Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20.498
Filtrar
2.
Development ; 151(17)2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-39250530

RESUMO

Developing bones can adapt their shape in response to mechanical stresses from neighbouring growing organs. In a new study, Koichi Matsuo and colleagues examine how bone-forming osteoblasts and bone-resorbing osteoclasts coordinate growth in the mouse fibula. They describe the process called 'endo-forming trans-pairing', where bone resorption by osteoclasts in the outer periosteum is paired with bone formation by osteoblasts in the inner endosteum to shape the growing bone. To learn more about the story behind the paper, we caught up with first author Yukiko Kuroda and the corresponding author Koichi Matsuo, Professor at the School of Medicine, Keio University, Japan.


Assuntos
Osso e Ossos , Microscopia/métodos , Osso e Ossos/citologia , Osteoclastos/citologia , Osteoclastos/metabolismo , Osteoblastos/citologia , Osteoblastos/metabolismo , Nervo Isquiático , Desenvolvimento Ósseo , Animais , Morfogênese
3.
Bone Res ; 12(1): 52, 2024 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-39231935

RESUMO

Osteoporosis remains incurable. The most widely used antiresorptive agents, bisphosphonates (BPs), also inhibit bone formation, while the anabolic agent, teriparatide, does not inhibit bone resorption, and thus they have limited efficacy in preventing osteoporotic fractures and cause some side effects. Thus, there is an unmet need to develop dual antiresorptive and anabolic agents to prevent and treat osteoporosis. Hydroxychloroquine (HCQ), which is used to treat rheumatoid arthritis, prevents the lysosomal degradation of TNF receptor-associated factor 3 (TRAF3), an NF-κB adaptor protein that limits bone resorption and maintains bone formation. We attempted to covalently link HCQ to a hydroxyalklyl BP (HABP) with anticipated low antiresorptive activity, to target delivery of HCQ to bone to test if this targeting increases its efficacy to prevent TRAF3 degradation in the bone microenvironment and thus reduce bone resorption and increase bone formation, while reducing its systemic side effects. Unexpectedly, HABP-HCQ was found to exist as a salt in aqueous solution, composed of a protonated HCQ cation and a deprotonated HABP anion. Nevertheless, it inhibited osteoclastogenesis, stimulated osteoblast differentiation, and increased TRAF3 protein levels in vitro. HABP-HCQ significantly inhibited both osteoclast formation and bone marrow fibrosis in mice given multiple daily PTH injections. In contrast, HCQ inhibited marrow fibrosis, but not osteoclast formation, while the HABP alone inhibited osteoclast formation, but not fibrosis, in the mice. HABP-HCQ, but not HCQ, prevented trabecular bone loss following ovariectomy in mice and, importantly, increased bone volume in ovariectomized mice with established bone loss because HABP-HCQ increased bone formation and decreased bone resorption parameters simultaneously. In contrast, HCQ increased bone formation, but did not decrease bone resorption parameters, while HABP also restored the bone lost in ovariectomized mice, but it inhibited parameters of both bone resorption and formation. Our findings suggest that the combination of HABP and HCQ could have dual antiresorptive and anabolic effects to prevent and treat osteoporosis.


Assuntos
Conservadores da Densidade Óssea , Reabsorção Óssea , Difosfonatos , Hidroxicloroquina , Ovariectomia , Animais , Ovariectomia/efeitos adversos , Feminino , Camundongos , Hidroxicloroquina/farmacologia , Hidroxicloroquina/uso terapêutico , Difosfonatos/farmacologia , Difosfonatos/uso terapêutico , Reabsorção Óssea/prevenção & controle , Reabsorção Óssea/tratamento farmacológico , Reabsorção Óssea/metabolismo , Conservadores da Densidade Óssea/farmacologia , Conservadores da Densidade Óssea/uso terapêutico , Camundongos Endogâmicos C57BL , Anabolizantes/farmacologia , Anabolizantes/uso terapêutico , Osteogênese/efeitos dos fármacos , Osteoporose/tratamento farmacológico , Osteoporose/prevenção & controle , Osteoporose/metabolismo , Osteoporose/patologia , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo
4.
Immun Inflamm Dis ; 12(9): e70011, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39264247

