Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.633
Filtrar
Más filtros











Intervalo de año de publicación
1.
BMC Pharmacol Toxicol ; 25(1): 40, 2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-38997762

RESUMEN

BACKGROUND: Periapical lesions are characterized by periapical inflammation and damage to periapical tissues and eventually lead to bone resorption and even tooth loss. H2O2 is widely used in root canal therapy for patients with periapical inflammation. Luteolin possesses high anti-inflammatory, antioxidant, and anticancer potential. However, the underlying mechanism of the efficacy of H2O2 and luteolin on oxidative stress and inflammatory tissue has not been previously addressed. We aimed to investigate the anti-inflammatory and antioxidative effects of luteolin on H2O2-induced cellular oxidative inflammation. METHODS: After human osteoblasts (hFOB1.19) were treated with lipopolysaccharide (LPS), luteolin, or H2O2, cell proliferation was analysed by using a cell counting kit-8 (CCK-8), cell apoptosis was measured by using flow cytometry, the production of reactive oxygen species (ROS) was evaluated by using an oxidation-sensitive probe DCFH-DA ROS assay kit, and the expression of genes and proteins was detected by using reverse transcription quantitative polymerase chain reaction (RT‒qPCR), Western blotting, and enzyme-linked immunosorbent assay (ELISA). RESULTS: We demonstrated that inflammation is closely related to oxidative stress and that the oxidative stress level in the inflammatory environment is increased. Luteolin inhibited the H2O2-induced increase in the expression of interleukin-6 (IL-6), interleukin-8 (IL-8) and tumour necrosis factor α (TNF-α) and significantly repressed the H2O2-induced increase in ROS, as well as markedly strengthened superoxide dismutase (SOD) activity in hFOB1.19 cells. Moreover, we detected that luteolin may inhibit H2O2-induced hFOB1.19 cell injury by suppressing the NF-κB pathway. CONCLUSION: We elucidated that luteolin protected human osteoblasts (hFOB1.19) from H2O2-induced cell injury and inhibited the production of proinflammatory cytokines by suppressing the NF-κB signalling pathway. Our findings provide a potential drug for treating H2O2-induced periodontitis and cell injury.


Asunto(s)
Antiinflamatorios , Peróxido de Hidrógeno , Inflamación , Luteolina , Osteoblastos , Estrés Oxidativo , Luteolina/farmacología , Humanos , Estrés Oxidativo/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Peróxido de Hidrógeno/toxicidad , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Línea Celular , Antiinflamatorios/farmacología , Apoptosis/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Lipopolisacáridos/farmacología , Proliferación Celular/efectos de los fármacos , Antioxidantes/farmacología , FN-kappa B/metabolismo , Microambiente Celular/efectos de los fármacos , Citocinas/metabolismo
2.
Int J Mol Sci ; 25(13)2024 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-39000355

RESUMEN

Postmenopausal osteoporosis, characterized by an imbalance between osteoclast-mediated bone resorption and osteoblast-driven bone formation, presents substantial health implications. In this study, we investigated the role of black goat extract (BGE), derived from a domesticated native Korean goat, estrogen-like activity, and osteoprotective effects in vitro. BGE's mineral and fatty acid compositions were analyzed via the ICP-AES method and gas chromatography-mass spectrometry, respectively. In vitro experiments were conducted using MCF-7 breast cancer cells, MC3T3-E1 osteoblasts, and RAW264.7 osteoclasts. BGE exhibits a favorable amount of mineral and fatty acid content. It displayed antimenopausal activity by stimulating MCF-7 cell proliferation and augmenting estrogen-related gene expression (ERα, ERß, and pS2). Moreover, BGE positively impacted osteogenesis and mineralization in MC3T3-E1 cells through Wnt/ß-catenin pathway modulation, leading to heightened expression of Runt-related transcription factor 2, osteoprotegerin, and collagen type 1. Significantly, BGE effectively suppressed osteoclastogenesis by curtailing osteoclast formation and activity in RAW264.7 cells, concurrently downregulating pivotal signaling molecules, including receptor activator of nuclear factor κ B and tumor necrosis factor receptor-associated factor 6. This study offers a shred of preliminary evidence for the prospective use of BGE as an effective postmenopausal osteoporosis treatment.


Asunto(s)
Diferenciación Celular , Cabras , Osteoblastos , Osteoclastos , Osteogénesis , Animales , Ratones , Células RAW 264.7 , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteogénesis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Osteoclastos/citología , Humanos , Estrógenos/farmacología , Proliferación Celular/efectos de los fármacos , Vía de Señalización Wnt/efectos de los fármacos , Células MCF-7 , Extractos de Tejidos/farmacología
3.
Chem Biol Drug Des ; 104(1): e14574, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38958121

RESUMEN

To develop novel bovine lactoferrin (bLF) peptides targeting bLF-tumour necrosis factor (TNF) receptor-associated factor 6 (TRAF6) binding sites, we identified two peptides that could target bLF-TRAF6 binding sites using structural analysis. Moreover, another peptide that could bind to the TRAF6 dimerization area was selected from the bLF sequence. The effects of each peptide on cytokine expression in lipopolysaccharide (LPS)-stimulated osteoblasts (ST2) and on osteoclastogenesis were examined using an LPS-treated co-culture of primary bone marrow cells (BMCs) with ST2 cells and a single culture of osteoclast precursor cells (RAW-D) treated with soluble receptor activator of NF-κB ligand. Finally, the effectiveness of these peptides against LPS-induced alveolar bone destruction was assessed. Two of the three peptides significantly suppressed LPS-induced TNF-α and interleukin-1ß expression in ST2 cells. Additionally, these peptides inhibited and reversed LPS-induced receptor activator of NF-κB ligand (RANKL) upregulation and osteoprotegerin (OPG) downregulation, respectively. Furthermore, both peptides significantly reduced LPS-induced osteoclastogenesis in the BMC-ST2 co-culture and RANKL-induced osteoclastogenesis in RAW-D cells. In vivo, topical application of these peptides significantly reduced the osteoclast number by downregulating RANKL and upregulating OPG in the periodontal ligament. It is indicated that the novel bLF peptides can be used to treat periodontitis-associated bone destruction.


