Osteoporotic bone defect

  • 文章类型: Journal Article
    背景:由于过量的活性氧(ROS),骨质疏松性骨缺损的修复仍然具有挑战性。持续性炎症,以及成骨和破骨细胞之间的不平衡。方法:这里,可注射的H2释放水凝胶(镁@聚乙二醇-聚(乳酸-共-乙醇酸),Mg@PEG-PLGA)的开发旨在重塑具有挑战性的骨骼环境并加速骨质疏松性骨缺损的修复。结果:这种Mg@PEG-PLGA凝胶显示出优异的可注射性,形状适应性,和相变能力,可以通过微创注射填充不规则的骨缺损区域,并且可以原位转变为多孔支架以提供机械支撑。随着H2和镁离子的适当释放,2Mg@PEG-PLGA凝胶(装载2mgMg)通过减少细胞内ROS表现出显着的免疫调节作用,引导巨噬细胞向M2表型极化,抑制IκB/NF-κB信号通路。此外,体外实验表明,2Mg@PEG-PLGA凝胶抑制破骨细胞生成,同时促进成骨。最值得注意的是,在动物实验中,2Mg@PEG-PLGA凝胶通过清除ROS,抑制炎症和破骨细胞生成,显著促进体内骨质疏松性骨缺损的修复。结论:总体而言,我们的研究为H2释放镁基水凝胶作为修复骨质疏松性骨缺损的潜在植入物的设计和开发提供了重要见解。
    Background: The repair of osteoporotic bone defects remains challenging due to excessive reactive oxygen species (ROS), persistent inflammation, and an imbalance between osteogenesis and osteoclastogenesis. Methods: Here, an injectable H2-releasing hydrogel (magnesium@polyethylene glycol-poly(lactic-co-glycolic acid), Mg@PEG-PLGA) was developed to remodel the challenging bone environment and accelerate the repair of osteoporotic bone defects. Results: This Mg@PEG-PLGA gel shows excellent injectability, shape adaptability, and phase-transition ability, can fill irregular bone defect areas via minimally invasive injection, and can transform into a porous scaffold in situ to provide mechanical support. With the appropriate release of H2 and magnesium ions, the 2Mg@PEG-PLGA gel (loaded with 2 mg of Mg) displayed significant immunomodulatory effects through reducing intracellular ROS, guiding macrophage polarization toward the M2 phenotype, and inhibiting the IκB/NF-κB signaling pathway. Moreover, in vitro experiments showed that the 2Mg@PEG-PLGA gel inhibited osteoclastogenesis while promoting osteogenesis. Most notably, in animal experiments, the 2Mg@PEG-PLGA gel significantly promoted the repair of osteoporotic bone defects in vivo by scavenging ROS and inhibiting inflammation and osteoclastogenesis. Conclusions: Overall, our study provides critical insight into the design and development of H2-releasing magnesium-based hydrogels as potential implants for repairing osteoporotic bone defects.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    羟基磷灰石(HAp)涂层目前具有有限的治疗应用,因为它们缺乏抗感染,骨诱导性,和差的机械特性。在钛基板上,电化学沉积(ECD)用于构建具有抗菌和药物递送性能的锶(Sr)特征羟基磷灰石(HAp)/氧化石墨烯(GO)/利奈唑胺(LZ)纳米材料。通过X射线衍射分析(XRD)证实了新制备的纳米材料,傅里叶变换红外光谱(FTIR),和X射线光电子能谱(XPS)分析和形态学特征的扫描电子显微镜(SEM)分析。结果显示由于GO的2D表面上的含氧部分,SrHAp/GO/LZ复合涂层的多个成核位点。它被证明有利于成骨细胞的增殖和分化。具有SrHAp/GO/LZ涂层的LZ纳米复合材料的弹性模量和硬度分别提高了67%和121%,分别。从SrHAp/GO/LZ涂层释放LZ最初5小时,然后逐渐释放14小时,由于LZ的物理和化学吸附。SrHAp/GO/LZ涂层能有效抑制表皮葡萄球菌和金黄色葡萄球菌,抑制持续了三天,如抑制区和菌落计数测定所证明的。当MG-63电池涂有SrHAp/GO/LZ复合涂层时,他们的附着力,扩散,和分化大大提高,当涂覆纯钛。一种用于治疗和预防骨质疏松性骨缺损的新型表面工程纳米材料,SrHAp/GO/LZ,被证明具有很高的机械特性,优越的抗菌能力,和骨诱导性。
    Hydroxyapatite (HAp) coatings currently have limited therapeutic applications because they lack anti-infection, osteoinductivity, and poor mechanical characteristics. On the titanium substrate, electrochemical deposition (ECD) was used to construct the strontium (Sr)-featuring hydroxyapatite (HAp)/graphene oxides (GO)/linezolid (LZ) nanomaterial coated with antibacterial and drug delivery properties. The newly fabricated nanomaterials were confirmed by X-ray diffraction analysis (XRD), Fourier-transform infrared spectroscopy (FTIR), and X-ray photoelectron spectroscopy (XPS) analysis