RESUMO

BACKGROUND: Bone regeneration is a well-regulated dynamic process, of which the prominent role of the immune system on bone homeostasis is more and more revealed by recent research. Before fully activation of the bone remodeling cells, the immune system needs to clean up the microenvironment in facilitating the bone repair initiation. Furthermore, this microenvironment must be maintained properly by various mechanisms over the entire bone regeneration process. OBJECTIVE: This review aims to summarize the role of the T-helper 17/Regulatory T cell (Th17/Treg) balance in bone cell remodeling and discuss the relevant progress in bone tissue engineering. RESULTS: The role of the immune response in the early stages of bone regeneration is crucial, especially the impact of the Th17/Treg balance on osteoclasts, mesenchymal stem cells (MSCs), and osteoblasts activity. By virtue of these knowledge advancements, innovative approaches in bone tissue engineering, such as nano-structures, hydrogel, and exosomes, are designed to influence the Th17/Treg balance and thereby augment bone repair and regeneration. CONCLUSION: Targeting the Th17/Treg balance is a promising innovative strategy for developing new treatments to enhance bone regeneration, thus offering potential breakthroughs in bone injury clinics.


Assuntos
Regeneração Óssea , Osso e Ossos , Linfócitos T Reguladores , Células Th17 , Engenharia Tecidual , Humanos , Linfócitos T Reguladores/imunologia , Engenharia Tecidual/métodos , Regeneração Óssea/imunologia , Animais , Células Th17/imunologia , Osso e Ossos/imunologia , Células-Tronco Mesenquimais/imunologia , Células-Tronco Mesenquimais/metabolismo , Remodelação Óssea/imunologia , Osteoblastos/imunologia , Osteoclastos/imunologia , Osteoclastos/metabolismo
5.
Int J Mol Sci ; 25(17)2024 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-39273569

RESUMO

G protein-coupled receptor (GPR)40 and GPR120 are receptors for medium- and long-chain free fatty acids. It has been well documented that GPR40 and GPR120 activation improves metabolic syndrome (MetS) and exerts anti-inflammatory effects. Since chronic periodontitis is a common oral inflammatory disease initiated by periodontal pathogens and exacerbated by MetS, we determined if GPR40 and GPR120 activation with agonists improves MetS-associated periodontitis in animal models in this study. We induced MetS and periodontitis by high-fat diet feeding and periodontal injection of lipopolysaccharide, respectively, and treated mice with GW9508, a synthetic GPR40 and GPR120 dual agonist. We determined alveolar bone loss, osteoclast formation, and periodontal inflammation using micro-computed tomography, osteoclast staining, and histology. To understand the underlying mechanisms, we further performed studies to determine the effects of GW9508 on osteoclastogenesis and proinflammatory gene expression in vitro. Results showed that GW9508 improved metabolic parameters, including glucose, lipids, and insulin resistance. Results also showed that GW9508 improves periodontitis by reducing alveolar bone loss, osteoclastogenesis, and periodontal inflammation. Finally, in vitro studies showed that GW9508 inhibited osteoclast formation and proinflammatory gene secretion from macrophages. In conclusion, this study demonstrated for the first time that GPR40/GPR120 agonist GW9508 reduced alveolar bone loss and alleviated periodontal inflammation in mice with MetS-exacerbated periodontitis, suggesting that activating GPR40/GPR120 with agonist GW9508 is a potential anti-inflammatory approach for the treatment of MetS-associated periodontitis.


Assuntos
Síndrome Metabólica , Metilaminas , Periodontite , Propionatos , Receptores Acoplados a Proteínas G , Animais , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo , Camundongos , Síndrome Metabólica/tratamento farmacológico , Síndrome Metabólica/metabolismo , Síndrome Metabólica/complicações , Propionatos/farmacologia , Propionatos/uso terapêutico , Periodontite/tratamento farmacológico , Periodontite/metabolismo , Metilaminas/farmacologia , Masculino , Camundongos Endogâmicos C57BL , Perda do Osso Alveolar/tratamento farmacológico , Perda do Osso Alveolar/etiologia , Dieta Hiperlipídica/efeitos adversos , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Modelos Animais de Doenças , Osteogênese/efeitos dos fármacos
6.
PLoS One ; 19(9): e0309807, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39236007

