mesoporous silica nanoparticle

  • 文章类型: Journal Article
    颞下颌关节骨关节炎(TMJOA)是口腔颌面外科常见的退行性关节疾病。最近的研究表明,Wnt/β-catenin信号的过度不平衡激活与TMJOA的发病机制有关,并且由于无法抑制过度激活的Wnt通路,而Wnt16缺陷小鼠的膝关节OA更严重。然而,TMJ内直接注射Wnt16缓解TMJOA的疗效仍未得到直接证实.此外,Wnt16等小分子药物通常表现出短期疗效和较差的治疗依从性.因此,为了在短期和长期内获得WNT16的稳定释放,这项研究制造了一种基于介孔二氧化硅纳米球(MSN)包裹在透明质酸(HA)水凝胶中的双层缓释Wnt16载体。在体外和体内分析生物功能水凝胶HA/Wnt16@MSN以评估TMJOA的治疗。因此,它显示出优越的前软骨基质恢复和破骨细胞生成抑制能力,并有效抑制Wnt/β-catenin通路的过度激活。一起来看,生物功能水凝胶HA/Wnt16@MSN是治疗TMJOA的有希望的候选药物。
    Temporomandibular joint osteoarthritis (TMJOA) is a commonly encountered degenerative joint disease in oral and maxillofacial surgery. Recent studies have shown that the excessive unbalanced activation of Wnt/β-catenin signaling is connected with the pathogenesis of TMJOA and due to the inability to inhibit the over-activated Wnt pathway, while Wnt16-deficient mice has a more severe Knee OA. However, the efficacy of direct intra-TMJ injection of Wnt16 for the relief of TMJOA is still not directly confirmed. Moreover, small-molecule drugs such as Wnt16 usually exhibit short-lived efficacy and poor treatment adherence. Therefore, in order to obtain a stable release of Wnt16 both in the short and long term, this study fabricates a double-layer slow-release Wnt16 carrier based on mesoporous silica nanospheres (MSNs) encased within hyaluronic acid (HA) hydrogels. The biofunctional hydrogel HA/Wnt16@MSN is analyzed both in vitro and in vivo to evaluate the treatment of TMJOA. As a result, it shows superior pro-cartilage matrix restoration and inhibition of osteoclastogenesis ability, and effectively inhibits the over-activation of the Wnt/β-catenin pathway. Taken together, biofunctional hydrogel HA/Wnt16@MSN is a promising candidate for the treatment of TMJOA.
    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
    晚期乳腺癌仍然是一个重大的肿瘤挑战,需要新的方法来改善临床结果。本研究使用MCM-41-NH2-DTPA-Gd3+-MIH纳米材料研究了一种创新的治疗药物,其结合用于检测的MRI成像和用于治疗的新型化疗药物(MIH2.4Bl)。纳米材料基于介孔二氧化硅类型,MCM-41,并通过胺基官能化和与DTPA缀合以及与Gd3+络合进行药物递送优化。MRI敏感性通过使用基于钆的造影剂增强,这对识别早期肿瘤性病变至关重要。MIH2.4Bl,以其独特的介离子结构,允许与促进其细胞内抗肿瘤活性的生物分子的有效相互作用。物理化学表征证实了纳米材料的合成和有效的药物掺入,其中15%的MIH2.4Bl被吸附。药物释放试验表明,大约50%在8小时内释放。MRI体模研究表明,纳米材料具有优越的成像能力,弛豫率明显高于商业剂Magnvist。体外细胞毒性测定,在EC50浓度为12.6mg/mL时,与之相比,在正常人乳腺上皮细胞(HMEC)中EC50浓度为68.9mg/mL时,证明了该纳米材料在杀伤MDA-MB-231乳腺癌细胞方面的有效性.在体内,4T1同基因小鼠模型中的MRI评估证实了其作为造影剂的功效。这项研究强调了MCM-41-NH2-DTPA-Gd3+-MIH的治疗能力及其增强乳腺癌管理的潜力。
    Advanced breast cancer remains a significant oncological challenge, requiring new approaches to improve clinical outcomes. This study investigated an innovative theranostic agent using the MCM-41-NH2-DTPA-Gd3⁺-MIH nanomaterial, which combined MRI imaging for detection and a novel chemotherapy agent (MIH 2.4Bl) for treatment. The nanomaterial was based on the mesoporous silica type, MCM-41, and was optimized for drug delivery via functionalization with amine groups and conjugation with DTPA and complexation with Gd3+. MRI sensitivity was enhanced by using gadolinium-based contrast agents, which are crucial in identifying early neoplastic lesions. MIH 2.4Bl, with its unique mesoionic structure, allows effective interactions with biomolecules that facilitate its intracellular antitumoral activity. Physicochemical characterization confirmed the nanomaterial synthesis and effective drug incorporation, with 15% of MIH 2.4Bl being adsorbed. Drug release assays indicated that approximately 50% was released within 8 h. MRI phantom studies demonstrated the superior imaging capability of the nanomaterial, with a relaxivity significantly higher than that of the commercial agent Magnevist. In vitro cellular cytotoxicity assays, the effectiveness of the nanomaterial in killing MDA-MB-231 breast cancer cells was demonstrated at an EC50 concentration of 12.6 mg/mL compared to an EC50 concentration of 68.9 mg/mL in normal human mammary epithelial cells (HMECs). In vivo, MRI evaluation in a 4T1 syngeneic mouse model confirmed its efficacy as a contrast agent. This study highlighted the theranostic capabilities of MCM-41-NH2-DTPA-Gd3⁺-MIH and its potential to enhance breast cancer management.
    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
    影响大脑的疾病是全球第二大死亡原因。针对脑疾病的药物的主要挑战之一是通过血脑屏障(BBB)。这里,评估了具有两种不同尖峰长度的介孔二氧化硅纳米星(MSiNS)穿过体外BBB多细胞模型的有效性,并与球形纳米颗粒(MSiNP)进行了比较。改进的溶胶-凝胶单胶束外延生长用于生产MSiNS,在外周血单核细胞和神经元细胞中浓度高达1μgmL-1时,没有细胞毒性或免疫原性。纳米星MSiNS在24小时后有效地穿透了BBB模型,和具有较短尖峰长度(9±2nm)的MSiNS-1在96h时比具有较长尖峰(18±4nm)或球形MSiNP的MSiNS-2更快地穿过体外BBB模型,积聚在顶端和基底外侧,分别。分子动力学模拟表明,在与MSiNS接触期间,脂质双层的构型灵活性增加。导致包裹,而MSiNP抑制膜波动。这项工作提出了一种基于病毒状MSiNS的有效脑给药系统,用于治疗不同的脑部疾病,以及它们与脂质双层相互作用的机制。
    Conditions affecting the brain are the second leading cause of death globally. One of the main challenges for drugs targeting brain diseases is passing the blood-brain barrier (BBB). Here, the effectiveness of mesoporous silica nanostars (MSiNSs) with two different spike lengths to cross an in vitro BBB multicellular model was evaluated and compared to spherical nanoparticles (MSiNP). A modified sol-gel single-micelle epitaxial growth was used to produce MSiNS, which showed no cytotoxicity or immunogenicity at concentrations of up to 1 μg mL-1 in peripheral blood mononuclear and neuronal cells. The nanostar MSiNS effectively penetrated the BBB model after 24 h, and MSiNS-1 with a shorter spike length (9 ± 2 nm) crossed the in vitro BBB model more rapidly than the MSiNS-2 with longer spikes (18 ± 4 nm) or spherical MSiNP at 96 h, which accumulated in the apical and basolateral sides, respectively. Molecular dynamic simulations illustrated an increase in configurational flexibility of the lipid bilayer during contact with the MSiNS, resulting in wrapping, whereas the MSiNP suppressed membrane fluctuations. This work advances an effective brain drug delivery system based on virus-like shaped MSiNS for the treatment of different brain diseases and a mechanism for their interaction with lipid bilayers.
    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
    介孔二氧化硅纳米粒子(MSNs)是物理和化学上稳定的无机纳米材料,在纳米医学领域作为药物递送系统的载体备受关注。在本研究中,我们研究了掺入抗原肽的MSN疫苗用于癌症免疫治疗的潜力.体外实验表明,荧光标记的MSN在小鼠树突状细胞(DC2.4细胞)中积累,颗粒定位于胞质溶胶。这些观察结果可能表明,MSN具有将负载的分子递送到抗原呈递细胞中的潜力,从而刺激宿主的获得性免疫系统。体内实验证明,在植入表达卵清蛋白(OVA)的淋巴瘤细胞的小鼠中,存活时间延长(E。G7-OVA细胞)皮下接种掺入OVA抗原肽的MSN。此外,血清和脾细胞中检测到OVA特异性免疫球蛋白G抗体和细胞毒性T淋巴细胞,分别,接种MSN-OVA疫苗的小鼠,表明在体液和细胞免疫系统中诱导抗原特异性反应。这些结果表明,掺入抗原肽的MSN疗法可以作为癌症免疫疗法的新型疫苗。
    Mesoporous silica nanoparticles (MSNs) are physically and chemically stable inorganic nanomaterials that have been attracting much attention as carriers for drug delivery systems in the field of nanomedicine. In the present study, we investigated the potential of MSN vaccines that incorporate antigen peptides for use in cancer immunotherapy. In vitro experiments demonstrated that fluorescently labeled MSNs accumulated in a line of mouse dendritic cells (DC2.4 cells), where the particles localized to the cytosol. These observations could suggest that MSNs have potential for use in delivering the loaded molecules into antigen-presenting cells, thereby stimulating the host acquired immune system. In vivo experiments demonstrated prolonged survival in mice implanted with ovalbumin (OVA)-expressing lymphoma cells (E.G7-OVA cells) following subcutaneous inoculation with MSNs incorporating OVA antigen peptides. Furthermore, OVA-specific immunoglobulin G antibodies and cytotoxic T lymphocytes were detected in the serum and the spleen cells, respectively, of mice inoculated with an MSN-OVA vaccine, indicating the induction of antigen-specific responses in both the humoral and cellular immune systems. These results suggested that the MSN therapies incorporating antigen peptides may serve as novel vaccines for cancer immunotherapy.
    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
    背景:介孔二氧化硅纳米颗粒(MSN)被广泛用作递送化疗药物的理想纳米载体。然而,在大多数相关研究中,高抗牙周炎活性和低生物毒性之间的平衡一直难以维持,因为二氧化硅在活生物体中的降解缓慢.
    方法:在本研究中,响应性羟基磷灰石(HAP)被掺杂到MSN骨架中,化疗药物盐酸米诺环素(MH)被加载到MSN的孔中,形成带负电荷的药物输送系统。阳离子壳聚糖(COS)是一种抗菌性能高、生物安全性好的生物可降解材料。在这项研究中,通过稳定的电荷相互作用将COS固定在载药颗粒表面,构建了复合给药系统(MH@MSNion@COS)。
    结果:体外和细胞实验证明了纳米载体系统的有效降解和药物的同步受控释放。值得注意的是,与单次MH管理相比,这个系统,其中MH和COS共同调节牙周炎相关炎症因子(TNF-α,IL-6,IL-1β,和iNOS),能较好地抑制牙周炎的进展,诱导组织再生,对细胞无明显毒副作用。
    结论:该系统为智能,高效,和安全的抗牙周炎药物递送系统。
    BACKGROUND: Mesoporous silica nanoparticles (MSN) are widely used as ideal nanovehicles for the delivery of chemotherapeutic drugs. However, the balance between high anti-periodontitis activity and low biotoxicity has been challenging to maintain in most relevant studies owing to the slow degradation of silica in living organisms.
    METHODS: In this study, -responsive hydroxyapatite (HAP) was doped into the MSN skeleton, and the chemotherapeutic drug minocycline hydrochloride (MH) was loaded into the pores of MSN, forming a negatively charged drug delivery system. Cationic chitosan (COS) is a biodegradable material with high antibacterial performance and good biosafety. In this study, COS was immobilized on the surface of the drug-loaded particles through stable charge interaction to construct a composite drug delivery system (MH@MSNion@COS).
    RESULTS: In vitro and cellular experiments demonstrated effective degradation of the nanocarrier system and synchronized controlled release of the drug. Notably, compared with single MH administration, this system, in which MH and COS jointly regulated the expression levels of periodontitis- associated inflammatory factors (TNF-α, IL-6, IL-1β, and iNOS), better inhibited the progress of periodontitis and induced tissue regeneration without showing significant toxic side effects in cells.
    CONCLUSIONS: This system provides a promising strategy for the design of intelligent, efficient, and safe anti-periodontitis drug delivery systems.
    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
    介孔二氧化硅纳米颗粒(MSN)是高度先进的工程颗粒,具有增加的表面积和对各种分子的极端吸附能力。在这里,合成了两种类型的MSN,并将其用作磷化氢气体的吸附剂。一个是没有官能团(MSN),另一种是用硼酸(MSN-BA)后改性。MSN和硼酸改性MSN的结构,具有约1025和650m2/g的高表面积,分别,已定义。通过透射电子显微镜(TEM)发现MSN具有尺寸为约30nm的颗粒。在本研究中,MSN被用作磷中毒的解毒剂,磷化锌(磷)粉末被用作毒性和致死剂。对大鼠进行体内分析以证明MSN化学吸附磷化氢气体的能力。在生存率评估中,在用MSN治疗后,phos中毒的动物保持存活,MSN-BA治疗组(剂量为5mg/kg)的存活率为60%。血清分析表明,MSNs具有很高的缓解器官血液损伤的潜力,用MSN-BA治疗磷化氢中毒的动物时,血清生物标志物急剧下降。
    Mesoporous silica nanoparticles (MSNs) are highly advanced engineered particles with increased surface area and extreme adsorption capacity for various molecules. Herein, two types of MSNs were synthesized and applied as adsorbents for phosphine gas. One was without functional groups (MSN), and the other was post-modified with boric acid (MSN-BA). The structures of MSN and boric acid-modified MSN with high surface areas of about 1025 and 650 m2/g, respectively, were defined. MSN was found to have particles with sizes around 30 nm by transmission electron microscopy (TEM). In the present study, MSNs were used as an antidote to phosphorus poisoning, and zinc phosphide (phosphorus) powder was used as the toxic and lethal agent. In vivo analysis was carried out on rats to demonstrate the ability of MSNs to chemisorb phosphine gas. In the survival percentage assessment, Phos-poisoned animals were kept alive after treatment with MSNs, and the MSN-BA-treated group (dose of 5 mg/kg) was shown to have a 60 % survival rate. Blood serum analysis showed that MSNs have a high potential to alleviate organ blood damage, and serum biomarkers dropped sharply while phosphine-poisoned animals were treated with MSN-BA.
    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
    与激活铁凋亡相关的癌症治疗的挑战主要来自与氧化还原和铁稳态相关的障碍。这阻碍了肿瘤细胞对铁性凋亡的敏感性。然而,铁死亡抗性的具体机制,尤其是那些与肿瘤细胞内异常代谢过程交织在一起的细胞,一直被低估。作为回应,我们提出了一种创新的谷胱甘肽响应磁热治疗纳米药物,称为LFMP。LFMP由装载到具有Fe3O4核的PEG修饰的磁性纳米颗粒中的lonidamine(LND)组成,并涂覆有二硫键桥接的介孔二氧化硅壳。这种纳米药物被设计为通过破坏稳态在肿瘤细胞中诱导加速的铁凋亡激活状态。在交变磁场和肿瘤微环境中高浓度谷胱甘肽的双重作用下,LFMP经历了崩解,释放药物。LND通过抑制糖酵解干预细胞代谢,最终增加铁死亡并导致合成谷胱甘肽消耗。二硫键通过消耗谷胱甘肽和灭活谷胱甘肽过氧化物酶4(GPX4)在破坏细胞内氧化还原稳态中起关键作用,与LND协同增强肿瘤细胞对铁凋亡的敏感性。这个过程加剧了氧化应激,进一步损害氧化还原稳态。此外,LFMP加剧线粒体功能障碍,在癌细胞中触发ROS形成和乳酸积累,导致酸度增加和随后的肿瘤细胞死亡。重要的是,LFMP显著抑制肿瘤细胞增殖,在体外和体内副作用最小,表现出令人满意的T2加权MR成像特性。总之,这种基于磁性热疗的纳米医学策略为抗肿瘤治疗提供了一种有前途的创新方法.