Asunto(s)
Lactoferrina , Lipopolisacáridos , Osteoclastos , Péptidos , Animales , Lactoferrina/farmacología , Lactoferrina/química , Lactoferrina/metabolismo , Lipopolisacáridos/farmacología , Ratas , Péptidos/farmacología , Péptidos/química , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Ligando RANK/metabolismo , Masculino , Pérdida de Hueso Alveolar/tratamiento farmacológico , Pérdida de Hueso Alveolar/metabolismo , Pérdida de Hueso Alveolar/patología , Bovinos , Ratones , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteoblastos/citología , Ratas Sprague-Dawley , Osteogénesis/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo , Sitios de Unión , Técnicas de Cocultivo , Osteoprotegerina/metabolismo , Modelos Animales de Enfermedad
4.
J Cell Mol Med ; 28(13): e18527, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38984969

RESUMEN

Infected bone defects (IBDs) are the common condition in the clinical practice of orthopaedics. Although surgery and anti-infective medicine are the firstly chosen treatments, in many cases, patients experience a prolonged bone union process after anti-infective treatment. Epimedium-Curculigo herb pair (ECP) has been proved to be effective for bone repair. However, the mechanisms of ECP in IBDs are insufficiency. In this study, Effect of ECP in IBDs was verified by micro-CT and histological examination. Qualitative and quantitative analysis of the main components in ECP containing medicated serum (ECP-CS) were performed. The network pharmacological approaches were then applied to predict potential pathways for ECP associated with bone repair. In addition, the mechanism of ECP regulating LncRNA MALAT1/miRNA-34a-5p/SMAD2 signalling axis was evaluated by molecular biology experiments. In vivo experiments indicated that ECP could significantly promote bone repair. The results of the chemical components analysis and the pathway identification revealed that TGF-ß signalling pathway was related to ECP. The results of in vitro experiments indicated that ECP-CS could reverse the damage caused by LPS through inhibiting the expressions of LncRNA MALAT1 and SMAD2, and improving the expressions of miR-34a-5p, ALP, RUNX2 and Collagen type І in osteoblasts significantly. This research showed that ECP could regulate the TGF-ß/SMADs signalling pathway to promote bone repair. Meanwhile, ECP could alleviate LPS-induced bone loss by modulating the signalling axis of LncRNA MALAT1/miRNA-34a-5p/ SMAD2 in IBDs.


Asunto(s)
Epimedium , MicroARNs , Osteoblastos , ARN Largo no Codificante , Transducción de Señal , Proteína Smad2 , MicroARNs/genética , MicroARNs/metabolismo , Osteoblastos/metabolismo , Osteoblastos/efectos de los fármacos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Animales , Proteína Smad2/metabolismo , Proteína Smad2/genética , Ratones , Epimedium/química , Transducción de Señal/efectos de los fármacos , Masculino , Regeneración Ósea/efectos de los fármacos , Humanos , Regulación de la Expresión Génica/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Osteogénesis/genética
5.
Biomolecules ; 14(7)2024 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-39062525

RESUMEN

Peptide-based drug development is a promising direction due to its excellent biological activity, minimal immunogenicity, high in vivo stability, and efficient tissue penetrability. GV1001, an amphiphilic peptide, has proven effective as an anti-cancer vaccine, but its effect on osteoblast differentiation is unknown. To identify proteins interacting with GV1001, biotin-conjugated GV1001 was constructed and confirmed by mass spectrometry. Proteomic analyses were performed to determine GV1001's interaction with osteogenic proteins. GV1001 was highly associated with peptidyl-prolyl isomerase A and co-immunoprecipitation assays revealed that GV1001 bound to peptidyl-prolyl cis-trans isomerase 1 (Pin1). GV1001 significantly increased alkaline phosphatase (ALP) activity, bone nodule formation, and the expression of osteogenic gene markers. GV1001-induced osteogenic activity was enhanced by Pin1 overexpression and abolished by Pin1 knockdown. GV1001 increased the protein stability and transcriptional activity of Runx2 and Osterix. Importantly, GV1001 administration enhanced bone mass density in the OVX mouse model, as verified by µCT analysis. GV1001 demonstrated protective effects against bone loss in OVX mice by upregulating osteogenic differentiation via the Pin1-mediated protein stabilization of Runx2 and Osterix. GV1001 could be a potential candidate with anabolic effects for the prevention and treatment of osteoporosis.