RESUMO

This study explored the mechanism of curcumin (CUR) suppressing osteoclastogenesis and evaluated its effects on osteoarthritis (OA) mouse. Bone marrow-derived macrophages were isolated as osteoclast precursors. In the presence or absence of CUR, cell proliferation was detected by CCK-8, osteoclastogenesis was detected by tartrate-resistant acid phosphatase (TRAP) staining, F-actin rings formation was detected by immunofluorescence, bone resorption was detected by bone slices, IκBα, nuclear factor kappa-B (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways were detected using western blot, osteoclastogenesis-related gens were measured using quantitative polymerase chain reaction. A knee OA mouse model was designed by destabilizing the medial meniscus (DMM). Thirty-six male mice were divided into sham+vehicle, OA+vehicle, and OA+CUR groups. Mice were administered with or without CUR at 25 mg/kg/d from the first post-operative day until sacrifice. After 4 and 8 weeks of OA induction, micro-computed tomography was performed to analyze microstructure changes in subchondral bone, hematoxylin and eosin staining was performed to calculate the thickness of the calcified and hyaline cartilage layers, toluidine blue O staining was performed to assess the degenerated cartilage, TRAP-stained osteoclasts were counted, and NF-κB, phosphorylated Jun N-terminal Kinases (p-JNK), and receptor activator of nuclear factor κB ligand (RANKL) were detected using immunohistochemistry. CUR suppressed osteoclastogenesis and bone resorption without cytotoxicity. CUR restrained RANKL-induced activation of NF-κB, p-JNK and up-regulation of osteoclastogenesis-related genes. CUR delayed cartilage degeneration by suppressing osteoclastogenesis and bone resorption in early OA. The mechanism of CUR inhibiting osteoclastogenesis might be associated with NF-κB/JNK signaling pathway, indicating a novel strategy for OA treatment.


Assuntos
Curcumina , Sistema de Sinalização das MAP Quinases , NF-kappa B , Osteoclastos , Osteogênese , Animais , Camundongos , Masculino , NF-kappa B/metabolismo , Curcumina/farmacologia , Osteogênese/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Osteoartrite/tratamento farmacológico , Osteoartrite/metabolismo , Osteoartrite/patologia , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças , Reabsorção Óssea/tratamento farmacológico , Reabsorção Óssea/metabolismo , Reabsorção Óssea/patologia
7.
Front Endocrinol (Lausanne) ; 15: 1450007, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39290327

RESUMO

Oxytocin (OT) is a posterior pituitary hormone that, in addition to its role in regulating childbirth and lactation, also exerts direct regulatory effects on the skeleton through peripheral OT and oxytocin receptor (OTR). Bone marrow mesenchymal stem cells (BMSCs), osteoblasts (OB), osteoclasts (OC), chondrocytes, and adipocytes all express OT and OTR. OT upregulates RUNX2, BMP2, ALP, and OCN, thereby enhancing the activity of BMSCs and promoting their differentiation towards OB rather than adipocytes. OT also directly regulates OPG/RANKL to inhibit adipocyte generation, increase the expression of SOX9 and COMP, and enhance chondrocyte differentiation. OB can secrete OT, exerting influence on the surrounding environment through autocrine and paracrine mechanisms. OT directly increases OC formation through the NκB/MAP kinase signaling pathway, inhibits osteoclast proliferation by triggering cytoplasmic Ca2+ release and nitric oxide synthesis, and has a dual regulatory effect on OCs. Under the stimulation of estrogen, OB synthesizes OT, amplifying the biological effects of estrogen and OT. Mediated by estrogen, the OT/OTR forms a feedforward loop with OB. Apart from estrogen, OT also interacts with arginine vasopressin (AVP), prostaglandins (PGE2), leptin, and adiponectin to regulate bone metabolism. This review summarizes recent research on the regulation of bone metabolism by OT and OTR, aiming to provide insights into their clinical applications and further research.


Assuntos
Osso e Ossos , Ocitocina , Receptores de Ocitocina , Ocitocina/metabolismo , Humanos , Animais , Osso e Ossos/metabolismo , Receptores de Ocitocina/metabolismo , Receptores de Ocitocina/genética , Osteoblastos/metabolismo , Células-Tronco Mesenquimais/metabolismo , Osteoclastos/metabolismo , Condrócitos/metabolismo , Osteogênese/fisiologia
8.
Redox Rep ; 29(1): 2398891, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-39284587

RESUMO

OBJECTIVE: The study will be to observe the effect of Sodium butyrate (NaB) on bone loss in lipopolysaccharide (LPS)-treated rats. METHODS: In the rat model, we observed that changes in the expression of oxidative stress regulators, inflammatory markers and target genes were measured by immunofluorescence and RT-PCR after treatment. Changes in viability and osteogenesis of MC3T3-E1, osteoclast differentiation in RAW264.7 cells in the presence of LPS were evaluated using CCK-8, ALP staining, RES staining, and TRAP staining. RESULTS: In vitro experiments have shown that LPS-induced inhibition of JC-1, SIRT1, GPX1 and SOD2 is associated with increased levels of inflammation and oxidative stress. In addition, NaB has been found to suppress oxidative stress, inflammation and Mito SOX, promote osteogenic differentiation, and inhibit osteoclast differentiation. In addition, NaB significantly promoted SITR1 expression, repaired impaired bone metabolism, and improved bone strength and bone mineral density. CONCLUSION: Given all this experimental evidence, the results strongly suggest that NaB can restore osteogenic activity in the presence of LPS by reducing intracellular ROS, inhibiting osteoclast differentiation and reducing bone loss in LPS-treated rat models.