and morphological features were examined by scanning electron microscope (SEM) analysis. The results reveal multiple nucleation sites for SrHAp/GO/LZ composite coatings due to oxygen-comprising moieties on the 2D surface of GO. It was shown to be favorable for osteoblast proliferation and differentiation. The elastic modulus and hardness of LZ nanocomposite with SrHAp/GO/LZ coatings were increased by 67 % and 121 %, respectively. An initial 5 h burst of LZ release from the SrHAp/GO/LZ coating was followed by 14 h of gradual release, owing to LZ\'s physical and chemical adsorption. The SrHAp/GO/LZ coating effectively inhibited both S. epidermidis and S. aureus, and the inhibition lasted for three days, as demonstrated by the inhibition zone and colony count assays. When MG-63 cells are coated with SrHAp/GO/LZ composite coating, their adhesion, proliferation, and differentiation greatly improve when coated with pure titanium. A novel surface engineering nanomaterial for treating and preventing osteoporotic bone defects, SrHAp/GO/LZ, was shown to have high mechanical characteristics, superior antibacterial abilities, and osteoinductivity.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    骨质疏松性骨缺损的治疗仍然是一个巨大的临床挑战,因为骨质疏松症(OP)与氧化应激和高水平的活性氧(ROS)有关。对骨骼形成有害的条件。抗氧化纳米材料如硒纳米颗粒(SeNPs)由于其多效药理活性而对骨生成具有积极作用,其可以发挥抗氧化应激功能以防止骨丢失并促进OP中的骨再生。在当前的研究中,通过将聚(乳酸-碳酸酯)(PDT)和β-磷酸三钙(β-TCP)与SeNP引入一锅法的策略,是用来准备注射剂的,防崩溃,形状适应性和粘性骨移植替代材料(PDT-TCP-SE)。PDT-TCP-SE骨移植替代品在生物微环境中表现出足够的粘附力和骨诱导活性,血管生成作用和抗炎以及抗氧化作用在体外和体内。此外,PDT-TCP-SE可以通过Sirt1/Nrf2/GPX4抗氧化通路保护骨髓间充质干细胞免受erastin诱导的铁凋亡,which,在一起,证明了骨移植替代材料是一种新兴的生物材料,具有潜在的临床应用前景。重要声明:可注射,防崩溃,粘合剂,塑料和生物活性骨移植替代物成功合成。通过调节骨质疏松性骨缺损区域的氧化应激,将SeNPs与PDT结合到β-TCP原位再生新骨中。PDT-TCP-SE骨移植替代物降低了骨质疏松性骨缺损微环境中的高ROS水平。骨移植替代物还可以通过体外Sirt1/Nrf2/GPX4途径减轻氧化应激并抑制铁性凋亡。此外,PDT-TCP-SE骨移植替代物能缓解体内骨质疏松性骨缺损的炎症环境,促进骨再生。这种生物材料具有合成简单的优点,生物相容性,防崩溃,可注射,和调节氧化应激水平,在骨组织工程中具有潜在的应用价值。
    The treatment of osteoporotic bone defect remains a big clinical challenge because osteoporosis (OP) is associated with oxidative stress and high levels of reactive oxygen species (ROS), a condition detrimental for bone formation. Anti-oxidative nanomaterials such as selenium nanoparticles (SeNPs) have positive effect on osteogenesis owing to their pleiotropic pharmacological activity which can exert anti-oxidative stress functions to prevent bone loss and facilitate bone regeneration in OP. In the current study a strategy of one-pot method by introducing Poly (lactic acid-carbonate) (PDT) and β-Tricalcium Phosphate (β-TCP) with SeNPs, is developed to prepare an injectable, anti-collapse, shape-adaptive and adhesive bone graft substitute material (PDT-TCP-SE). The PDT-TCP-SE bone graft substitute exhibits sufficient adhesion in biological microenvironments and osteoinductive activity, angiogenic effect and anti-inflammatory as well as anti-oxidative effect in vitro and in vivo. Moreover, the PDT-TCP-SE can protect BMSCs from erastin-induced ferroptosis through the Sirt1/Nrf2/GPX4 antioxidant pathway, which, in together, demonstrated the bone graft substitute material as an emerging biomaterial with potential clinical application for the future treatment of osteoporotic bone defect. STATEMENT OF SIGNIFICANCE: Injectable, anti-collapse, adhesive, plastic