Asunto(s)
Péptidos de Penetración Celular , Subunidad alfa 1 del Factor de Unión al Sitio Principal , Peptidilprolil Isomerasa de Interacción con NIMA , Osteogénesis , Factor de Transcripción Sp7 , Animales , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Osteogénesis/efectos de los fármacos , Peptidilprolil Isomerasa de Interacción con NIMA/metabolismo , Peptidilprolil Isomerasa de Interacción con NIMA/genética , Ratones , Péptidos de Penetración Celular/farmacología , Péptidos de Penetración Celular/química , Factor de Transcripción Sp7/metabolismo , Factor de Transcripción Sp7/genética , Humanos , Femenino , Estabilidad Proteica/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteoblastos/citología
6.
Mol Biol Rep ; 51(1): 838, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-39042226

RESUMEN

BACKGROUND: Bioglass materials have gained significant attention in the field of tissue engineering due to their osteoinductive and biocompatible properties that promote bone cell differentiation. In this study, a novel composite scaffold was developed using a sol-gel technique to combine bioglass (BG) 58 S with a poly L-lactic acid (PLLA). METHODS AND RESULTS: The physiochemical properties, morphology, and osteoinductive potential of the scaffolds were investigated by X-ray diffraction analysis, scanning electron microscopy, and Fourier-transform infrared spectroscopy. The results showed that the SiO2-CaO-P2O5 system was successfully synthesized by the sol-gel method. The PLLA scaffolds containing BG was found to be osteoinductive and promoted mineralization, as demonstrated by calcium deposition assay, upregulation of alkaline phosphatase enzyme activity, and Alizarin red staining data. CONCLUSIONS: These in vitro studies suggest that composite scaffolds incorporating hBMSCs are a promising substitute material to be implemented in bone tissue engineering. The PLLA/BG scaffolds promote osteogenesis and support the differentiation of bone cells, such as osteoblasts, due to their osteoinductive properties.


Asunto(s)
Materiales Biocompatibles , Diferenciación Celular , Cerámica , Osteogénesis , Poliésteres , Ingeniería de Tejidos , Andamios del Tejido , Poliésteres/química , Andamios del Tejido/química , Cerámica/química , Cerámica/farmacología , Ingeniería de Tejidos/métodos , Materiales Biocompatibles/farmacología , Materiales Biocompatibles/química , Osteogénesis/efectos de los fármacos , Humanos , Diferenciación Celular/efectos de los fármacos , Regeneración Ósea/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Espectroscopía Infrarroja por Transformada de Fourier , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Difracción de Rayos X , Huesos/efectos de los fármacos , Huesos/metabolismo , Fosfatasa Alcalina/metabolismo , Microscopía Electrónica de Rastreo
7.
J Orthop Surg Res ; 19(1): 444, 2024 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-39075522

RESUMEN

BACKGROUND: Dysregulation of osteogenic differentiation is a crucial event during osteoporosis. The bioactive phytochemical icariin has become an anti-osteoporosis candidate. Here, we elucidated the mechanisms underlying the promoting function of icariin in osteogenic differentiation. METHODS: Murine pre-osteoblast MC3T3-E1 cells were stimulated with dexamethasone (DEX) to induce osteogenic differentiation, which was evaluated by an Alizarin Red staining assay and ALP activity measurement. The mRNA amounts of SPI1 and SMAD5 were detected by real-time quantitative PCR. Expression analysis of proteins, including osteogenic markers (OPN, OCN and RUNX2) and autophagy-associated proteins (LC3, Beclin-1, and ATG5), was performed by immunoblotting. The binding of SPI1 and the SMAD5 promoter was predicted by the Jaspar2024 algorithm and confirmed by chromatin immunoprecipitation (ChIP) experiments. The regulation of SPI1 in SMAD5 was examined by luciferase assays. RESULTS: During osteogenic differentiation of MC3T3-E1 cells, SPI1 and SMAD5 were upregulated. Functionally, SPI1 overexpression enhanced autophagy and osteogenic differentiation of MC3T3-E1 cells, while SMAD5 downregulation exhibited opposite effects. Mechanistically, SPI1 could enhance SMAD5 transcription and expression. Downregulation of SMAD5 also reversed SPI1 overexpression-induced autophagy and osteogenic differentiation in MC3T3-E1 cells. In MC3T3-E1 cells under DEX stimulation, icariin increased SMAD5 expression by upregulating SPI1. Furthermore, icariin could attenuate SPI1 depletion-imposed inhibition of autophagy and osteogenic differentiation of MC3T3-E1 cells. CONCLUSION: Our findings demonstrate that the SPI1/SMAD5 cascade, with the ability to enhance osteogenic differentiation, underlies the promoting effect of icariin on osteogenic differentiation of MC3T3-E1 cells.


Asunto(s)
Diferenciación Celular , Flavonoides , Osteoblastos , Osteogénesis , Proteína Smad5 , Flavonoides/farmacología , Animales , Ratones , Osteogénesis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proteína Smad5/metabolismo , Proteína Smad5/genética , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Autofagia/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Células 3T3
8.
Theranostics ; 14(10): 3945-3962, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38994035