Assuntos
Ácido Butírico , Inflamação , Lipopolissacarídeos , Estresse Oxidativo , Animais , Lipopolissacarídeos/toxicidade , Lipopolissacarídeos/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ácido Butírico/farmacologia , Inflamação/metabolismo , Inflamação/tratamento farmacológico , Camundongos , Células RAW 264.7 , Osteogênese/efeitos dos fármacos , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Diferenciação Celular/efeitos dos fármacos , Densidade Óssea/efeitos dos fármacos , Masculino , Ratos Sprague-Dawley , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/metabolismo
9.
Front Immunol ; 15: 1425670, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39281679

RESUMO

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.


Assuntos
Reabsorção Óssea , Diferenciação Celular , Camundongos Transgênicos , Osteoclastos , Osteogênese , Receptores de IgG , Animais , Receptores de IgG/genética , Receptores de IgG/metabolismo , Camundongos , Osteoclastos/metabolismo , Osteogênese/genética , Reabsorção Óssea/genética , Reabsorção Óssea/metabolismo , Ligante RANK/metabolismo , Ligante RANK/genética , Artrite Experimental/imunologia , Artrite Experimental/genética , Transdução de Sinais , Artrite Reumatoide/metabolismo , Artrite Reumatoide/imunologia , Artrite Reumatoide/genética , Osteoporose/genética , Osteoporose/etiologia , Osteoporose/metabolismo
10.
J Agric Food Chem ; 72(37): 20383-20395, 2024 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-39238071

RESUMO

Postmenopausal osteoporosis (PMOP) arises from the disruption in bone remodeling caused by estrogen deficiency, leading to a heightened susceptibility to osteoporotic fractures in aging women. Tetrahydroberberine (THB) is a chemical compound extracted from Corydalis yanhusuo, a member of the traditional Chinese medicine series "Zhejiang eight taste", possessing a variety of pharmacological functions such as lowering lipids and preventing muscle atrophy. However, the impact of THB on PMOP has not been systematically explored. In vitro experiments supported that THB suppresses osteoclast formation and resorption of bone concentration-dependently. Further experiments confirmed that these inhibitory effects of THB were related to inhibition on expressions of osteoclast-specific genes, the mitogen-activated protein kinase (MAPK) pathway, and the nuclear factor kappa-B (NF-κB) pathway and an increased apoptosis level in mature osteoclasts. Additionally, THB treatment mitigated the ovariectomy-induced bone loss and improved the skeletal microarchitecture in vivo. In conclusion, THB has such potential to improve the PMOP status.


Assuntos
Apoptose , NF-kappa B , Osteoclastos , Osteogênese , Ovariectomia , Ligante RANK , Animais , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Apoptose/efeitos dos fármacos , Feminino , Ligante RANK/metabolismo , Ligante RANK/genética , Ovariectomia/efeitos adversos , Camundongos , Osteogênese/efeitos dos fármacos , Humanos , NF-kappa B/metabolismo , NF-kappa B/genética , Osteoporose Pós-Menopausa/prevenção & controle , Osteoporose Pós-Menopausa/metabolismo , Osteoporose Pós-Menopausa/fisiopatologia , Osteoporose Pós-Menopausa/genética , Osteoporose Pós-Menopausa/tratamento farmacológico , Alcaloides de Berberina/farmacologia , Camundongos Endogâmicos C57BL , Reabsorção Óssea/prevenção & controle , Reabsorção Óssea/tratamento farmacológico , Células RAW 264.7
11.
Mol Med ; 30(1): 151, 2024 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-39278948