and bioactive bone graft substitute was successfully synthesized. Incorporation of SeNPs with PDT into β-TCP regenerated new bone in-situ by moderating oxidative stress in osteoporotic bone defects area. The PDT-TCP-SE bone graft substitute reduced high ROS levels in osteoporotic bone defect microenvironment. The bone graft substitute could also moderate oxidative stress and inhibit ferroptosis via Sirt1/Nrf2/GPX4 pathway in vitro. Moreover, the PDT-TCP-SE bone graft substitute could alleviate the inflammatory environment and promote bone regeneration in osteoporotic bone defect in vivo. This biomaterial has the advantages of simple synthesis, biocompatibility, anti-collapse, injectable, and regulation of oxidative stress level, which has potential application value in bone tissue engineering.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    骨质疏松,以骨稳态不平衡为特征,是全球健康问题。骨质疏松患者骨缺损难以愈合。由于副作用和不精确的递送问题,用于骨质疏松性骨缺损的经典药物治疗具有不令人满意的功效。在这项研究中,磁性聚集诱导的骨靶向聚(乳酸-共-乙醇酸,合成了基于PLGA)的纳米载体(ZOL-PLGA@Yoda1/SPIO),以实现双靶向递送和精确的Piezo1激活治疗骨质疏松性骨缺损。Piezo1是一种重要的机械换能器,在调节骨稳态中起着关键作用。为了实现双目标属性,ZOL-PLGA@Yoda1/SPIO使用唑来膦酸盐(ZOL)装饰的PLGA制造,超顺磁性氧化铁(SPIO),和Piezo1通过乳液溶剂扩散法激活分子Yoda1。骨靶向分子介导和磁聚集诱导的特性可以共同有效地实现对局部骨缺损的精确递送。此外,Yoda1加载能够有针对性地和有效地模拟机械信号和激活Piezo1。体内和体外实验表明,ZOL-PLGA@Yoda1/SPIO可以激活骨质疏松小鼠骨缺损区域的Piezo1,通过YAP/β-catenin信号轴促进成骨,促进良好协调的成骨-血管生成耦合,并显着加速缺损内的骨骼重建,而没有明显的副作用。总的来说,这种新型的双靶向纳米载体为临床治疗骨质疏松性骨缺损提供了潜在的有效策略。
    Osteoporosis, characterized by an imbalance in bone homeostasis, is a global health concern. Bone defects are difficult to heal in patients with osteoporosis. Classical drug treatments for osteoporotic bone defects have unsatisfactory efficacy owing to side effects and imprecise delivery problems. In this study, a magnetic aggregation-induced bone-targeting poly(lactic-co-glycolic acid, PLGA)-based nanocarrier (ZOL-PLGA@Yoda1/SPIO) is synthesized to realize dual-targeted delivery and precise Piezo1-activated therapy for osteoporotic bone defects. Piezo1 is an important mechanotransducer that plays a key role in regulating bone homeostasis. To achieve dual-targeting properties, ZOL-PLGA@Yoda1/SPIO is fabricated using zoledronate (ZOL)-decorated PLGA, superparamagnetic iron oxide (SPIO), and Piezo1-activated molecule Yoda1 via the emulsion solvent diffusion method. Bone-targeting molecular mediation and magnetic aggregation-induced properties can jointly and effectively achieve precise delivery to localized bone defects. Moreover, Yoda1 loading enables targeted and efficient mimicking of mechanical signals and activation of Piezo1. Experiments in vivo and in vitro demonstrate that ZOL-PLGA@Yoda1/SPIO can activate Piezo1 in bone defect areas of osteoporotic mice, improve osteogenesis through YAP/β-catenin signaling axis, promote a well-coordinated osteogenesis-angiogenesis coupling, and significantly accelerate bone reconstruction within the defects without noticeable side effects. Overall, this novel dual-targeting nanocarrier provides a potentially effective strategy for the clinical treatment of osteoporotic bone defects.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    骨质疏松症的骨缺损通常存在过量的活性氧(ROS),异常炎症水平,不规则的形状和受损的骨再生能力;因此,骨质疏松性骨缺损难以修复。在这项研究中,一种可注射的热敏水凝胶聚(D,L-丙交酯-聚(乙二醇)-聚(D,含有白藜芦醇(Res)和地塞米松(DEX)的L-丙交酯(PLEL)系统旨在创造有利于骨质疏松性骨缺损中成骨的微环境。该PLEL水凝胶被注入并填充不规则缺陷区域,并在原位实现快速的溶胶-凝胶转变。Res具有很强的抗炎作用,可以有效清除受损部位多余的自由基,引导巨噬细胞极化到M2表型,调节免疫反应。此外,DEX可以促进成骨分化。体外实验表明,该水凝胶能有效促进间充质干细胞的成骨分化,去除过量的细胞内ROS,调节巨噬细胞极化以减少炎症反应。体内实验表明,水凝胶促进骨质疏松性骨缺损的再生并调节免疫反应。总的来说,这项研究证实了水凝胶可以通过协同调节骨损伤微环境来治疗骨质疏松性骨缺损,缓解炎症反应,促进成骨;因此,它代表了一种有希望的药物递送策略来修复骨质疏松性骨缺损。