RESUMEN

Rationale: NLRP3 inflammasome is critical in the development and progression of many metabolic diseases driven by chronic inflammation, but its effect on the pathology of postmenopausal osteoporosis (PMOP) remains poorly understood. Methods: We here firstly examined the levels of NLRP3 inflammasome in PMOP patients by ELISA. Then we investigated the possible mechanisms underlying the effect of NLRP3 inflammasome on PMOP by RNA sequencing of osteoblasts treated with NLRP3 siRNA and qPCR. Lastly, we accessed the effect of decreased NLRP3 levels on ovariectomized (OVX) rats. To specifically deliver NLRP3 siRNA to osteoblasts, we constructed NLRP3 siRNA wrapping osteoblast-specific aptamer (CH6)-functionalized lipid nanoparticles (termed as CH6-LNPs-siNLRP3). Results: We found that the levels of NLRP3 inflammasome were significantly increased in patients with PMOP, and were negatively correlated with estradiol levels. NLRP3 knock-down influenced signal pathways including immune system process, interferon signal pathway. Notably, of the top ten up-regulated genes in NLRP3-reduced osteoblasts, nine genes (except Mx2) were enriched in immune system process, and five genes were related to interferon signal pathway. The in vitro results showed that CH6-LNPs-siNLRP3 was relatively uniform with a dimeter of 96.64 ± 16.83 nm and zeta potential of 38.37 ± 1.86 mV. CH6-LNPs-siNLRP3 did not show obvious cytotoxicity and selectively delivered siRNA to bone tissue. Moreover, CH6-LNPs-siNLRP3 stimulated osteoblast differentiation by activating ALP and enhancing osteoblast matrix mineralization. When administrated to OVX rats, CH6-LNPs-siNLRP3 promoted bone formation and bone mass, improved bone microarchitecture and mechanical properties by decreasing the levels of NLRP3, IL-1ß and IL-18 and increasing the levels of OCN and Runx2. Conclusion: NLRP3 inflammasome may be a new biomarker for PMOP diagnosis and plays a key role in the pathology of PMOP. CH6-LNPs-siNLRP3 has potential application for the treatment of PMOP.


Asunto(s)
Inflamasomas , Liposomas , Proteína con Dominio Pirina 3 de la Familia NLR , Nanopartículas , Osteoblastos , Osteoporosis Posmenopáusica , Animales , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Femenino , Humanos , Ratas , Inflamasomas/metabolismo , Nanopartículas/química , Osteoporosis Posmenopáusica/metabolismo , Regulación hacia Abajo/efectos de los fármacos , Ratas Sprague-Dawley , ARN Interferente Pequeño/administración & dosificación , Aptámeros de Nucleótidos/farmacología , Aptámeros de Nucleótidos/administración & dosificación , Modelos Animales de Enfermedad , Persona de Mediana Edad , Ovariectomía
9.
Int J Med Sci ; 21(8): 1511-1517, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38903930

RESUMEN

Bone marrow-derived mesenchymal stem cells (MSCs), which are capable of differentiating into osteoblasts, are used in effective regenerative therapies. MSCs must be prompted to differentiate into osteoblasts for MSC transplantation to be effective. In this study, osteoblast differentiation markers involved in bone formation were evaluated to investigate the stress resistance of bone marrow-derived rat MSCs to dexamethasone and hypoxia and their ability to differentiate into osteoblasts. MSCs were allowed to differentiate into osteoblasts for 21 days in three different environments (dexamethasone treatment, hypoxic conditions [1% oxygen], or both). Osteoblast differentiation potential was evaluated according to alkaline phosphatase levels and a mineralisation assay. Immunofluorescence staining was used to determine the protein expression of the osteoblast differentiation markers type I collagen and osteopontin. MSCs differentiated into osteoblasts under hypoxic conditions but differentiated more slowly upon treatment with dexamethasone and dexamethasone plus hypoxia relative to the control. MSCs preconditioned with dexamethasone or hypoxia and then allowed to differentiate into osteoblasts under similar conditions differentiated comparably to control MSCs. MSCs that developed resistance to dexamethasone or hypoxia differentiated more quickly into osteoblasts than those that did not. The findings suggest that increasing the resistance of MSCs to stress by preconditioning them via dexamethasone or hypoxia exposure could result in more rapid differentiation into osteoblasts following transplantation.


Asunto(s)
Diferenciación Celular , Hipoxia de la Célula , Dexametasona , Células Madre Mesenquimatosas , Osteoblastos , Dexametasona/farmacología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Animales , Osteoblastos/efectos de los fármacos , Osteoblastos/citología , Osteoblastos/metabolismo , Diferenciación Celular/efectos de los fármacos , Ratas , Hipoxia de la Célula/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Células Cultivadas , Fosfatasa Alcalina/metabolismo , Humanos , Trasplante de Células Madre Mesenquimatosas/métodos , Colágeno Tipo I/metabolismo , Masculino
10.
J Mater Sci Mater Med ; 35(1): 33, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38900208

RESUMEN

Phosphate bioactive glass has been studied for its advanced biodegradability and active ion release capability. Our previous research found that phosphate glass containing (P2O5)-(Na2O)-(TiO2)-(CaO)-(SrO) or (ZnO) showed good biocompatibility with MG63 and hMSCs. This study further investigated the application of 5 mol% zinc oxide or 17.5 mol% strontium oxide in titanium-doped phosphate glass for bone tissue engineering. Ti-Ca-Na-Phosphate glasses, incorporating 5% zinc oxide or 17.5% strontium oxide, were made with melting quenching technology. The pre-osteoblast cell line MC3T3-E1 was cultured for indirect contact tests with graded diluted phosphate glass extractions and for direct contact tests by seeding cells on glass disks. The cell viability and cytotoxicity were analysed in vitro over 7 days. In vivo studies utilized the tibial defect model with or without glass implants. The micro-CT analysis was performed after surgery and then at 2, 6, and 12 weeks. Extractions from both zinc and strontium phosphate glasses showed no negative impact on MC3T3-E1 cell viability. Notably, non-diluted Zn-Ti-Ca-Na-phosphate glass extracts significantly increased cell viability by 116.8% (P < 0.01). Furthermore, MC3T3-E1 cells cultured with phosphate glass disks exhibited no increase in LDH release compared with the control group. Micro-CT images revealed that, over 12 weeks, both zinc-doped and strontium-doped phosphate glasses demonstrated good bone incorporation and longevity compared to the no-implant control. Titanium-doped phosphate glasses containing 5 mol% zinc oxide, or 17.5 mol% strontium oxide have promising application potential for bone regeneration research.