RESUMO

Erythropoietin (EPO), expressed in red blood progenitor cells, primarily regulates erythropoiesis by binding to its receptor. Besides anemia, recent studies have identified new therapeutic indications for EPO that are not connected to red blood cell formation. Elevated EPO levels harm bone homeostasis in adult organisms and are associated with increased osteoclast; however, the underlying molecular mechanisms remain unclear. This study demonstrated that EPO enhanced osteoclast differentiation and bone resorption in vitro. We showed that EPO promoted osteoclast formation by up-regulating PPARγ expression through activating the Jak2/ERK signaling pathway. Consistently, PPARγ antagonists rescued the hyperactivation of osteoclasts due to EPO, while PPARγ agonists reversed the EMP9-mediated decrease in osteoclast differentiation. Further, exposing female mice to EPO for two months led to a decrease in bone mass and increased osteoclast numbers. The present results suggested that EPO promotes osteoclastogenesis by regulating the Jak2/ERK/ PPARγ signaling pathway. From a clinical perspective, the risk of compromised bone health should be considered when using EPO to treat anemia in post-operative patients with intertrochanteric fractures of the femur, as it could significantly impact the patient's recovery and quality of life.


Assuntos
Diferenciação Celular , Eritropoetina , Osteoclastos , PPAR gama , Eritropoetina/farmacologia , Eritropoetina/metabolismo , Animais , PPAR gama/metabolismo , Osteoclastos/metabolismo , Osteoclastos/efeitos dos fármacos , Camundongos , Feminino , Diferenciação Celular/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Janus Quinase 2/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Humanos , Regulação para Cima/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Reabsorção Óssea/metabolismo , Camundongos Endogâmicos C57BL
12.
Angle Orthod ; 94(5): 566-573, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-39230015

RESUMO

OBJECTIVES: To investigate whether the inhibition of 12/15-lipoxygenase (12/15-LOX), one of the core enzymes of the arachidonic acid cascade, suppresses orthodontically induced root resorption (OIRR), and examine the involvement of the hyaline degeneration of periodontal ligament cells and odontoclast differentiation. MATERIALS AND METHODS: The left maxillary first molars of 10-week-old male Wistar rats were moved mesially for 14 days using a closed-coil spring (25 cN) inserted between the first molar and incisor. The rats were intraperitoneally administered with a 12/15-LOX specific inhibitor (ML-351; 0.05 mmol/kg) daily in the experimental group or vehicle (dimethyl sulfoxide) in the control group. Tooth movement was measured using microcomputed tomography on day 14. The appearance of OIRR, hyaline degeneration, osteoclasts, and odontoclasts was evaluated via histological analysis. Immunohistochemical staining for receptor-activated NF-kB ligand (RANKL) and osteoprotegerin was performed. RESULTS: OIRR observed on day 14 in the control group was strongly suppressed by ML-351 treatment. Hyaline degeneration observed on the compression side on day 3 and the appearance of osteoclasts and odontoclasts on days 3 and 14 were significantly suppressed by ML-351. RANKL expression on day 3 was significantly suppressed by ML-351. These key processes in OIRR were substantially suppressed by ML-351 treatment. CONCLUSIONS: Inhibition of 12/15-LOX reduced OIRR by suppressing hyaline degeneration and subsequent odontoclast differentiation.


Assuntos
Araquidonato 12-Lipoxigenase , Araquidonato 15-Lipoxigenase , Inibidores de Lipoxigenase , Osteoclastos , Ratos Wistar , Reabsorção da Raiz , Técnicas de Movimentação Dentária , Animais , Masculino , Técnicas de Movimentação Dentária/métodos , Reabsorção da Raiz/etiologia , Reabsorção da Raiz/prevenção & controle , Reabsorção da Raiz/patologia , Ratos , Araquidonato 15-Lipoxigenase/metabolismo , Araquidonato 12-Lipoxigenase/metabolismo , Inibidores de Lipoxigenase/farmacologia , Inibidores de Lipoxigenase/uso terapêutico , Osteoclastos/efeitos dos fármacos , Microtomografia por Raio-X , Ligante RANK/metabolismo , Diferenciação Celular/efeitos dos fármacos , Ligamento Periodontal/efeitos dos fármacos , Ligamento Periodontal/patologia , Osteoprotegerina/metabolismo , Dente Molar
13.
Cell Mol Biol (Noisy-le-grand) ; 70(7): 186-192, 2024 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-39097878