    Bone defects in osteoporosis usually present excessive reactive oxygen species (ROS), abnormal inflammation levels, irregular shapes and impaired bone regeneration ability; therefore, osteoporotic bone defects are difficult to repair. In this study, an injectable thermosensitive hydrogel poly (D, L-lactide)-poly (ethylene glycol)- poly (D, L-lactide) (PLEL) system containing resveratrol (Res) and dexamethasone (DEX) is designed to create a microenvironment conducive to osteogenesis in osteoporotic bone defects. This PLEL hydrogel is injected and filled irregular defect areas and achieving a rapid sol-gel transition in situ. Res has a strong anti-inflammatory effects that can effectively remove excess free radicals at the damaged site, guide macrophage polarization to the M2 phenotype, and regulate immune responses. Additionally, DEX can promote osteogenic differentiation. In vitro experiments showed that the hydrogel effectively promoted osteogenic differentiation of mesenchymal stem cells, removed excess intracellular ROS, and regulated macrophage polarization to reduce inflammatory responses. In vivo experiments showed that the hydrogel promoted osteoporotic bone defect regeneration and modulated immune responses. Overall, this study confirmed that the hydrogel can treat osteoporotic bone defects by synergistically modulating bone damage microenvironment, alleviating inflammatory responses, and promoting osteogenesis; thus, it represents a promising drug delivery strategy to repair osteoporotic bone defects.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • 文章类型: Journal Article
    载药生物活性支架材料的局部应用是解决临床骨质疏松性骨缺损问题的重要方向之一。这项研究保留了天然3D生物活性支架的载药量和机械性能的优势。通过化学和自组装方法对支架进行功能修饰,并应用聚多巴胺(PDA)纳米颗粒和甲状旁腺激素相关肽-1(PTHrP-1)进行有效的局部载药。这项研究调查了新型生物活性支架对骨化的影响,破骨细胞生成,和巨噬细胞极化。这项工作阐明了支架在体外调节破骨细胞活性和新骨形成中的作用。对小动物OP骨缺损的建立和修复进行了进一步的研究,并初步验证了天然生物活性多孔支架材料促进OP骨缺损修复的潜力。制备安全、经济的抗OP骨修复材料为临床转化应用提供了理论依据。
    The local application of drug-loaded bioactive scaffold materials is one of the important directions to solve the clinical problem of osteoporotic (OP) bone defects. This study retains the advantages of drug loading and mechanical properties of natural 3D bioactive scaffolds. The scaffolds are functionally modified through chemical and self-assembly approaches with application of polydopamine (PDA) nanoparticles and parathyroid hormone-related peptide-1 (PTHrP-1) for efficient local drug loading. This study investigates the effects of the novel bioactive scaffolds on ossification, osteoclastogenesis, and macrophage polarization. This work elucidates the effects of the scaffolds in regulating osteoclastic activity and new bone formation in vitro. Further studies on the establishment and repair of OP bone defects in small animals are conducted, and the potential of natural bioactive porous scaffold materials to promote the repair of OP bone defects is initially verified. The preparation of safe and economical anti-OP bone repair material provides a theoretical basis for clinical translational applications.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    骨疾病和疾病的治疗可能是困难的,因为它的复杂性。每年有数百万患者需要骨骼替代品,这可能有助于他们从各种疾病中迅速康复。反映结构的合成骨替代物,化学,和骨基质结构的生物学特征将非常有帮助和高要求。在这项研究中,在这项研究中,用有机胶原蛋白和丝素蛋白结构(COL/SF/CaO-SiO2)基质的无机生物活性玻璃纳米颗粒用于创建多功能骨样纤维,我们在这里描述。纤维结构以与形成磷灰石和新组织的顺序相当的分层方式组织。COL和SF中的氨基与CaO-SiO2结合以稳定所得复合纳米纤维。形态和功能研究证实,平均尺寸为20±5nm的结晶CaO-SiO2纳米颗粒锚定在115±10nm的COL/SF纳米纤维基质上。X射线光电子能谱(XPS)结果证实了C,N,O,Ca,和Si在复合纤维中的原子百分比分别为59.46、3.30、20.25、3.38和13.61%。分别。与成骨细胞(Saos-2)的生物相容性检查表明,CAL/SF/CaO-SiO2复合纳米纤维具有增强的成骨活性。最后,CAL/SF/CaO-SiO2复合纳米纤维支架用于治疗大鼠骨质疏松性骨缺损,复合纳米纤维支架显著促进骨再生和血管化。这种新型纤维支架类别代表了用于复杂骨再生的先进材料设计的潜在突破。
    Bone disease and disorder treatment might be difficult because of its complicated nature. Millions of patients each year need bone substitutes that may help them recover quickly from a variety of illnesses. Synthetic bone replacements that mirror the structural, chemical, and biological features of bone matrix structure will be very helpful and in high demand. In this research, the inorganic bioactive glass nanoparticles matrixed with organic collagen and silk fibroin structure (COL/SF/CaO-SiO2) were used to create multifunctional bone-like fibers in this study, which we describe here. The fiber structure is organized in a layered fashion comparable to the sequence in which apatite and neo tissue are formed. The amino groups in COL and SF combined with CaO-SiO2 to stabilize the resulting composite nanofiber. Morphological and functional studies confirmed that crystalline CaO-SiO2 nanoparticles with average sizes of 20 ± 5 nm are anchored on a 115 ± 10 nm COL/SF nanofiber matrix. X-ray photoelectron spectroscopic (XPS) results confirmed the presence of C, N, O, Ca, and Si in the composite fiber with an atomic percentage of 59.46, 3.30, 20.25, 3.38 and 13.61%. respectively. The biocompatibility examination with osteoblast cells (Saos-2) revealed that the CAL/SF/CaO-SiO2 composite nanofiber had enhanced osteogenic activity. Finally, when the CAL/SF/CaO-SiO2 composite nanofiber scaffolds were used to treat an osteoporotic bone defect in a rat model, the composite nanofiber scaffolds significantly promoted bone regeneration and vascularization. This novel fibrous scaffold class represents a potential breakthrough in the design of advanced materials for complicated bone regeneration.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    氧化应激是骨质疏松症发展的重要因素。褪黑激素,松果体分泌的吲哚胺,具有抗氧化性能。本研究旨在探讨褪黑素是否可以促进骨形成,并阐明这一过程的潜在机制。在这项研究中,我们采用过氧化氢(H2O2)诱导的MC3T3-E1细胞氧化应激模型和大鼠去卵巢骨质疏松性骨缺损模型,探讨褪黑素对骨质疏松性骨缺损的保护作用及其机制.我们发现褪黑素显著增加碱性磷酸酶活性,矿化能力,H2O2处理的MC3T3-E1细胞中BMP2、RUNX2和OPN的表达。此外,发现褪黑激素激活SIRT1,SIRT3并抑制p66Shc,降低细胞内活性氧的水平,稳定线粒体,降低丙二醛水平,增加超氧化物歧化酶活性,并减少H2O2处理的MC3T3-E1细胞的凋亡。有趣的是,这些效应可被SIRT1抑制剂EX527逆转.体内实验证实,褪黑素改善了股骨远端骨小梁的微观结构和骨矿物质密度,并促进了骨形成。同时,褪黑素激活SIRT1,抑制p66Shc并增加SIRT3表达。一起来看,我们的发现表明,褪黑素可以通过激活SIRT1来调节SIRT3的活性并抑制p66Shc的表达,从而抑制MC3T3-E1细胞的氧化损伤并促进成骨。提示褪黑素可能是骨质疏松症相关骨代谢疾病的潜在治疗药物。
    Oxidative stress is an important contributor to the development of osteoporosis. Melatonin, an indoleamine secreted by the pineal gland, has antioxidant properties. This study aims to explore whether melatonin can promote bone formation and elucidate the mechanisms underlying this process. In this study, we used an in vitro hydrogen peroxide (H2O2)-induced oxidative stress model in MC3T3-E1 cells and an in vivo ovariectomized osteoporotic bone defect model in rats to explore the protective effects of melatonin against osteoporotic bone defects along with the mechanism underlying these effects. We found that melatonin significantly increased alkaline phosphatase activity, mineralization capacity, and the expression of BMP2, RUNX2, and OPN in MC3T3-E1 cells treated with H2O2. Furthermore, melatonin was found to activate SIRT1, SIRT3 and inhibit p66Shc, reduce the intracellular reactive oxygen species levels, stabilize mitochondria, reduce malondialdehyde levels, increase superoxide dismutase activity, and reduce apoptosis in MC3T3-E1 cells treated with H2O2. Intriguingly, these effects could be reversed by the SIRT1 inhibitor EX527. In vivo experiments confirmed that melatonin improves the microstructure and bone mineral density of the distal femoral bone trabecula and promotes bone formation. Meanwhile, melatonin activated SIRT1, inhibited p66Shc and increased SIRT3 expression. Taken together, our findings showed that melatonin can restrain oxidative damage in MC3T3-E1 cells and promote osteogenesis by activating SIRT1 which regulate the activity of SIRT3 and inhibit the expression of p66Shc, suggesting that melatonin could be a potential therapeutic agent for osteoporosis-related bone metabolic diseases.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

  • DOI:
    文章类型: Journal Article
    Local application of lithium or aspirin with biological scaffold has been identified as a potent means to improve bone formation. In this study, lithium and aspirin modified calcium phosphate cement (Asp-Li/CPC) was prepared, and the feasibility of this biological scaffold in the treatment of osteoporotic bone defect was observed in vivo and in vitro. In vitro experiments confirmed that Asp-Li/CPC had better ability to promote MC3T3-E1 cells differentiation into osteoblasts, osteoblast mineralization and viability, and promote cell expression of ALP, OP, RUNX-2, OC and COL-1 protein than simple CPC or lithium modified CPC by MTT, Alizarin red staining and Western blot evaluation. In vivo experiments confirmed that Asp-Li/CPC presented the strongest effect on bone regeneration and bone mineralization through the comparison with CPC group and Li/CPC group with X-ray images, Micro-CT and Histological evaluation. RT-qPCR analysis showed that Asp-Li/CPC, Li/CPC group and CPC group demonstrated increased BMP2, Smad1, OPG than the OVX group (P<0.05), while Asp-Li/CPC exhibited decreased TNF-α, IFN-γ and RANKL than the OVX group (P<0.05). Experiments in vivo and in vitro show that Asp-Li/CPC is a scheme for rapid repair of femoral condylar defects, and these effects may be achieved by inhibiting local inflammation and through BMP-2/Smad1 and OPG/RANKL signaling pathway.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: Journal Article
    背景:抗癫痫药物的使用和雌激素缺乏对骨缺损的再生提出了更高的要求。本研究研究了在卡马西平(CBZ)的影响下,阿仑膦酸钠(ALN)对去卵巢(OVX)大鼠股骨骨缺损的影响。