Asunto(s)
Regeneración Ósea , Supervivencia Celular , Vidrio , Fosfatos , Estroncio , Titanio , Estroncio/química , Estroncio/farmacología , Regeneración Ósea/efectos de los fármacos , Animales , Ratones , Fosfatos/química , Fosfatos/farmacología , Vidrio/química , Titanio/química , Supervivencia Celular/efectos de los fármacos , Ensayo de Materiales , Zinc/química , Línea Celular , Osteoblastos/efectos de los fármacos , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Ingeniería de Tejidos/métodos , Sustitutos de Huesos/química , Sustitutos de Huesos/farmacología , Microtomografía por Rayos X
11.
J Nanobiotechnology ; 22(1): 320, 2024 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-38849820

RESUMEN

Simultaneously modulating the inflammatory microenvironment and promoting local bone regeneration is one of the main challenges in treating bone defects. In recent years, osteoimmunology has revealed that the immune system plays an essential regulatory role in bone regeneration and that macrophages are critical components. In this work, a mussel-inspired immunomodulatory and osteoinductive dual-functional hydroxyapatite nano platform (Gold/hydroxyapatite nanocomposites functionalized with polydopamine - PDA@Au-HA) is developed to accelerate bone tissues regeneration by regulating the immune microenvironment. PDA coating endows nanomaterials with the ability to scavenge reactive oxygen species (ROS) and anti-inflammatory properties, and it also exhibits an immunomodulatory ability to inhibit M1 macrophage polarization and activate M2 macrophage secretion of osteogenesis-related cytokines. Most importantly, this nano platform promotes the polarization of M2 macrophages and regulates the crosstalk between macrophages and pre-osteoblast cells to achieve bone regeneration. Au-HA can synergistically promote vascularized bone regeneration through sustained release of Ca and P particles and gold nanoparticles (NPs). This nano platform has a synergistic effect of good compatibility, scavenging of ROS, and anti-inflammatory and immunomodulatory capability to accelerate the bone repair process. Thus, our research offers a possible therapeutic approach by exploring PDA@Au-HA nanocomposites as a bifunctional platform for tissue regeneration.


Asunto(s)
Bivalvos , Regeneración Ósea , Durapatita , Oro , Indoles , Macrófagos , Osteogénesis , Regeneración Ósea/efectos de los fármacos , Durapatita/química , Durapatita/farmacología , Animales , Ratones , Oro/química , Oro/farmacología , Bivalvos/química , Células RAW 264.7 , Macrófagos/efectos de los fármacos , Indoles/química , Indoles/farmacología , Osteogénesis/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Polímeros/química , Polímeros/farmacología , Nanocompuestos/química , Nanopartículas del Metal/química , Osteoblastos/efectos de los fármacos , Antiinflamatorios/farmacología , Antiinflamatorios/química , Factores Inmunológicos/farmacología , Factores Inmunológicos/química , Citocinas/metabolismo
12.
Biochem Biophys Res Commun ; 725: 150265, 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-38901225

RESUMEN

With the substantial increase in the overuse of glucocorticoids (GCs) in clinical medicine, the prevalence of glucocorticoid-induced osteonecrosis of the femoral head (GC-ONFH) continues to rise in recent years. However, the optimal treatment for GC-ONFH remains elusive. Rotating magnetic field (RMF), considered as a non-invasive, safe and effective approach, has been proved to have multiple beneficial biological effects including improving bone diseases. To verify the effects of RMF on GC-ONFH, a lipopolysaccharide (LPS) and methylprednisolone (MPS)-induced invivo rat model, and an MPS-induced invitro cell model have been employed. The results demonstrate that RMF alleviated bone mineral loss and femoral head collapse in GC-ONFH rats. Meanwhile, RMF reduced serum lipid levels, attenuated cystic lesions, raised the expression of anti-apoptotic proteins and osteoprotegerin (OPG), while suppressed the expression of pro-apoptotic proteins and nuclear factor receptor activator-κB (RANK) in GC-ONFH rats. Besides, RMF also facilitated the generation of ALP, attenuated apoptosis and inhibits the expression of pro-apoptotic proteins, facilitated the expression of OPG, and inhibited the expression of RANK in MPS-stimulated MC3T3-E1 cells. Thus, this study indicates that RMF can improve GC-ONFH in rat and cell models, suggesting that RMF have the potential in the treatment of clinical GC-ONFH.


Asunto(s)
Diferenciación Celular , Necrosis de la Cabeza Femoral , Glucocorticoides , Osteoblastos , Ratas Sprague-Dawley , Animales , Osteoblastos/metabolismo , Osteoblastos/efectos de los fármacos , Necrosis de la Cabeza Femoral/inducido químicamente , Necrosis de la Cabeza Femoral/patología , Necrosis de la Cabeza Femoral/metabolismo , Necrosis de la Cabeza Femoral/terapia , Ratas , Diferenciación Celular/efectos de los fármacos , Masculino , Campos Magnéticos , Magnetoterapia/métodos , Cabeza Femoral/patología , Cabeza Femoral/metabolismo , Modelos Animales de Enfermedad , Rotación , Ratones
13.
Biomed Mater ; 19(5)2024 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-38917820

RESUMEN

Metastatic bone lesions are often osteolytic, which causes advanced-stage cancer sufferers to experience severe pain and an increased risk of developing a pathological fracture. Gallium (Ga) ion possesses antineoplastic and anti-bone resorption properties, suggesting the potential for its local administration to impede the growth of metastatic bone lesions. This study investigated the chemotherapeutic potential, cytotoxicity, and osteogenic effects of a Ga-doped glass polyalkenoate cement (GPC) (C-TA2) compared to its non-gallium (C-TA0) counterpart. Ion release profiles revealed a biphasic pattern characterized by an initial burst followed by a gradually declining release of ions. C-TA2 continued to release Ga steadily throughout the experimentation period (7 d) and exhibited prolonged zinc (Zn) release compared to C-TA0. Interestingly, the Zn release from both GPCs appeared to cause a chemotherapeutic effect against H1092 lung cancer cellsin vitro, with the prolonged Zn release from C-TA2 extending this effect. Unfortunately, both GPCs enhanced the viability of HCC2218 breast cancer cells, suggesting that the chemotherapeutic effects of Zn could be tied to cellular differences in preferred Zn concentrations. The utilization of SAOS-2 and MC3T3 cell lines as bone cell models yielded conflicting results, with the substantial decline in MC3T3 viability closely associated with silicon (Si) release, indicating cellular variations in Si toxicity. Despite this ambiguity, both GPCs exhibited harmful effects on the osteogenesis of primary rat osteoblasts, raising concerns about excessive burst Zn release. While Ga/Zn-doped GPCs hold promise for treating metastatic bone lesions caused by lung cancers, further optimization is required to mitigate cytotoxicity on healthy bone.


Asunto(s)
Supervivencia Celular , Galio , Osteogénesis , Galio/química , Animales , Humanos , Línea Celular Tumoral , Osteogénesis/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Ratones , Zinc/química , Ratas , Cementos de Ionómero Vítreo/química , Antineoplásicos/química , Antineoplásicos/farmacología , Ensayo de Materiales , Neoplasias Óseas/tratamiento farmacológico , Osteoblastos/efectos de los fármacos , Femenino , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Materiales Biocompatibles/química , Materiales Biocompatibles/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/patología
14.
PLoS One ; 19(6): e0301618, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38843277

RESUMEN

Periprosthetic tissue inflammation is a challenging complication arising in joint replacement surgeries, which is often caused by wear debris from polyethylene (PE) components. In this study, we examined the potential biological effects of grafting a [2-(methacryloyloxy)ethyl]dimethyl-(3-sulfopropyl)ammonium hydroxide (MEDSAH) polymer onto the surface of PE through a solvent-evaporation technique. J774A.1 macrophage-like cells and primary cultured mouse osteoblasts were treated with PE powder with or without the MEDSAH coating. MEDSAH grafting on PE substantially reduced the expression of pro-inflammatory cytokines and other mediators in primary cultured mouse osteoblasts, but did not significantly impact macrophage-mediated inflammation. Our findings suggest that a MEDSAH coating on PE-based materials has potential utility in mitigating periprosthetic tissue inflammation and osteolysis and preventing aseptic loosening in total joint replacements. Further research, including large-scale clinical trials and biomechanical analyses, is needed to assess the long-term performance and clinical implications of MEDSAH-coated PE-based materials in total joint arthroplasty.


Asunto(s)
Inflamación , Osteoblastos , Polietileno , Animales , Ratones , Inflamación/patología , Osteoblastos/metabolismo , Osteoblastos/efectos de los fármacos , Macrófagos/metabolismo , Línea Celular , Citocinas/metabolismo , Osteólisis/etiología , Osteólisis/patología , Materiales Biocompatibles Revestidos/química , Metacrilatos/química , Artroplastia de Reemplazo/efectos adversos
15.
PeerJ ; 12: e17488, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38827303

RESUMEN

Epigallocatechin gallate (EGCG), an active constituent of tea, is recognized for its anticancer and anti-inflammatory properties. However, the specific mechanism by which EGCG protects osteoblasts from cadmium-induced damage remains incompletely understood. Here, the action of EGCG was investigated by exposing MC3T3-E1 osteoblasts to EGCG and CdCl2 and examining their growth, apoptosis, and differentiation. It was found that EGCG promoted the viability of cadmium-exposed MC3T3-E1 cells, mitigated apoptosis, and promoted both maturation and mineralization. Additionally, CdCl2 has been reported to inhibit both the phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) and nuclear factor erythroid 2-related factor 2/heme oxygenase-1(Nrf2/HO-1) signaling pathways. EGCG treatment attenuated cadmium-induced apoptosis in osteoblasts and restored their function by upregulating both signaling pathways. The findings provide compelling evidence for EGCG's role in attenuating cadmium-induced osteoblast apoptosis and dysfunction through activating the PI3K/AKT/mTOR and Nrf2/HO-1 pathways. This suggests the potential of using EGCG for treating cadmium-induced osteoblast dysfunction.