RESUMO

This study aimed to explore the regulatory effect of remifentanil-mediated mitochondrial autophagy on osteoclast formation and further investigate its mechanism. Macrophage cell line RAW264.7 was taken and induced to differentiate into mature osteoclasts using nuclear factor kB receptor activating factor ligand (RANKL). The cell model was treated with different concentrations of remifentanil or down-regulated expression of mitochondrial autophagy-related gene PINK1. The survival, death and ROS production of osteoclasts were detected by CCK8 kit and flow cytometry, MMP level was detected by JC-1 method, mitochondrial morphology and autophagy were observed by transmission electron microscopy, and mitochondrial autophagy-related protein expression was detected by Western blot. The number of osteoclasts in the remifentanil-treated group was significantly reduced compared to the control group, accompanied by a reduction in reactive oxygen species (ROS) and mitochondrial membrane potential levels (MMP). Further results showed that remifentanil could significantly up-regulate the activity of PINK1/Parkin pathway, promote the occurrence of mitochondrial autophagy, and damaged mitochondria, and inhibit the formation of osteoclasts. Remifentanil successfully inhibited osteoclast formation by regulating mitochondrial autophagy mediated by PINK1/Parkin pathway. The results of this study revealed that remifentanil plays an important role in the physiology and pathology of osteoclasts, which may provide new ideas and strategies for the clinical treatment of remifentanil in tibial fractures.


Assuntos
Autofagia , Potencial da Membrana Mitocondrial , Mitocôndrias , Osteoclastos , Proteínas Quinases , Espécies Reativas de Oxigênio , Remifentanil , Ubiquitina-Proteína Ligases , Camundongos , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Animais , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteínas Quinases/metabolismo , Proteínas Quinases/genética , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Remifentanil/farmacologia , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitina-Proteína Ligases/genética , Células RAW 264.7 , Autofagia/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos
15.
Nutrients ; 16(15)2024 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-39125380

RESUMO

BACKGROUND: Excessive fluoride exposure induces skeletal fluorosis, but the specific mechanism responsible is still unclear. Therefore, this study aimed to identify the pathogenesis of fluoride-induced bone injuries. METHODS: We systematically searched fluoride-induced bone injury-related genes from five databases. Then, these genes were subjected to enrichment analyses. A TF (transcription factor)-mRNA-miRNA network and protein-protein interaction (PPI) network were constructed using Cytoscape, and the Human Protein Atlas (HPA) database was used to screen the expression of key proteins. The candidate pharmacological targets were predicted using the Drug Signature Database. RESULTS: A total of 85 studies were included in this study, and 112 osteoblast-, 35 osteoclast-, and 41 chondrocyte-related differential expression genes (DEGs) were identified. Functional enrichment analyses showed that the Atf4, Bcl2, Col1a1, Fgf21, Fgfr1 and Il6 genes were significantly enriched in the PI3K-Akt signaling pathway of osteoblasts, Mmp9 and Mmp13 genes were enriched in the IL-17 signaling pathway of osteoclasts, and Bmp2 and Bmp7 genes were enriched in the TGF-beta signaling pathway of chondrocytes. With the use of the TF-mRNA-miRNA network, the Col1a1, Bcl2, Fgfr1, Mmp9, Mmp13, Bmp2, and Bmp7 genes were identified as the key regulatory factors. Selenium methyl cysteine, CGS-27023A, and calcium phosphate were predicted to be the potential drugs for skeletal fluorosis. CONCLUSIONS: These results suggested that the PI3K-Akt signaling pathway being involved in the apoptosis of osteoblasts, with the IL-17 and the TGF-beta signaling pathways being involved in the inflammation of osteoclasts and chondrocytes in fluoride-induced bone injuries.


Assuntos
Apoptose , Fluoretos , Inflamação , Osteoblastos , Transdução de Sinais , Humanos , Fluoretos/efeitos adversos , Apoptose/efeitos dos fármacos , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Inflamação/induzido quimicamente , Transdução de Sinais/efeitos dos fármacos , MicroRNAs/metabolismo , MicroRNAs/genética , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Mapas de Interação de Proteínas , RNA Mensageiro/metabolismo , RNA Mensageiro/genética , Redes Reguladoras de Genes , Regulação da Expressão Gênica/efeitos dos fármacos , Doenças Ósseas/induzido quimicamente , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
16.
ACS Nano ; 18(33): 22431-22443, 2024 Aug 20.
Artigo em Inglês | MEDLINE | ID: mdl-39103298

RESUMO

Osteoclastic inhibition using antiresorptive bisphosphonates and osteogenic promotion using antisclerostin agents represent two distinct osteoporosis treatments in clinical practice, each individual treatment suffers from unsatisfactory therapeutic efficacy due to its indirect intervention in osteoclasis and promotion of osteogenesis simultaneously. Although this issue is anticipated to be resolved by drug synergism, a tempting carrier-free dual-medication nanoassembly remains elusive. Herein, we prepare such a nanoassembly made of antiresorptive alendronate (ALN) crystal and antisclerostin polyaptamer (Apt) via a nucleic acid-driven crystallization method. This nanoparticle can protect Apt from rapid nuclease degradation, avoid the high cytotoxicity of free ALN, and effectively concentrate in the cancellous bone by virtue of the bone-binding ability of DNA and ALN. More importantly, the acid microenvironment of cancellous bone triggers the disassociation of nanoparticles for sustained drug release, from which ALN inhibits the osteoclast-mediated bone resorption while Apt promotes osteogenic differentiation. Our work represents a pioneering demonstration of nucleic acid-driven crystallization of a bisphosphonate into a tempting carrier-free dual-medication nanoassembly. This inaugural advancement augments the antiosteoporosis efficacy through direct inhibition of osteoclasis and promotion of osteogenesis simultaneously and establishes a paradigm for profound understanding of the underlying synergistic antiosteoporosis mechanism of antiresorptive and antisclerostin components. It is envisioned that this study provides a highly generalizable strategy applicable to the tailoring of a diverse array of DNA-inorganic nanocomposites for targeted regulation of intricate pathological niches.


Assuntos
Alendronato , Cristalização , Osteoclastos , Osteogênese , Osteoporose , Alendronato/química , Alendronato/farmacologia , Osteogênese/efeitos dos fármacos , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Osteoporose/tratamento farmacológico , Animais , Camundongos , Conservadores da Densidade Óssea/farmacologia , Conservadores da Densidade Óssea/química , Aptâmeros de Nucleotídeos/química , Aptâmeros de Nucleotídeos/farmacologia , Células RAW 264.7 , Humanos , Sinergismo Farmacológico
17.
Chem Biol Interact ; 401: 111164, 2024 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-39111524

RESUMO

Ganoderic Acid A (GAA) has demonstrated beneficial effects in anti-inflammatory and anti-oxidative stress studies. However, it remains unknown whether GAA exerts positive impacts on bone loss induced by lipopolysaccharide (LPS). This study aims to investigate the influence of GAA on bone loss in LPS-treated rats. The study assesses changes in the viability and osteogenic potential of MC3T3-E1 cells, as well as osteoclast differentiation in RAW264.7 cells in the presence of LPS using CCK-8, ALP staining, AR staining, and Tartrate-resistant acid phosphatase (TRAP) staining. In vitro experiments indicate that LPS-induced inhibition of osteoclasts (OC) and Superoxide Dismutase 2 (SOD2) correlates with heightened levels of inflammation and oxidative stress. Furthermore, GAA has displayed the ability to alleviate oxidative stress and inflammation, enhance osteogenic differentiation, and suppress osteoclast differentiation. Animal experiment also proves that GAA notably upregulates SOD2 expression and downregulates TNF-α expression, leading to the restoration of impaired bone metabolism, improved bone strength, and increased bone mineral density. The collective experimental findings strongly suggest that GAA can enhance osteogenic activity in the presence of LPS by reducing inflammation and oxidative stress, hindering osteoclast differentiation, and mitigating bone loss in LPS-treated rat models.


Assuntos
Diferenciação Celular , Ácidos Heptanoicos , Inflamação , Lanosterol , Lipopolissacarídeos , Osteoclastos , Osteogênese , Estresse Oxidativo , Ratos Sprague-Dawley , Superóxido Dismutase , Animais , Lipopolissacarídeos/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Masculino , Camundongos , Ratos , Células RAW 264.7 , Superóxido Dismutase/metabolismo , Inflamação/metabolismo , Inflamação/tratamento farmacológico , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Diferenciação Celular/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Lanosterol/análogos & derivados , Lanosterol/farmacologia , Lanosterol/uso terapêutico , Ácidos Heptanoicos/farmacologia , Ácidos Heptanoicos/uso terapêutico , Densidade Óssea/efeitos dos fármacos , Fator de Necrose Tumoral alfa/metabolismo , Reabsorção Óssea/prevenção & controle , Reabsorção Óssea/tratamento farmacológico , Reabsorção Óssea/metabolismo
18.
Mol Med ; 30(1): 125, 2024 Aug 16.
Artigo em Inglês | MEDLINE | ID: mdl-39152382

RESUMO

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.


Assuntos
Diferenciação Celular , NF-kappa B , Osteoclastos , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Transdução de Sinais , Fator 6 Associado a Receptor de TNF , Animais , Fator 6 Associado a Receptor de TNF/metabolismo , Fator 6 Associado a Receptor de TNF/genética , Osteoclastos/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Feminino , Fosfatidilinositol 3-Quinases/metabolismo , Ratos , Camundongos , Células RAW 264.7 , Flavonoides/farmacologia , Osteogênese/efeitos dos fármacos , Ratos Sprague-Dawley , Osteoporose/metabolismo , Osteoporose/etiologia , Ovariectomia/efeitos adversos , Regulação da Expressão Gênica/efeitos dos fármacos , Densidade Óssea/efeitos dos fármacos
19.
Biomed Pharmacother ; 178: 117208, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39088966

RESUMO

Rosiglitazone (RSG), as an insulin-sensitizing drug to treat type 2 diabetes mellitus (T2DM) is reported to decrease bone quality and increase bone fracture risk. The multiple off-target effects of Resveratrol (RSV), a natural specific agonist of Sirtuin1 (Sirt1) with pro-osteoblastogenesis and anti-adipogenesis effects, on bone loss in T2DM are still under discussion. In this study, successfully ovariectomized rats were fed with high-fat diet and STZ (HFD/STZ) to induced T2DM mice. RSV alone, RSG alone or co-administration of RSV and RSG were given orally to T2DM rats for 8 weeks to determine whether RSV administration had any prevention effect on T2DM osteoporosis. Bone mesenchymal stem cells (BMSCs) and bone marrow­derived macrophages (BMMs) were cultured under high glucose condition and were induced to osteoblasts or adipocytes and osteoclasts, respectively. µCT and HE staining showed that in T2DM osteoporotic rats, RSV co-administration prevents RSG induced-bone loss. ELISA results confirmed that RSV suppressed osteoclast activity and promoted osteoblast activity in diabetic osteoporosis rats and RSG-administrated diabetic osteoporosis rats. In vitro study showed that RSV significantly reversed RSG induced inhibition on osteogenesis and promotion on adiopogenesis of BMSC under high glucose (HG). Moreover, RSV significantly reverse RSG induced osteoclast formation and mature under HG. Taken together, these findings uncover a previously unappreciated anti-osteoporosis effect of concomitant treatment with RSV in RSG-administrated diabetic rats, suggesting the clinical use of RSV as an adjuvant in the treatment of T2DM for preventing or reversing RSG administration-associated bone loss.


Assuntos
Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2 , Osteogênese , Osteoporose , Ratos Sprague-Dawley , Resveratrol , Rosiglitazona , Animais , Resveratrol/farmacologia , Rosiglitazona/farmacologia , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/tratamento farmacológico , Feminino , Osteoporose/tratamento farmacológico , Osteoporose/induzido quimicamente , Osteoporose/patologia , Osteoporose/prevenção & controle , Ratos , Osteogênese/efeitos dos fármacos , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/induzido quimicamente , Células-Tronco Mesenquimais/efeitos dos fármacos , Osteoblastos/efeitos dos fármacos , Hipoglicemiantes/farmacologia , Dieta Hiperlipídica/efeitos adversos , Osteoclastos/efeitos dos fármacos , Osteoclastos/patologia , Adipócitos/efeitos dos fármacos
20.
Biomed Pharmacother ; 178: 117271, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39121589

RESUMO

Osteoblast-mediated bone formation and osteoclast-mediated bone resorption are critical processes in bone metabolism. Annexin A, a calcium-phospholipid binding protein, regulates the proliferation and differentiation of bone cells, including bone marrow mesenchymal stem cells, osteoblasts, and osteoclasts, and has gradually become a marker gene for the diagnosis of osteoporosis. As calcium channel proteins, the annexin A family members are closely associated with mechanical stress, which can target annexins A1, A5, and A6 to promote bone cell differentiation. Despite the significant clinical potential of annexin A family members in bone metabolism, few studies have reported on these mechanisms. Therefore, based on a review of relevant literature, this article elaborates on the specific functions and possible mechanisms of annexin A family members in bone metabolism to provide new ideas for their application in the prevention and treatment of bone diseases, such as osteoporosis.


Assuntos
Osso e Ossos , Humanos , Animais , Osso e Ossos/metabolismo , Osteoporose/metabolismo , Anexinas/metabolismo , Anexinas/genética , Osteogênese/fisiologia , Osteogênese/genética , Diferenciação Celular , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Reabsorção Óssea/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA