Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.212
Filtrar
Más filtros

Intervalo de año de publicación
1.
Bioact Mater ; 43: 441-459, 2025 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-39399835

RESUMEN

The osteoporotic bone defect caused by excessive activity of osteoclasts has posed a challenge for public healthcare. However, most existing bioinert bone cement fails to effectively regulate the pathological bone microenvironment and reconstruct bone homeostasis in the presence of osteoclast overactivity and osteoblast suppression. Herein, inspired by natural bone tissue, an in-situ modulation system for osteoporotic bone regeneration is developed by fabricating an injectable double-crosslinked PEGylated poly(glycerol sebacate) (PEGS)/calcium phosphate cement (CPC) loaded with sodium alendronate (ALN) (PEGS/CPC@ALN) adhesive bone cement. By incorporating ALN, the organic-inorganic interconnection within PEGS/CPC@ALN results in a 100 % increase in compression modulus and energy dissipation efficiency. Additionally, PEGS/CPC@ALN effectively adheres to the bone by bonding with amine and calcium ions present on the bone surface. Moreover, this in-situ regulation system comprehensively mitigates excessive bone resorption through the buffering effect of CPC to improve the acidic microenvironment of osteoporotic bone and the release of ALN to inhibit hyperactive osteoclasts, and facilitates stem cell proliferation and differentiation into osteoblasts through calcium ion release. Overall, the PEGS/CPC@ALN effectively regulates the pathological microenvironment of osteoporosis while promoting bone regeneration through synergistic effects of drugs and materials, thereby improving bone homeostasis and enabling minimally invasive treatment for osteoporotic defects.

2.
J Bone Miner Metab ; 2024 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-39373772

RESUMEN

INTRODUCTION: Disulfiram (DSF), known as an anti-alcoholism drug, has been reported to suppress osteoclast differentiation in vitro; however, it remains uncertain whether DSF is effective in preventing osteoclastogenesis in vivo. This study aimed to investigate the effect of DSF administration in osteoporotic mice and its contribution to osteoclastogenesis in vivo. MATERIALS AND METHODS: The bone phenotype of ovariectomized mice, both treated and untreated with DSF, was examined using microcomputed tomography analysis. Osteoclastic and osteoblastic parameters were assessed through bone morphometric analysis. The direct effect of DSF on osteoblastogenesis in vitro was evaluated via a primary osteoblast culture experiment. The expression of genes related to DSF targets (Nup85, Ccr2, and Ccr5) in osteoclast-lineage cells was examined using scRNA-seq analysis and flow cytometry analysis using the bone marrow cells from ovariectomized mice. The impact of DSF on osteoclast-lineage cells was assessed using primary cultures of osteoclasts. RESULTS: DSF administration ameliorated ovariectomy-induced bone loss and mitigated the increase of osteoclasts without affecting osteoblastogenesis. The scRNA-seq data revealed that osteoclast precursor cells expressed Nup85, Ccr2, and Ccr5. CCR2 and CCR5-positive cells in osteoclast precursor cells within bone marrow increased following ovariectomy, and this increase was canceled by DSF administration. Finally, we found that DSF had a significant inhibitory effect on osteoclastogenesis in the early stage by suppressing Tnfrsf11a expression. CONCLUSION: This study demonstrates that DSF could be a candidate for osteoporosis therapies because it suppresses osteoclastogenesis from an early stage in vivo.

3.
Int Immunopharmacol ; 143(Pt 1): 113321, 2024 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-39388890

RESUMEN

As the most predominant form of arthritis, osteoarthritis (OA) is featured with irreversible progress and involvement of the whole joint. Since OA onset, abnormal mechanical load initiates excessive osteoclastogenesis, evolving a rapid turnover of subchondral bone, cyst creation, synovitis, cartilage degradation, and ultimately resulting in joint failure. Additionally, aberrant vascularization and nociceptive pain are invoked by osteoclast-induced angiogenesis and sensory innervation in the subchondral bone. Rhizoma anemarrhenae (Zhimu) has been extensively demonstrated to show multiple pharmacological effects including anti-inflammation, anti-aging, and immunomodulation. Herein, Broussonin a (BRA), Markogein (MAN), and Isosakuranetin (ISN) derived from Rhizoma anemarrhenae, were initially discovered for their affinity with Bone marrow mononuclear cell (BMMC) membranes using the Cell membrane chromatography/Time of flight mass spectrometry (CMC/TOFMS) method, while only ISN exerted a significant inhibitory effect on RANKL-induced osteoclastogenesis in BMMC in vitro. Intriguingly, we disclosed that ISN blunted the overactivation of Tartrate-resistant acid phosphatase positive (TRAP+) osteoclasts in subchondral bone in OA mice, as indicated by enhanced bone volume/total volume (BV/TV), trabecular number (Tb.N), and trabeculae thickness (Tb.Th), as well as diminished trabecular pattern factor (Tb.pf). Treatment with ISN also impaired aberrant angiogenesis and nociceptive reaction in the subchondral bone marrow. Moreover, ISN hindered the loss of articular cartilage proteoglycan and lowered the Osteoarthritis Research Society International (OARSI) grade, boosting the expression amount of Aggrecan (ACAN) and Collagen II (COL II) positive cells while reducing Matrix metalloproteinase 13 (MMP-13) positive cells. For mechanisms, We verified that ISN hampered subchondral osteoclastogenesis by blocking nuclear factor kappa light chain enhancer of activated B cells (NF-κB) signaling and C-X-C Motif Chemokine Ligand 2 (CXCL2) stimulation. Taken together, we reveal that ISN impedes the progression of OA by preventing hyperactivated subchondral osteoclastogenesis via suppressing the NF-κB/CXCL2 axis.