Asunto(s)
Apoptosis , Catequina , Hemo-Oxigenasa 1 , Factor 2 Relacionado con NF-E2 , Osteoblastos , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Serina-Treonina Quinasas TOR , Catequina/análogos & derivados , Catequina/farmacología , Apoptosis/efectos de los fármacos , Factor 2 Relacionado con NF-E2/metabolismo , Animales , Ratones , Serina-Treonina Quinasas TOR/metabolismo , Transducción de Señal/efectos de los fármacos , Hemo-Oxigenasa 1/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Cadmio/toxicidad , Diferenciación Celular/efectos de los fármacos , Línea Celular , Proteínas de la Membrana
16.
Nanoscale ; 16(27): 12934-12946, 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38913123

RESUMEN

Polyetheretherketone (PEEK), renowned for its exceptional mechanical properties and bio-stability, is considered a promising alternative to traditional metal-based implants. However, the inferior bactericidal activity and the limited angiogenic and osteogenic properties of PEEK remain the three major obstacles to osseointegration in vivo. To overcome these obstacles, in this work, a versatile heterostructured nanocoating was conceived and equipped on PEEK. This nanocoating was designed to endow PEEK with the ability of photo-activated pathogen disinfection, along with enhanced angiogenesis and osteogenesis, effectively addressing the triple-barrier challenge towards osseointegration. The crafted nanocoating, encompassing diverse nutritional metal elements (Fe3+, Mg2+, and Sr2+) and a fusion peptide adept at promoting angiogenesis and osteogenesis, was seamlessly decorated onto PEEK. The engineered implant exhibited an antibacterial activity of over 94% upon near-infrared illumination by virtue of the photothermal conversion of the polyphenol nanocoating. Simultaneously, the decorated hierarchical nanocoatings synergistically promoted cellular adhesion and proliferation and up-regulated angiogenesis-/osteogenesis-associated cytokine expression in endothelial/osteoblast cells, resulting in superior angiogenic differentiation and osteoinductive capability in vitro. Moreover, an in vivo assay in a rabbit femoral defect model revealed that the decorated implant can achieve ameliorative osseointegrative fixation. Collectively, this work offers a practical and instructive clinical strategy to address the triple-barrier challenge associated with PEEK-based implants.


Asunto(s)
Antibacterianos , Benzofenonas , Oseointegración , Polietilenglicoles , Polímeros , Animales , Oseointegración/efectos de los fármacos , Antibacterianos/farmacología , Antibacterianos/química , Conejos , Polímeros/química , Polímeros/farmacología , Polietilenglicoles/química , Humanos , Osteogénesis/efectos de los fármacos , Prótesis e Implantes , Péptidos/química , Péptidos/farmacología , Materiales Biocompatibles Revestidos/química , Materiales Biocompatibles Revestidos/farmacología , Staphylococcus aureus/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Osteoblastos/citología , Osteoblastos/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Cetonas/química , Cetonas/farmacología , Proliferación Celular/efectos de los fármacos , Línea Celular , Escherichia coli/efectos de los fármacos , Adhesión Celular/efectos de los fármacos
17.
Nat Commun ; 15(1): 5027, 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38871693

RESUMEN

Generating 3D bone cell networks in vitro that mimic the dynamic process during early bone formation remains challenging. Here, we report a synthetic biodegradable microporous hydrogel for efficient formation of 3D networks from human primary cells, analysis of cell-secreted extracellular matrix (ECM) and microfluidic integration. Using polymerization-induced phase separation, we demonstrate dynamic in situ formation of microporosity (5-20 µm) within matrix metalloproteinase-degradable polyethylene glycol hydrogels in the presence of living cells. Pore formation is triggered by thiol-Michael-addition crosslinking of a viscous precursor solution supplemented with hyaluronic acid and dextran. The resulting microporous architecture can be fine-tuned by adjusting the concentration and molecular weight of dextran. After encapsulation in microporous hydrogels, human mesenchymal stromal cells and osteoblasts spread rapidly and form 3D networks within 24 hours. We demonstrate that matrix degradability controls cell-matrix remodeling, osteogenic differentiation, and deposition of ECM proteins such as collagen. Finally, we report microfluidic integration and proof-of-concept osteogenic differentiation of 3D cell networks under perfusion on chip. Altogether, this work introduces a synthetic microporous hydrogel to efficiently differentiate 3D human bone cell networks, facilitating future in vitro studies on early bone development.


Asunto(s)
Técnicas de Cultivo Tridimensional de Células , Diferenciación Celular , Matriz Extracelular , Hidrogeles , Células Madre Mesenquimatosas , Osteoblastos , Osteogénesis , Humanos , Hidrogeles/química , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Matriz Extracelular/metabolismo , Porosidad , Técnicas de Cultivo Tridimensional de Células/métodos , Polietilenglicoles/química , Ingeniería de Tejidos/métodos , Ácido Hialurónico/química , Células Cultivadas , Andamios del Tejido/química , Dextranos/química
18.
Int J Mol Sci ; 25(12)2024 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-38928460

RESUMEN

Osteoporosis, a prevalent chronic health issue among the elderly, is a global bone metabolic disease. Flavonoids, natural active compounds widely present in vegetables, fruits, beans, and cereals, have been reported for their anti-osteoporotic properties. Onion is a commonly consumed vegetable rich in flavonoids with diverse pharmacological activities. In this study, the trabecular structure was enhanced and bone mineral density (BMD) exhibited a twofold increase following oral administration of onion flavonoid extract (OFE). The levels of estradiol (E2), calcium (Ca), and phosphorus (P) in serum were significantly increased in ovariectomized (OVX) rats, with effects equal to alendronate sodium (ALN). Alkaline phosphatase (ALP) and tartrate-resistant acid phosphatase (TRAP) levels in rat serum were reduced by 35.7% and 36.9%, respectively, compared to the OVX group. In addition, the effects of OFE on bone health were assessed using human osteoblast-like cells MG-63 and osteoclast precursor RAW 264.7 cells in vitro as well. Proliferation and mineralization of MG-63 cells were promoted by OFE treatment, along with increased ALP activity and mRNA expression of osteoprotegerin (OPG)/receptor activator of nuclear factor-kappaB ligand (RANKL). Additionally, RANKL-induced osteoclastogenesis and osteoclast activity were inhibited by OFE treatment through decreased TRAP activity and down-regulation of mRNA expression-related enzymes in RAW 264.7 cells. Overall findings suggest that OFE holds promise as a natural functional component for alleviating osteoporosis.


Asunto(s)
Flavonoides , Cebollas , Osteoblastos , Osteogénesis , Osteoporosis , Extractos Vegetales , Animales , Femenino , Humanos , Ratones , Ratas , Densidad Ósea/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Flavonoides/farmacología , Cebollas/química , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Osteogénesis/efectos de los fármacos , Osteoporosis/tratamiento farmacológico , Osteoporosis/metabolismo , Osteoporosis/patología , Osteoprotegerina/metabolismo , Osteoprotegerina/genética , Ovariectomía , Extractos Vegetales/farmacología , Extractos Vegetales/química , Ligando RANK/metabolismo , Ratas Sprague-Dawley , Células RAW 264.7
19.
Toxicol Lett ; 398: 127-139, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38914176

RESUMEN

Increasing epidemiological evidence has shown that PM2.5 exposure is significantly associated with the occurrence of osteoporosis. It has been well demonstrated that PM2.5 exposure enhanced the differentiation and function of osteoclasts by indirectly causing chronic inflammation, while the mechanism in osteoblasts remains unclear. In our study, toxic effects were evaluated by direct exposure of 20-80 µg/ml PM2.5 to MC3T3-E1 cells and BMSCs. The results showed that PM2.5 exposure did not affect cell viability via proliferation and apoptosis, but significantly inhibited osteoblast differentiation in a dose-dependent manner. Osteogenic transcription factors Runx2 and Sp7 and other biomarkers Alp and Ocn decreased after PM2.5 exposure. RNA-seq revealed TGF-ß signaling was involved in PM2.5 exposure inhibited osteoblast differentiation, which led to P-Smad1/5 and P-Smad2 reduction in the nucleus by increasing the ubiquitination and degradation of Smad4. At last, the inflammation response increased in MC3T3-E1 cells with PM2.5 exposure. Moreover, the mRNA levels of Mmp9 increased in bone marrow-derived macrophage cells treated with the conditional medium collected from MC3T3-E1 cells exposed to PM2.5. Overall, these results indicated that PM2.5 exposure inhibits osteoblast differentiation and concurrently increases the maturation of osteoclasts. Our study provides in-depth mechanistic insights into the direct impact of PM2.5 exposure on osteoblast, which would indicate the unrecognized role of PM2.5 on osteoporosis.


Asunto(s)
Diferenciación Celular , Osteoblastos , Material Particulado , Proteína Smad4 , Ubiquitinación , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Proteína Smad4/metabolismo , Proteína Smad4/genética , Ratones , Material Particulado/toxicidad , Ubiquitinación/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Contaminantes Atmosféricos/toxicidad , Línea Celular , Supervivencia Celular/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Proteolisis/efectos de los fármacos
20.
Biomed Pharmacother ; 177: 117032, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38941894

RESUMEN

In cell-based bone augmentation, transplanted cell dysfunction and apoptosis can occur due to oxidative stress caused by the overproduction of reactive oxygen species (ROS). Edaravone (EDA) is a potent free radical scavenger with potential medical applications. This study aimed to investigate the effect of controlling oxidative stress on bone regeneration using EDA. Bone marrow-derived cells were collected from 4-week-old rats, and EDA effects on cell viability and osteogenic differentiation were evaluated. Collagen gels containing PKH26-prelabeled cells were implanted into the calvarial defects of 12-week-old rats, followed by daily subcutaneous injections of normal saline or 500 µM EDA for 4 d. Bone formation was examined using micro-computed tomography and histological staining. Immunofluorescence staining was performed for markers of oxidative stress, macrophages, osteogenesis, and angiogenesis. EDA suppressed ROS production and hydrogen peroxide-induced apoptosis, recovering cell viability and osteoblast differentiation. EDA treatment in vivo increased new bone formation. EDA induced the transition of the macrophage population toward the M2 phenotype. The EDA group also exhibited stronger immunofluorescence for vascular endothelial growth factor and CD31. In addition, more PKH26-positive and PKH26-osteocalcin-double-positive cells were observed in the EDA group, indicating that transplanted cell survival was prolonged, and they differentiated into bone-forming cells. This could be attributed to oxidative stress suppression at the transplantation site by EDA. Collectively, local administration using EDA facilitates bone regeneration by improving the local environment and angiogenesis, prolonging survival, and enhancing the osteogenic capabilities of transplanted cells.


Asunto(s)
Regeneración Ósea , Diferenciación Celular , Supervivencia Celular , Edaravona , Osteogénesis , Oxidación-Reducción , Estrés Oxidativo , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno , Animales , Regeneración Ósea/efectos de los fármacos , Edaravona/farmacología , Osteogénesis/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Ratas , Supervivencia Celular/efectos de los fármacos , Masculino , Apoptosis/efectos de los fármacos , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Cráneo/efectos de los fármacos , Células Cultivadas , Depuradores de Radicales Libres/farmacología , Depuradores de Radicales Libres/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA