May 22.
Article in English | MEDLINE | ID: mdl-38713511

ABSTRACT

While sclerostin-neutralizing antibodies (Scl-Abs) transiently stimulate bone formation by activating Wnt signaling in osteoblast lineage cells, they exert sustained inhibition of bone resorption, suggesting an alternate signaling pathway by which Scl-Abs control osteoclast activity. Since sclerostin can activate platelet-derived growth factor receptors (PDGFRs) in osteoblast lineage cells in vitro and PDGFR signaling in these cells induces bone resorption through M-CSF secretion, we hypothesized that the prolonged anticatabolic effect of Scl-Abs could result from PDGFR inhibition. We show here that inhibition of PDGFR signaling in osteoblast lineage cells is sufficient and necessary to mediate prolonged Scl-Ab effects on M-CSF secretion and osteoclast activity in mice. Indeed, sclerostin coactivates PDGFRs independently of Wnt/ß-catenin signaling inhibition, by forming a ternary complex with LRP6 and PDGFRs in preosteoblasts. In turn, Scl-Ab prevents sclerostin-mediated coactivation of PDGFR signaling and consequent M-CSF upregulation in preosteoblast cultures, thereby inhibiting osteoclast activity in preosteoblast/osteoclast coculture assays. These results provide a potential mechanism explaining the dissociation between anabolic and antiresorptive effects of long-term Scl-Ab.


Subject(s)
Adaptor Proteins, Signal Transducing , Bone Resorption , Osteoblasts , Osteoclasts , Receptors, Platelet-Derived Growth Factor , Signal Transduction , Animals , Osteoblasts/metabolism , Mice , Adaptor Proteins, Signal Transducing/metabolism , Bone Resorption/metabolism , Osteoclasts/metabolism , Receptors, Platelet-Derived Growth Factor/metabolism , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Wnt Signaling Pathway/drug effects , Antibodies, Neutralizing/pharmacology , Low Density Lipoprotein Receptor-Related Protein-6/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Cell Lineage , Osteogenesis/drug effects , Cell Differentiation
20.
Sci Bull (Beijing) ; 69(12): 1964-1979, 2024 Jun 30.
Article in English | MEDLINE | ID: mdl-38760248

ABSTRACT

Mechanical loading is required for bone homeostasis, but the underlying mechanism is still unclear. Our previous studies revealed that the mechanical protein polycystin-1 (PC1, encoded by Pkd1) is critical for bone formation. However, the role of PC1 in bone resorption is unknown. Here, we found that PC1 directly regulates osteoclastogenesis and bone resorption. The conditional deletion of Pkd1 in the osteoclast lineage resulted in a reduced number of osteoclasts, decreased bone resorption, and increased bone mass. A cohort study of 32,500 patients further revealed that autosomal dominant polycystic kidney disease, which is mainly caused by loss-of-function mutation of the PKD1 gene, is associated with a lower risk of hip fracture than those with other chronic kidney diseases. Moreover, mice with osteoclast-specific knockout of Pkd1 showed complete resistance to unloading-induced bone loss. A mechanistic study revealed that PC1 facilitated TAZ nuclear translocation via the C-terminal tail-TAZ complex and that conditional deletion of Taz in the osteoclast lineage resulted in reduced osteoclastogenesis and increased bone mass. Pharmacological regulation of the PC1-TAZ axis alleviated unloading- and estrogen deficiency- induced bone loss. Thus, the PC1-TAZ axis may be a potential therapeutic target for osteoclast-related osteoporosis.


Subject(s)
Bone Resorption , Mice, Knockout , Osteoclasts , Osteogenesis , TRPP Cation Channels , Animals , TRPP Cation Channels/genetics , TRPP Cation Channels/metabolism , Bone Resorption/metabolism , Bone Resorption/genetics , Bone Resorption/pathology , Osteoclasts/metabolism , Mice , Humans , Osteoporosis/genetics , Osteoporosis/metabolism , Osteoporosis/pathology , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/metabolism , Polycystic Kidney, Autosomal Dominant/pathology , Male , Female , Adaptor Proteins, Signal Transducing
SELECTION OF CITATIONS
SEARCH DETAIL