4.
PeerJ ; 12: e18222, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39399421

RESUMEN

Background: Osteoporosis (OP) is a chronic disease characterized by decreased bone mass, loss of skeletal structural integrity and increased susceptibility to fracture. Available studies have shown that the pyruvate dehydrogenase kinase (PDK) family is associated with osteoclastogenesis and bone loss, but the specific role of Pdk3 in bone pathology has not been systematically investigated. Methods: A cell OP model was established in receptor activator for nuclear factor-κB Ligand (RANKL)-induced bone marrow macrophages (BMMs). Hereafter, the expression levels of Pdk3 and osteoclastogenesis feature genes including nuclear factor of activated T cells 1 (Nfatc1), Cathepsin K (Ctsk), osteoclast associated Ig-like receptor (Oscar) in BMMs-derived osteoclasts were examined based on real-time quantitative PCR and western blotting methods. Further, the phosphorylation of ERK, P65 and JAK/STAT and their correlation was Pdk3 was gauged. In particular, changes in the activity of these signaling pathways were observed by silencing experiments of the Pdk3 gene (using small interfering RNA). Finally, the effects of Pdk3 gene silencing on signaling pathway activity, osteoclastogenesis, and related inflammatory and apoptotic indicators were observed by transfection with PDK3-specific siRNA. Results: Following RANKL exposure, the levels of Pdk3 and osteoclastogenesis feature genes were all elevated, and a positive correlation between Pdk3 and osteoclastogenesis feature genes was seen. Meanwhile, ERK, P65 and JAK/STAT phosphorylation was increased by RANKL, and Pdk3 was confirmed to be positively correlated with the phosphorylation of ERK, P65 and JAK/STAT. Additionally, in RANKL-exposed osteoclasts, Pdk3 knockdown diminished the phosphorylation of ERK, P65 and JAK/STAT, reduced the expressions of osteoclastogenesis feature genes. Importantly, knockdown of Pdk3 also reduced the expression of inflammatory cytokines and resulted in elevated levels of Bax and Casp3 expression, as well as downregulation of Bcl2 expression. Conclusion: This study reveals for the first time the role of Pdk3 in RANKL-induced osteoclastogenesis and OP. These findings provide a foundation for future studies on the role of Pdk3 in other bone diseases and provide new ideas for the development of OP therapeutics targeting Pdk3.


Asunto(s)
Diferenciación Celular , Macrófagos , Osteoclastos , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Ligando RANK , Animales , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Ligando RANK/metabolismo , Ligando RANK/farmacología , Diferenciación Celular/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Ratones , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/metabolismo , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/genética , Transducción de Señal/efectos de los fármacos , Osteoporosis/patología , Osteoporosis/genética , Osteoporosis/metabolismo , Osteogénesis/efectos de los fármacos , Apoptosis/efectos de los fármacos , Factores de Transcripción NFATC/metabolismo , Factores de Transcripción NFATC/genética , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/efectos de los fármacos , Catepsina K
5.
Mater Today Bio ; 28: 101225, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39309162

RESUMEN

Wear particles produced by joint replacements induce inflammatory responses that lead to periprosthetic osteolysis and aseptic loosening. However, the precise mechanisms driving wear particle-induced osteolysis are not fully understood. Recent evidence suggests that autophagy, a cellular degradation process, plays a significant role in this pathology. This study aimed to clarify the role of autophagy in mediating inflammation and osteolysis triggered by wear particles and to evaluate the therapeutic potential of zinc oxide nanoparticles (ZnO NPs). We incorporated ZnO into the prosthetic material itself, ensuring that the wear particles inherently carried ZnO, providing a targeted and sustained intervention. Our findings reveal that polymer wear particles induce excessive autophagic activity, which is closely associated with increased inflammation and osteolysis. We identified secretory autophagy as a key mechanism for IL-1ß secretion, exacerbating osteolysis. Both in vitro and in vivo experiments demonstrated that ZnO-doped particles significantly inhibit autophagic overactivation, thereby reducing inflammation and osteolysis. In summary, this study establishes secretory autophagy as a critical mechanism in wear particle-induced osteolysis and highlights the potential of ZnO-doped prosthetic polymers for targeted, sustained mitigation of periprosthetic osteolysis.