    The challenges associated with activating ferroptosis for cancer therapy primarily arise from obstacles related to redox and iron homeostasis, which hinder the susceptibility of tumor cells to ferroptosis. However, the specific mechanisms of ferroptosis resistance, especially those intertwined with abnormal metabolic processes within tumor cells, have been consistently underestimated. In response, we present an innovative glutathione-responsive magnetocaloric therapy nanodrug termed LFMP. LFMP consists of lonidamine (LND) loaded into PEG-modified magnetic nanoparticles with a Fe3O4 core and coated with disulfide bonds-bridged mesoporous silica shells. This nanodrug is designed to induce an accelerated ferroptosis-activating state in tumor cells by disrupting homeostasis. Under the dual effects of alternating magnetic fields and high concentrations of glutathione in the tumor microenvironment, LFMP undergoes disintegration, releasing drugs. LND intervenes in cell metabolism by inhibiting glycolysis, ultimately enhancing iron death and leading to synthetic glutathione consumption. The disulfide bonds play a pivotal role in disrupting intracellular redox homeostasis by depleting glutathione and inactivating glutathione peroxidase 4 (GPX4), synergizing with LND to enhance the sensitivity of tumor cells to ferroptosis. This process intensifies oxidative stress, further impairing redox homeostasis. Furthermore, LFMP exacerbates mitochondrial dysfunction, triggering ROS formation and lactate buildup in cancer cells, resulting in increased acidity and subsequent tumor cell death. Importantly, LFMP significantly suppresses tumor cell proliferation with minimal side effects both in vitro and in vivo, exhibiting satisfactory T2-weighted MR imaging properties. In conclusion, this magnetic hyperthermia-based nanomedicine strategy presents a promising and innovative approach for antitumor therapy.
    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
    他克莫司(FK506)是临床上治疗移植排斥的有效药物,主要通过抑制淋巴结(LN)中同种异体T细胞的活化和增殖来抑制排斥反应。然而,传统的给药方法在直接向LN递送游离FK506方面面临挑战。在这项研究中,我们介绍了一种基于介孔二氧化硅纳米颗粒的新型LN靶向递送系统(MSNs-FK506-MECA79)。这些颗粒被设计为选择性地靶向LN中的高内皮小静脉;这是通过用MECA79抗体进行表面修饰来实现的。它们的平均大小和ζ电位为201.18±5.98nm和-16.12±0.36mV,分别。我们的发现表明,与游离FK506治疗组相比,MSNs-FK506-MECA79可以在LNs中积累,并将FK506的局部浓度从28.02±7.71ng/g增加到123.81±76.76ng/g。随后,在皮肤移植模型中评估了MSNs-FK506-MECA79的疗效.用MSNs-FK506-MECA79处理可导致移植物中T细胞浸润减少,拒绝等级的降低,和显著延长生存期。因此,本研究为主动LN靶向递送FK506和改善移植排斥的免疫治疗效果提供了一种有前景的策略.
    Tacrolimus (FK506) is an effective therapeutic for transplant rejection in clinical practice, primarily inhibiting rejection by suppressing the activation and proliferation of allogeneic T cells in the lymph nodes (LNs). However, conventional administration methods face challenges in directly delivering free FK506 to the LNs. In this study, we introduce a novel LN-targeted delivery system based on mesoporous silica nanoparticles (MSNs-FK506-MECA79). These particles were designed to selectively target high endothelial venules in LNs; this was achieved through surface modification with MECA79 antibodies. Their mean size and zeta potential were 201.18 ± 5.98 nm and - 16.12 ± 0.36 mV, respectively. Our findings showed that MSNs-FK506-MECA79 could accumulate in LNs and increase the local concentration of FK506 from 28.02 ± 7.71 ng/g to 123.81 ± 76.76 ng/g compared with the free FK506 treatment group. Subsequently, the therapeutic efficacy of MSNs-FK506-MECA79 was evaluated in a skin transplantation model. The treatment with MSNs-FK506-MECA79 could lead to a decrease in the infiltration of T cells in the grafts, a reduction in the grade of rejection, and a significant prolongation of survival. Consequently, this study presents a promising strategy for the active LN-targeted delivery of FK506 and improving the immunotherapeutic effects on transplant rejection.
    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
    控制药物释放并限制其在健康器官中的存在是非常有价值的。在这项研究中,介孔二氧化硅纳米粒子(MSN)为核心,负载紫杉醇(PTX),用二硫键官能化的无孔二氧化硅壳涂覆。通过二硫键用聚乙二醇(PEG)进一步涂覆纳米颗粒。我们分析了纳米粒子的物理化学性质,包括通过动态光散射(DLS)的流体动力学尺寸,zeta电位,X射线衍射(XRD)傅里叶变换红外(FTIR)光谱,并通过透射电子显微镜(TEM)和扫描电子显微镜(SEM)成像。测量了2μM和10μM两种不同谷胱甘肽(GSH)浓度下的药物释放曲线。使用共聚焦激光扫描显微镜(CLSM)图像和流式细胞术确定MCF-7细胞系对纳米颗粒的细胞摄取。此外,用MTT法和流式细胞术研究了MCF-7细胞的细胞活力和纳米颗粒诱导细胞凋亡的能力,分别。我们的研究表明,PTX从药物递送系统中的释放是氧化还原反应性的。此外,结果表明,细胞摄取水平升高,细胞凋亡有效诱导,强调了这种氧化还原反应性药物递送系统在乳腺癌治疗中的潜力。
    Controlling the drug release and restricting its presence in healthy organs is extremely valuable. In this study, mesoporous silica nanoparticles (MSN) as the core, loaded with paclitaxel (PTX), were coated with a non-porous silica shell functionalized with disulfide bonds. The nanoparticles were further coated with polyethylene glycol (PEG) via disulfide linkages. We analyzed the physicochemical properties of nanoparticles, including hydrodynamic size via Dynamic Light Scattering (DLS), zeta potential, X-ray Diffraction (XRD) patterns, Fourier-Transform Infrared (FTIR) spectra, and imaging through Transmission Electron Microscopy (TEM) and Scanning Electron Microscopy (SEM). The drug release profile in two distinct glutathione (GSH) concentrations of 2 µM and 10 µM was measured. The cellular uptake of nanoparticles by MCF-7 cell line was determined using Confocal Laser Scanning Microscopy (CLSM) images and flow cytometry. Furthermore, the cell viability and the capability of nanoparticles to induce apoptosis in MCF-7 cell line were studied using the MTT assay and flow cytometry, respectively. Our investigations revealed that the release of PTX from the drug delivery system was redox-responsive. Also, results indicated an elevated level of cellular uptake and efficient induction of apoptosis, underscoring the promising potential of this redox-responsive drug delivery system for breast cancer therapy.
    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
    细菌感染和骨整合延迟是钛基骨科植入物的两个主要挑战。在本研究中,我们开发了一种功能化的钛植入物Ti-M@A通过固定抗菌肽(AMP)HHC36负载二硒化物桥接介孔二氧化硅纳米颗粒(MSN)在表面,表现出良好的长期和机械稳定性。功能化的植入物可以在30天内实现AMP的持续释放,并且对四种类型的临床细菌表现出超过95.71%的抗菌活性(S.金黄色葡萄球菌,大肠杆菌,铜绿假单胞菌和MRSA),这源于破坏细菌膜的能力。此外,Ti-M@A能有效抑制细菌生物膜的形成。由于MSN中的Se,功能化的植入物还可以显着促进小鼠骨髓来源的间充质干细胞(mBMSC)的成骨分化。值得注意的是,它可以通过清除LPS激活的巨噬细胞中的ROS来触发巨噬细胞向M2极化。因此,感染和非感染骨缺损模型的体内试验表明,这种生物活性植入物不仅可以杀死98.82%以上的金黄色葡萄球菌,还能促进骨融合。因此,本研究为解决钛植入物的细菌感染和骨整合延迟提供了一个联合策略。
    Bacterial infection and delayed osseointegration are two major challenges for titanium-based orthopedic implants. In the present study, we developed a functionalized titanium implant Ti-M@A by immobilizing antimicrobial peptide (AMP) HHC36-loaded diselenide-bridged mesoporous silica nanoparticles (MSNs) on the surface, which showed good long-term and mechanical stability. The functionalized implants can realize the sustained release of AMP over 30 days and exhibit over 95.71 % antimicrobial activity against four types of clinical bacteria (S. aureus, E. coli, P. aeruginosa and MRSA), which arose from the capability to destroy the bacterial membranes. Moreover, Ti-M@A can efficiently inhibit the biofilm formation of the bacteria. The functionalized implants can also significantly promote the osteogenic differentiation of mouse bone marrow-derived mesenchymal stem cells (mBMSCs) because of the Se in MSNs. Notably, it can trigger macrophages toward M2 polarization in vitro by scavenging ROS in LPS-activated macrophages. Consequently, in vivo assays with infection and non-infection bone defect models demonstrated that such bioactive implants can not only kill over 98.82 % of S. aureus, but also promote osseointegration. Hence, this study provides a combined strategy to resolve bacterial infection and delayed osseointegration for titanium implants.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

    求助全文

公众号