    方法:将100只3月龄的雌性SD大鼠进行假手术或OVX,分为四组:假对照(CON);OVX对照(OVX);通过管饲法用CBZ治疗的卵巢切除大鼠(75mg/kg/天;CBZ);用CBZ加ALN(2mg/kg/天)治疗的卵巢切除大鼠。在所有雌性SD大鼠中都建立了临界大小的股骨干phy端骨缺损。来自CBZ和CBZ-ALN组的动物在进行骨缺损手术后的第二天通过管饲法接受药物。老鼠被处死后,采集位于股骨远端的缺损区域进行显微计算机断层扫描(micro-CT)评估,苏木精和伊红(HE)染色,和马森的三色染色。还通过生物力学和免疫组织化学评估(IHC)分析样品。此外,生化分析评估所有血清样本。
    结果:本研究表明卵巢切除术改变了骨的微观结构参数。CBZ的使用进一步减少了股骨骨量,而用ALN治疗可预防骨丢失。与OVX和CBZ组相比,CBZ-ALN组促进骨新生,增强股骨极限负荷。然而,与CON组相比,CBZ-ALN组未恢复至正常水平.此外,我们注意到与CBZ组相比,CBZ-ALN组降低了抗酒石酸酸性磷酸酶-5b(Tracp-5b)的表达,对IHC中骨钙蛋白(OCN)和I型胶原(Col-I)的表达没有显着影响。生化分析结果表明,CBZ的全身递送对卵巢切除大鼠的骨形成和骨吸收具有有害作用,对I型胶原蛋白(CTX-1)的C端交联端肽的影响较差。此外,与CBZ组相比,CBZ-ALN组CTX-1水平显著下降.
    结论:这些结果表明,ALN可以有效地逆转CBZ对骨骼微观结构特性的影响,因此可以对骨质疏松症大鼠的局部骨肿瘤形成产生积极影响。
    结论:2mg/kgALN的剂量可以改善75mg/kgCBZ的负面影响,并促进股骨骨性缺损的骨新生。
    BACKGROUND: The use of antiepileptic drugs and estrogen deficiency put forward higher requirements for bone defect regeneration. The present study investigated the effects of alendronate (ALN) on femoral bone defect in ovariectomized (OVX) rats under the influence of carbamazepine (CBZ).
    METHODS: One hundred female SD rats at 3 months of age were either sham-operated or OVX and divided into four groups: sham control (CON); OVX control (OVX); ovariectomized rats treated with CBZ via gavage (75 mg/kg/day; CBZ); ovariectomized rats treated with CBZ plus ALN (2 mg/kg/day; CBZ-ALN). A critical-sized femoral metaphyseal bone defect was established in all female SD rats. Animals from the CBZ and CBZ-ALN groups received drugs by gavage the day after bone defect surgery was performed. After the rats were sacrificed, the defected area located in the distal femur was harvested for evaluation by microcomputed tomography (micro-CT), hematoxylin and eosin (HE) staining, and Masson\'s trichrome staining. The samples were also analyzed by biomechanics and immunohistochemical evaluation (IHC). Besides, biochemical analysis evaluates all serum samples.
    RESULTS: The present study showed that ovariectomy changed the microstructural parameters of bone. The use of CBZ further decreased femur bone mass while treatment with ALN prevented bone loss. Compared to OVX and CBZ groups, CBZ-ALN group promoted bone neoformation and enhanced the ultimate load of the femur bone. However, the group of CBZ-ALN did not return to normal levels compared with the CON group. Besides, we noticed that CBZ-ALN group reduced tartrate-resistant acid phosphatase-5b (Tracp-5b) expression and had no significant effect on the expression of osteocalcin (OCN) and type I collagen (Col-I) in IHC compared with CBZ group. Biochemical analysis results presented that systemic delivery of CBZ showed pernicious effects on bone formation and resorption in ovariectomized rats, with the worse effects on C-terminal crosslinked telopeptide of type I collagen (CTX-1). Besides, a significant decrease in CTX-1 levels was observed in CBZ-ALN group as compared to the group of CBZ.
    CONCLUSIONS: These results demonstrated that ALN can effectively reverse the effects of CBZ on the microarchitectural properties of bone, and thus can have a positive effect on local bone neoformation in rats with osteoporosis.
    CONCLUSIONS: The dose of 2 mg/kg ALN improves the negative effect of prescription of CBZ at 75 mg/kg and promotes bone neoformation of femoral bony deficits.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号