6.
Lasers Med Sci ; 39(1): 247, 2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39349883

RESUMEN

Appropriate regeneration of jawbone after dental or surgical procedures relies on the recruitment of osteoprogenitor cells able to differentiate into matrix-producing osteoblasts. In this context, photobiomodulation (PBM) has emerged as promising therapy to improve tissue regeneration and to facilitate wound healing processes. The aim of this study was to determine the effect of PBM on human osteoprogenitor cells isolated from mandibular trabecular bone.Bone marrow stromal cell cultures were established from 4 donors and induced toward osteogenic differentiation for 14 days in a standard osteogenic assay. Cells were irradiated with a combined red/near-infrared (NIR) laser following different schedules and expression of osteogenic, matrix-related, osteoclastogenic and inflammatory genes was analyzed by quantitative PCR.Gene expression analysis revealed no overall effects of PBM on osteogenic differentiation. However, a statistically significant reduction was observed in the transcripts of COL1A1 and MMP13, two important genes involved in the bone matrix homeostasis. Most important, PBM significantly downregulated the expression of RANKL, IL6 and IL1B, three genes that are involved in both osteoclastogenesis and inflammation.In conclusion, PBM with a red/NIR laser did not modulate the osteogenic phenotype of mandibular osteoprogenitors but markedly reduced their expression of matrix-related genes and their pro-osteoclastogenic and pro-inflammatory profile.


Asunto(s)
Diferenciación Celular , Terapia por Luz de Baja Intensidad , Mandíbula , Osteogénesis , Humanos , Terapia por Luz de Baja Intensidad/métodos , Osteogénesis/efectos de la radiación , Mandíbula/efectos de la radiación , Diferenciación Celular/efectos de la radiación , Interleucina-1beta/metabolismo , Interleucina-1beta/genética , Ligando RANK/metabolismo , Ligando RANK/genética , Células Madre Mesenquimatosas/efectos de la radiación , Células Madre Mesenquimatosas/metabolismo , Metaloproteinasa 13 de la Matriz/metabolismo , Metaloproteinasa 13 de la Matriz/genética , Interleucina-6/metabolismo , Interleucina-6/genética , Osteoclastos/efectos de la radiación , Células Cultivadas , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I , Expresión Génica/efectos de la radiación , Inflamación/radioterapia , Rayos Infrarrojos/uso terapéutico
7.
J Pharm Bioallied Sci ; 16(Suppl 3): S2488-S2490, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39346162

RESUMEN

Background: Osteoclastogenesis, the formation of osteoclasts from precursor cells, plays a pivotal role in bone remodeling and associated pathologies like osteoporosis and rheumatoid arthritis. Host modulatory agents (HMAs) have emerged as potential therapeutic candidates for modulating osteoclastogenesis. However, their effects need comprehensive evaluation through in vitro studies. Materials and Methods: In this study, we conducted an in vitro assessment of the effects of a novel HMA on osteoclastogenesis. Primary murine bone marrow-derived macrophages were cultured with the receptor activator of nuclear factor kappa-B ligand to induce osteoclast differentiation. The HMA was administered at various concentrations, and osteoclastogenesis was evaluated through tartrate-resistant acid phosphatase (TRAP) staining, osteoclast size measurement, and gene expression analysis of osteoclast markers. Results: Treatment with the HMA resulted in a dose-dependent inhibition of osteoclast formation. At the highest concentration (100 µM), osteoclastogenesis was significantly suppressed, with a reduction in the number of TRAP-positive multinucleated cells from 50 ± 5 to 10 ± 2 per field (P < 0.001). Moreover, the osteoclast size was markedly reduced, with an average diameter of 20 ± 3 µm compared to 35 ± 4 µm in the control group (P < 0.05). Gene expression analysis revealed downregulation of osteoclast-specific markers, including TRAP, Cathepsin K, and NFATc1, confirming the inhibitory effect of the HMA on osteoclastogenesis. Conclusion: Our findings demonstrate the potential of the investigated HMA as a modulator of osteoclastogenesis. By suppressing osteoclast formation and activity, this agent holds promise for the development of novel therapeutic strategies targeting bone resorption-associated disorders.

8.
Orthod Craniofac Res ; 2024 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-39319631

RESUMEN

OBJECTIVES: The objective of this study was to determine changes in gene expression by establishing an orthodontic tooth movement (OTM) rat model with appropriate and excessive orthodontic force. MATERIALS AND METHODS: Using a closed coil nickel-titanium spring, the OTM was carried out to apply a mesial force of 50 or 100 g to the maxillary first molars. Micro-CT, histological and immunohistochemical staining were used to evaluate the bone formation at the tension site and the bone resorption and bone formation at pressure site. Then RNA sequencing and bioinformatic analysis were performed. RESULTS: According to the results of the Mirco-CT scan of OTM rat models, both the 50 g group and the 100 g group showed variable degrees of reduction in alveolar bone density on the tension and pressure sides. The results of histological and immunohistochemical staining demonstrated that the periodontal tissue and osteogenic ability of the 50 g group were restored at the 14 days, while the 100 g group caused severe periodontal tissue damage. The GO and KEGG analysis results, as well as the number of differentially expressed genes (DEGs), varied depending on the loading time and value of appliance, according to the results of the RNA sequencing. And the immunohistochemical staining results showed that Sfrp4 functioned by efficiently influencing both bone formation and bone absorption. CONCLUSIONS: Appropriate orthodontic force value could cause appropriate movement of teeth in rats without adverse periodontal damage. Simultaneously, distinct gene expression patterns were observed at various force levels and time intervals.

9.
Front Immunol ; 15: 1425670, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39281679

RESUMEN

Rheumatoid arthritis (RA) is a chronic autoimmune disease accompanied by local and systemic bone loss. FcγRs, especially FcγRIIa (hFcγRIIa), have been implicated in the pathogenesis of RA. However, the contribution of hFcγRIIa to bone loss has not been fully elucidated. In the present study, we demonstrated the double-edged sword role of hFcγRIIa on osteoclast differentiation through investigations involving hFcγRIIa-transgenic (hFcγRIIa-Tg) mice. Our findings reveal that hFcγRIIa-Tg mice, previously shown to exhibit heightened susceptibility to collagen-induced arthritis (CIA), displayed increased osteoporosis during CIA or at advanced ages (40 weeks), accompanied by heightened in vivo osteoclast differentiation. Notably, bone marrow cells from hFcγRIIa-Tg mice exhibited enhanced efficiency in differentiating into osteoclasts and bone resorption in vitro compared to wild-type mice when stimulated with receptor activators of NF-κB ligand (RANKL). Additionally, hFcγRIIa-Tg mice exhibited augmented sensitivity to RANKL-induced bone loss in vivo, highlighting the osteoclast-promoting role of hFcγRIIa. Mechanistically, bone marrow cells from hFcγRIIa-Tg mice displayed heightened Syk self-activation, leading to mTOR-pS6 pathway activation, thereby promoting RANKL-driven osteoclast differentiation. Intriguingly, while hFcγRIIa crosslinking hindered RANKL-induced osteoclast differentiation, it activated the kinase cAbl, subsequently triggering STAT5 activation and inhibiting the expression of osteoclast-associated genes. This study provides novel insights into hFcγRIIa-mediated osteoclast biology, suggesting promising therapeutic targets for managing bone remodeling disorders.


Asunto(s)
Resorción Ósea , Diferenciación Celular , Osteoclastos , Osteogénesis , Receptores de IgG , Animales , Ratones , Artritis Experimental/inmunología , Artritis Experimental/genética , Artritis Reumatoide/metabolismo , Artritis Reumatoide/inmunología , Artritis Reumatoide/genética , Resorción Ósea/genética , Resorción Ósea/metabolismo , Ratones Transgénicos , Osteoclastos/metabolismo , Osteoporosis/genética , Osteoporosis/etiología , Osteoporosis/metabolismo , Ligando RANK/metabolismo , Ligando RANK/genética , Receptores de IgG/genética , Receptores de IgG/metabolismo , Transducción de Señal
10.
Biochim Biophys Acta Mol Cell Res ; 1871(8): 119850, 2024 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-39303785

RESUMEN

Osteoclasts play a crucial role in bone destruction in rheumatoid arthritis (RA). This study aimed to investigate the inhibitory effects of chaperonin 60 (CPN60), identified in the surface proteins of Propionibacterium freudenreichii MJ2, on receptor activator of nuclear factor kappa-B ligand (RANKL)-induced osteoclast differentiation, and elucidate the underlying mechanisms. Treatment with CPN60 inhibited RANKL-induced osteoclast differentiation by decreasing the expression of osteoclast differentiation-related genes and proteins. CPN60 interfered with the binding of RANKL to RANK, as elucidated using surface plasmon resonance (SPR) and immunofluorescence. In silico molecular docking analysis further supported the interference of CPN60 with the binding of RANKL and RANK. CPN60 suppressed the expression of molecules linked to the calcium-dependent pathway in RANKL-induced osteoclast differentiation at both mRNA and protein levels. Microarray analysis showed elevated expression of lipocalin 2 (Lcn2), which was closely linked to the inhibition of osteoclast differentiation in CPN60-treated RAW 264.7 cells. Inhibition of Lcn2 decreased the inhibitory effect of CPN60 on osteoclast differentiation, indicating that increased expression of Lcn2 by CPN60 contributes to the inhibition of osteoclastogenesis. In addition, CPN60 treatment alleviated arthritis symptoms in collagen-induced arthritis mice by reducing the generation of collagen-specific antibodies and inhibiting osteoclast differentiation. In conclusion, CPN60 of P. freudenreichii MJ2 interfered with RANKL-RANK binding, reduced the expression of genes and proteins related to osteoclast differentiation and upregulated Lcn2 expression, thereby inhibiting RANKL-induced osteoclast differentiation, which might contribute to ameliorate collagen-induced arthritis.

11.
Genes (Basel) ; 15(9)2024 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-39336725

RESUMEN

ZC4H2 encodes a C4H2-type zinc finger protein, mutations of which lead to a spectrum of diseases known as ZC4H2 associated rare disorders (ZARD). In addition to neurological phenotypes, the most typical symptoms of ZARD are multiple joint contractures of varying degrees, accompanied by abnormal development of muscles and bones, and osteoporosis in some cases. The pathogenic mechanisms of such bone related phenotypes, however, remain unclear. Here, we showed that ZC4H2 is expressed in the developing bones in mice. ZC4H2 knockout mice were neonatal-lethal and smaller in size, with reduced calcification of long bones. Upon induced loss of ZC4H2 postnatally, the femoral bones developed an osteoporosis-like phenotype, with reduced bone mineral density, bone-volume fraction, and trabecular bone number. Knockdown of ZC4H2 showed no clear effect on the expression of osteogenic differentiation genes in in vitro models using mesenchymal stem cells. Interestingly, ZC4H2 knockdown significantly enhanced osteoclast differentiation and bone resorption in induced bone marrow-derived macrophages. We further confirmed that the number of osteoclasts in the long bone of ZC4H2 knockout mice was increased, as well as the expression of the serum bone resorption/osteoporosis marker CTX-1. Our study unveils a new role of ZC4H2 in osteoclast differentiation and bone development, providing new clues on the pathology of ZARD.


Asunto(s)
Artrogriposis , Ratones Noqueados , Osteoclastos , Osteogénesis , Animales , Ratones , Osteoclastos/metabolismo , Osteoclastos/patología , Osteogénesis/genética , Artrogriposis/genética , Artrogriposis/patología , Diferenciación Celular/genética , Osteoporosis/genética , Osteoporosis/patología , Resorción Ósea/genética , Resorción Ósea/patología , Células Madre Mesenquimatosas/metabolismo , Densidad Ósea/genética
12.
Genes (Basel) ; 15(9)2024 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-39336808

RESUMEN

BACKGROUND/OBJECTIVES: Glass, bone, and dentin are commonly applied substrates for osteoclast cultures; however, the impact of these substrates on osteoclastogenesis remains underexplored. This study aimed to address a significant gap in understanding how different substrates influence the process of osteoclastogenesis. METHODS: RAW 264.7 cells were cultured and induced with RANKL on glass, bone, and dentin slides. Histological and molecular techniques were used to identify patterns and differences in osteoclast behavior on each substrate. RESULTS: Osteoclasts cultured on glass slides possessed the greatest number of nuclei and the highest expression levels of ACP5 (TRAP) and CTSK, with osteoclasts on bone and dentin slides displaying progressively lower levels. Src expression was also most pronounced in osteoclasts on glass slides, with decreased levels observed on bone and dentin. This variation in Src expression likely contributed to differences in cytoskeletal remodeling and oxidative phosphorylation (OXPHOS), resulting in substrate-dependent divergences in osteoclastogenesis. CONCLUSIONS: Glass slides were the most favorable substrate for inducing osteoclastogenesis, while bone and dentin slides were less effective. The substrate-induced expression of Src played a fundamental role in shaping the phenotypic divergence of osteoclasts. These insights fill important knowledge gaps and have significant implications for the development and selection of in vitro models for bone-related diseases and drug screening platforms.


Asunto(s)
Osteoclastos , Osteogénesis , Animales , Ratones , Osteoclastos/metabolismo , Osteoclastos/citología , Células RAW 264.7 , Osteogénesis/genética , Ligando RANK/metabolismo , Ligando RANK/genética , Familia-src Quinasas/metabolismo , Familia-src Quinasas/genética , Diferenciación Celular , Dentina/metabolismo , Vidrio , Huesos/metabolismo , Huesos/citología
13.
Int Immunopharmacol ; 142(Pt B): 113245, 2024 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-39340985

RESUMEN

Periprosthetic osteolysis (PPO), caused by wear particles, is a significant complication of total joint replacement, leading to prosthesis failure. Previous research has highlighted the crucial role of osteoclast-induced bone destruction in PPO progression. Albiflorin (AF), a monoterpene glycoside from Paeonia lactiflora, is a key active ingredient known for its antioxidant and anti-inflammatory properties. Although AF has shown promise in treating various conditions, its impact on osteoclasts and PPO remains unexplored. Our study revealed that AF could effectively inhibit osteoclast differentiation to reduce overactivated bone resorption and effectively inhibit the accumulation of reactive oxygen species (ROS) induced by wear particles. In vitro experiments also confirmed that AF could effectively inhibit the PI3K/AKT signaling pathway and inhibit inflammation to regulate osteoclast generation. Studies in animal models have also verified the antioxidant and anti-inflammatory properties of AF. In summary, the above studies indicate that AF inhibits osteoclastogenesis via inhibiting ROS accumulation and the PI3K/AKT signaling pathway, which may be a potential therapeutic method for PPO.


Asunto(s)
Osteoclastos , Osteogénesis , Osteólisis , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Especies Reactivas de Oxígeno , Transducción de Señal , Titanio , Animales , Proteínas Proto-Oncogénicas c-akt/metabolismo , Osteólisis/tratamiento farmacológico , Osteólisis/inducido químicamente , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Osteoclastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Células RAW 264.7 , Ratones Endogámicos C57BL , Masculino , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Humanos , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Diferenciación Celular/efectos de los fármacos , Paeonia/química , Hidrocarburos Aromáticos con Puentes
14.
Bone Rep ; 22: 101798, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39252697

RESUMEN

In periodontitis, gingival fibroblasts (GF) appear to produce a multitude of paracrine factors. However, the influence of GF-derived soluble factors on osteoclastogenesis remains unclear. In this case study, production of paracrine factors by GF was assessed under inflammatory and non-inflammatory conditions, as well as their effect on osteoclastogenesis. Human primary GF were cultured in a transwell system and primed with a cocktail of IL-1ß, IL-6 and TNF-α to mimic inflammation. GF were co-cultured directly and indirectly with human peripheral blood mononuclear cells (PBMC). Cytokines and chemokines in supernatants (flow cytometry based multiplex assay), osteoclastogenesis (TRAcP staining) and gene expression (qPCR) were quantified on days 7 and 21. Results from this case study showed that GF communicated via soluble factors with PBMC resulting in a two-fold induction of osteoclasts. Reversely, PBMC induced gene expression of IL-6, OPG and MCP-1 by GF. Remarkably, after priming of GF with cytokines, this communication was impaired and resulted in fewer osteoclasts. This could be partly explained by an increase in IL-10 expression and a decrease in MCP-1 expression. Intriguingly, the short priming of GF resulted in significantly higher expression of inflammatory cytokines that was sustained at both 7 and 21 days. GF appear to produce paracrine factors capable of stimulating osteoclastogenesis in the absence of physical cell-cell interactions. GF cultured in the presence of PBMC or osteoclasts had a remarkably inflammatory phenotype. Given profound expression of both pro- and anti-inflammatory cytokines after the inflammatory stimulus, it is probably the effector hierarchy that leads to fewer osteoclasts.

15.
Eur J Pharmacol ; 982: 176956, 2024 Nov 05.
Artículo en Inglés | MEDLINE | ID: mdl-39209096

RESUMEN

Excessive activity of osteoclasts(OCs) lead to bone resorption in chronic inflammatory conditions. The use of natural compounds to target OCs offers significant promise in the treatment or prevention of OC-associated diseases. Irilin D (IRD), a natural isoflavone derived from Belamcanda chinensis (L.) DC., has potential effects on OC differentiation both in vitro and in vivo that have yet to be thoroughly explored. In our study, we found that IRD inhibited receptor activator of nuclear factor-κB ligand (RANKL)-induced OC differentiation, actin ring formation, and bone resorption in vitro without compromising cell viability. However, IRD did not exhibit anti-inflammatory effects in lipopolysaccharide (LPS)-stimulated macrophages. Furthermore, IRD reduced LPS-induced inflammatory bone loss by blocking osteoclastogenesis in a mouse model. Mechanistically, IRD disrupted RANKL-induced activation of mitogen-activated protein kinases (MAPKs) and nuclear factor-κB (NF-κB), leading to the inhibition of c-Fos and nuclear factor of activated T cells cytoplasmic 1 (NFATc1) activation. We also demonstrated that IRD inhibited RANKL-induced osteoclastic NFATc1 target genes, including DC-STAMP, ACP5, and CtsK. Our results indicate that IRD mitigates LPS-induced inflammatory bone resorption in mice by inhibiting RANKL-activated MAPKs and NF-κB signaling pathways, suggesting its potential as a natural isoflavone for preventing or treating OC-associated diseases.


Asunto(s)
Resorción Ósea , Inflamación , Isoflavonas , Sistema de Señalización de MAP Quinasas , FN-kappa B , Osteoclastos , Osteogénesis , Ligando RANK , Animales , Masculino , Ratones , Resorción Ósea/prevención & control , Resorción Ósea/inducido químicamente , Resorción Ósea/tratamiento farmacológico , Resorción Ósea/patología , Resorción Ósea/metabolismo , Diferenciación Celular/efectos de los fármacos , Inflamación/patología , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Isoflavonas/farmacología , Isoflavonas/uso terapéutico , Lipopolisacáridos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Factores de Transcripción NFATC/metabolismo , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Osteogénesis/efectos de los fármacos , Ligando RANK/metabolismo , Células RAW 264.7
16.
J Bone Miner Res ; 39(10): 1503-1517, 2024 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-39095084

RESUMEN

Bone-resorbing osteoclasts (OCLs) are formed by differentiation and fusion of monocyte precursor cells, generating large multinucleated cells. Tightly regulated cell fusion during osteoclastogenesis leads to formation of resorption-competent OCLs, whose sizes fall within a predictable physiological range. The molecular mechanisms that regulate the onset of OCL fusion and its subsequent arrest are, however, largely unknown. We have previously shown that OCLs cultured from mice homozygous for the R51Q mutation in the vesicle trafficking-associated protein sorting nexin 10, a mutation that induces autosomal recessive osteopetrosis in humans and in mice, display deregulated and continuous fusion that generates gigantic, inactive OCLs. Fusion of mature OCLs is therefore arrested by an active, genetically encoded, cell-autonomous, and SNX10-dependent mechanism. To directly examine whether SNX10 performs a similar role in vivo, we generated SNX10-deficient (SKO) mice and demonstrated that they display massive osteopetrosis and that their OCLs fuse uncontrollably in culture, as do homozygous R51Q SNX10 (RQ/RQ) mice. OCLs that lack SNX10 exhibit persistent presence of DC-STAMP protein at their periphery, which may contribute to their uncontrolled fusion. To visualize endogenous SNX10-mutant OCLs in their native bone environment, we genetically labeled the OCLs of WT, SKO, and RQ/RQ mice with enhanced Green Fluorescent Protein (EGFP), and then visualized the 3D organization of resident OCLs and the pericellular bone matrix by 2-photon, confocal, and second harmonics generation microscopy. We show that the volumes, surface areas and, in particular, the numbers of nuclei in the OCLs of both mutant strains were on average 2-6-fold larger than those of OCLs from WT mice, indicating that deregulated, excessive fusion occurs in the mutant mice. We conclude that the fusion of OCLs, and consequently their size, is regulated in vivo by SNX10-dependent arrest of fusion of mature OCLs.


Osteoclasts (OCLs) are cells that degrade bone. These cells are generated by fusion of monocyte precursor cells, but the mechanisms that regulate this process and eventually arrest it are unknown. We had previously shown that OCLs cultured from mice carrying the R51Q mutation in the protein sorting nexin 10 (SNX10) lose their resorptive capacity and become gigantic due to uncontrolled fusion. To examine whether SNX10 is required for OCL fusion arrest also in vivo, we inactivated the Snx10 gene in mice and fluorescently labeled their OCLs and OCLs of R51Q SNX10 mice, isolated their femurs, and used advanced 3D microscopy methods to visualize OCLs within the bone matrix. As expected, mice lacking SNX10 exhibited excessive bone mass, indicating that their OCLs are inactive. OCLs within bones of both mutant mouse strains were on average 2­6-fold larger than in control mice and contained proportionally more nuclei. We conclude that OCL fusion is arrested in control, but not SNX10 mutant, mice, indicating that the sizes of mature OCLs are limited in vivo by an active, SNX10-dependent mechanism that suppresses cell fusion.


Asunto(s)
Fusión Celular , Osteoclastos , Nexinas de Clasificación , Animales , Nexinas de Clasificación/metabolismo , Nexinas de Clasificación/genética , Osteoclastos/metabolismo , Osteoclastos/patología , Ratones , Osteopetrosis/patología , Osteopetrosis/genética , Osteopetrosis/metabolismo , Tamaño de la Célula
17.
Hum Exp Toxicol ; 43: 9603271241269028, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39197164

RESUMEN

BACKGROUND AND OBJECTIVE: The objective of this study was to investigate the potential of salidroside (SAL) (a major active compound in Rhodiola rosea L.) in regulating osteoclast differentiation and function by modulating the HIF-1α pathway and its downstream target genes. METHODS: The expression of HIF-1α and its downstream target genes was examined at both mRNA and protein levels in osteoclasts treated with SAL. Immunofluorescence analysis was performed to assess the nuclear translocation and transcriptional activity of HIF-1α in response to SAL. MTT, flow cytometry, qPCR, TRAP staining and bone resorption assays were used to evaluate the potential effect of salidroside on osteoclasts. RESULTS: SAL enhanced the expression of HIF-1α and its downstream target genes in osteoclasts. Immunofluorescence analysis confirmed the facilitation of HIF-1α nuclear translocation and transcriptional activity by SAL. In addition, SAL enhanced osteoclast viability, differentiation and bone resorption activity in an autocrine manner through HIF-1α/VEGF, IL-6 and ANGPTL4 pathways. CONCLUSION: SAL promotes osteoclast proliferation, differentiation and bone resorption through HIF-1α/VEGF, IL-6 and ANGPTL4 pathways.


Asunto(s)
Glucósidos , Subunidad alfa del Factor 1 Inducible por Hipoxia , Osteoclastos , Osteogénesis , Fenoles , Glucósidos/farmacología , Fenoles/farmacología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Osteoclastos/efectos de los fármacos , Animales , Ratones , Osteogénesis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Células RAW 264.7 , Interleucina-6/metabolismo , Interleucina-6/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Resorción Ósea , Transducción de Señal/efectos de los fármacos , Proteína 4 Similar a la Angiopoyetina/metabolismo , Proteína 4 Similar a la Angiopoyetina/genética , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos
18.
Mol Med ; 30(1): 125, 2024 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-39152382

RESUMEN

BACKGROUND: Epimedin A (EA) has been shown to suppress extensive osteoclastogenesis and bone resorption, but the effects of EA remain incompletely understood. The aim of our study was to investigate the effects of EA on osteoclastogenesis and bone resorption to explore the corresponding signalling pathways. METHODS: Rats were randomly assigned to the sham operation or ovariectomy group, and alendronate was used for the positive control group. The therapeutic effect of EA on osteoporosis was systematically analysed by measuring bone mineral density and bone biomechanical properties. In vitro, RAW264.7 cells were treated with receptor activator of nuclear factor kappa-B ligand (RANKL) and macrophage colony-stimulating factor (M-CSF) to induce osteoclast differentiation. Cell viability assays, tartrate-resistant acid phosphatase (TRAP) staining, and immunofluorescence were used to elucidate the effects of EA on osteoclastogenesis. In addition, the expression of bone differentiation-related proteins or genes was evaluated using Western blot analysis or quantitative polymerase chain reaction (PCR), respectively. RESULTS: After 3 months of oral EA intervention, ovariectomized rats exhibited increased bone density, relative bone volume, trabecular thickness, and trabecular number, as well as reduced trabecular separation. EA dose-dependently normalized bone density and trabecular microarchitecture in the ovariectomized rats. Additionally, EA inhibited the expression of TRAP and NFATc1 in the ovariectomized rats. Moreover, the in vitro results indicated that EA inhibits osteoclast differentiation by suppressing the TRAF6/PI3K/AKT/NF-κB pathway. Further studies revealed that the effect on osteoclast differentiation, which was originally inhibited by EA, was reversed when the TRAF6 gene was overexpressed. CONCLUSIONS: The findings indicated that EA can negatively regulate osteoclastogenesis by inhibiting the TRAF6/PI3K/AKT/NF-κB axis and that ameliorating ovariectomy-induced osteoporosis in rats with EA may be a promising potential therapeutic strategy for the treatment of osteoporosis.


Asunto(s)
Diferenciación Celular , FN-kappa B , Osteoclastos , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Factor 6 Asociado a Receptor de TNF , Animales , Factor 6 Asociado a Receptor de TNF/metabolismo , Factor 6 Asociado a Receptor de TNF/genética , Osteoclastos/metabolismo , FN-kappa B/metabolismo , Transducción de Señal/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Femenino , Fosfatidilinositol 3-Quinasas/metabolismo , Ratas , Ratones , Células RAW 264.7 , Flavonoides/farmacología , Osteogénesis/efectos de los fármacos , Ratas Sprague-Dawley , Osteoporosis/metabolismo , Osteoporosis/etiología , Ovariectomía/efectos adversos , Regulación de la Expresión Génica/efectos de los fármacos , Densidad Ósea/efectos de los fármacos
19.
Biochimie ; 2024 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-39154977

RESUMEN

Bone metabolism plays a crucial role in maintaining normal bone tissue homeostasis and function. Imbalances between bone formation and resorption can lead to osteoporosis, osteoarthritis, and other bone diseases. The dynamic and complex process of bone remodeling is driven by various factors, including epigenetics. Histone modification, one of the most important and well-studied components of epigenetic regulation, has emerged as a promising area of research in bone metabolism. Different histone proteins and modification sites exert diverse effects on osteogenesis and osteoclastogenesis. In this review, we summarize recent progress in understanding histone modifications in bone metabolism, including specific modification sites and potential regulatory enzymes. Comprehensive knowledge of histone modifications in bone metabolism could reveal new therapeutic targets and treatment strategies for bone diseases.

20.
Adv Sci (Weinh) ; : e2403177, 2024 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-39120025

RESUMEN

Epigenetic regulation of metabolism profoundly influences cell fate commitment. During osteoclast differentiation, the activation of RANK signaling is accompanied by metabolic reprogramming, but the epigenetic mechanisms by which RANK signaling induces this reprogramming remain elusive. By transcriptional sequence and ATAC analysis, this study identifies that activation of RANK signaling upregulates PRMT6 by epigenetic modification, triggering a metabolic switching from fatty acids oxidation toward glycolysis. Conversely, Prmt6 deficiency reverses this shift, markedly reducing HIF-1α-mediated glycolysis and enhancing fatty acid oxidation. Consequently, PRMT6 deficiency or inhibitor impedes osteoclast differentiation and alleviates bone loss in ovariectomized (OVX) mice. At the molecular level, Prmt6 deficiency reduces asymmetric dimethylation of H3R2 at the promoters of genes including Ppard, Acox3, and Cpt1a, enhancing genomic accessibility for fatty acid oxidation. PRMT6 thus emerges as a metabolic checkpoint, mediating metabolic switch from fatty acid oxidation to glycolysis, thereby supporting osteoclastogenesis. Unveiling PRMT6's critical role in epigenetically orchestrating metabolic shifts in osteoclastogenesis offers a promising target for anti-resorptive therapy.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA