Integrin alphaVbeta3

整合素 α V β 3
  • 文章类型: Journal Article
    小干扰RNA(siRNA)突出了癌症治疗的巨大治疗潜力。siRNA治疗的主要挑战是有效的RNA纳米药物递送系统,这是由配体和载体促进的。在这项研究中,我们分析了线性RGD和环状RGD对αVβ3整合素的结合特异性,通过使用超分辨率直接随机光学重建显微镜绘制形态学图。同时,使用单分子力谱研究了结合动力学。然后,通过力追踪技术在单颗粒水平上评估配体和载体对RNA纳米药物细胞进入动力学参数的影响。此外,使用基于AFM的纳米压痕在单细胞水平评估RNA纳米药物的递送效率.本报告将为旨在提高纳米药物递送系统效率的合理设计策略提供有价值的见解。
    Small interfering RNA (siRNA) highlights the immense therapeutic potential for cancer treatment. The major challenge in siRNA therapy is the effective RNA nanodrug delivery system, which is facilitated by the ligand and the carrier. In this study, we analyzed the binding specificity of linear RGD and circular RGD to αVβ3 integrins by mapping the morphology using super-resolution direct stochastic optical reconstruction microscopy. Meanwhile, the binding dynamics was investigated using single-molecule force spectroscopy. Then, the effects of the ligand and carrier on RNA nanodrug cell entry dynamic parameters were evaluated at the single particle level by the force tracing technique. Furthermore, the delivery efficiency of RNA nanodrugs was assessed using AFM-based nanoindentation at the single cell level. This report will provide valuable insights for rational design strategies aiming to achieve improved efficiency for nanodrug 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
    在斑马鱼中,脑淋巴管内皮细胞(BLECs)是脑膜血管生成和脑血管再生所必需的。尽管表皮生长因子样结构域7(Egfl7)已被报道作为促血管生成因子,其在淋巴管生成中的作用尚不清楚.这里,我们显示Egfl7在血液和淋巴内皮细胞中均有表达。我们产生了在外显子3中具有13bp缺失的egfl7cq180突变体,导致Egfl7的表达降低。egfl7cq180突变体斑马鱼表现出BLEC双侧环状结构的缺陷形成,虽然躯干和面部淋巴发育未受影响。此外,而egfl7cq180突变体显示正常的BLEC谱系规格,这些细胞的迁移和增殖受损。此外,我们确定整合素αvβ3为Egfl7的受体。αvβ3在CVP和发芽BLECs中表达,阻断该整合素抑制BLEC双侧环状结构的形成。因此,这项研究确定了Egfl7在通过整合素αvβ3介导的BLEC发育中的作用。
    In zebrafish, brain lymphatic endothelial cells (BLECs) are essential for meningeal angiogenesis and cerebrovascular regeneration. Although epidermal growth factor-like domain 7 (Egfl7) has been reported to act as a pro-angiogenic factor, its roles in lymphangiogenesis remain unclear. Here, we show that Egfl7 is expressed in both blood and lymphatic endothelial cells. We generate an egfl7 cq180 mutant with a 13-bp-deletion in exon 3 leading to reduced expression of Egfl7. The egfl7 cq180 mutant zebrafish exhibit defective formation of BLEC bilateral loop-like structures, although trunk and facial lymphatic development remains unaffected. Moreover, while the egfl7 cq180 mutant displays normal BLEC lineage specification, the migration and proliferation of these cells are impaired. Additionally, we identify integrin αvβ3 as the receptor for Egfl7. αvβ3 is expressed in the CVP and sprouting BLECs, and blocking this integrin inhibits the formation of BLEC bilateral loop-like structures. Thus, this study identifies a role for Egfl7 in BLEC development that is mediated through the integrin αvβ3.
    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
    腹主动脉瘤(AAA)是一种高度致命的心血管疾病,鉴于该疾病的复杂病理生理学,目前缺乏有效的药物治疗。这里,我们分析了AAA患者和小鼠模型的单细胞RNA测序数据,揭示了关键的病理变化,包括巨噬细胞的M1样极化和平滑肌细胞(SMC)收缩功能的丧失。两种细胞类型都表达整联蛋白αvβ3,从而允许用单个合理设计的分子进行双重靶向。为此,我们设计了一种生物相容性纳米药物,我们称之为EVMS@R-HNC,由封装有乙型肝炎病毒核心蛋白的多功能药物依维莫司(EVMS)组成,该蛋白修饰为包含RGD序列以特异性结合整联蛋白αvβ3。体外和体内结果显示EVMS@R-HNC可以靶向巨噬细胞和SMC。纳米药物结合后,EVMS在细胞内释放,在那里它表现出多种功能,包括抑制M1巨噬细胞极化,从而抑制自我传播的炎症级联反应和免疫微环境失衡,同时保持SMC的正常收缩功能。总的来说,这些结果表明,EVMS@R-HNC为AAA的治疗提供了一种非常有前景的治疗方法.本文受版权保护。保留所有权利。
    Abdominal aortic aneurysm (AAA) is a highly lethal cardiovascular disease that currently lacks effective pharmacological treatment given the complex pathophysiology of the disease. Here, single-cell RNA-sequencing data from patients with AAA and a mouse model are analyzed, which reveals pivotal pathological changes, including the M1-like polarization of macrophages and the loss of contractile function in smooth muscle cells (SMCs). Both cell types express the integrin αvβ3, allowing for their dual targeting with a single rationally designed molecule. To this end, a biocompatible nanodrug, which is termed EVMS@R-HNC, that consists of the multifunctional drug everolimus (EVMS) encapsulated by the hepatitis B virus core protein modifies to contain the RGD sequence to specifically bind to integrin αvβ3 is designed. Both in vitro and in vivo results show that EVMS@R-HNC can target macrophages as well as SMCs. Upon binding of the nanodrug, the EVMS is released intracellularly where it exhibits multiple functions, including inhibiting M1 macrophage polarization, thereby suppressing the self-propagating inflammatory cascade and immune microenvironment imbalance, while preserving the normal contractile function of SMCs. Collectively, these results suggest that EVMS@R-HNC presents a highly promising therapeutic approach for the management of AAA.
    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
    内皮高通透性是脓毒症相关多器官功能障碍的关键。增加血管性血友病因子(vWF)血浆水平,源于脓毒症期间活化的血小板和内皮损伤,可与整合素αvβ3结合,加剧内皮通透性。因此,靶向该途径为脓毒症提供了潜在的治疗途径.最近,我们鉴定了isaridinE(ISE),一种海洋真菌环己肽,作为一种有前途的抗血小板和抗血栓药物,出血风险低。ISE对脓毒症小鼠模型脓毒症死亡率和脓毒症肺损伤的影响,盲肠结扎和穿刺诱导,在这项研究中进行了调查。ISE剂量依赖性地提高生存率,减轻肺损伤,血小板减少症,肺内皮通透性,和小鼠模型中的血管炎症。ISE通过抑制囊泡相关膜蛋白8和可溶性N-乙基马来酰亚胺敏感因子附着蛋白23的过度表达,显着减少了脓毒症小鼠活化血小板中vWF的释放。此外,ISE抑制健康人血小板对培养的脂多糖(LPS)刺激的人脐静脉内皮细胞(HUVECs)的粘附,从而显著降低vWF分泌和内皮高通透性。使用cilengitide,选择性整合素αvβ3抑制剂,发现ISE可以通过抑制vWF与αvβ3的结合来改善内皮通透性。整合素αvβ3-FAK/Src途径的激活可能是vWF诱导的脓毒症内皮功能障碍的基础。总之,ISE通过抑制内皮高通透性和血小板-内皮相互作用来预防脓毒症。
    Endothelial hyperpermeability is pivotal in sepsis-associated multi-organ dysfunction. Increased von Willebrand factor (vWF) plasma levels, stemming from activated platelets and endothelium injury during sepsis, can bind to integrin αvβ3, exacerbating endothelial permeability. Hence, targeting this pathway presents a potential therapeutic avenue for sepsis. Recently, we identified isaridin E (ISE), a marine-derived fungal cyclohexadepsipeptide, as a promising antiplatelet and antithrombotic agent with a low bleeding risk. ISE\'s influence on septic mortality and sepsis-induced lung injury in a mouse model of sepsis, induced by caecal ligation and puncture, is investigated in this study. ISE dose-dependently improved survival rates, mitigating lung injury, thrombocytopenia, pulmonary endothelial permeability, and vascular inflammation in the mouse model. ISE markedly curtailed vWF release from activated platelets in septic mice by suppressing vesicle-associated membrane protein 8 and soluble N-ethylmaleide-sensitive factor attachment protein 23 overexpression. Moreover, ISE inhibited healthy human platelet adhesion to cultured lipopolysaccharide (LPS)-stimulated human umbilical vein endothelial cells (HUVECs), thereby significantly decreasing vWF secretion and endothelial hyperpermeability. Using cilengitide, a selective integrin αvβ3 inhibitor, it was found that ISE can improve endothelial hyperpermeability by inhibiting vWF binding to αvβ3. Activation of the integrin αvβ3-FAK/Src pathway likely underlies vWF-induced endothelial dysfunction in sepsis. In conclusion, ISE protects against sepsis by inhibiting endothelial hyperpermeability and platelet-endothelium interactions.
    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
    细胞外囊泡(EV),以低免疫原性为特征,高生物相容性和靶向特异性以及优异的血脑屏障通透性,越来越被认为是治疗各种疾病的有前途的药物输送工具,比如癌症,炎症和病毒感染。然而,最近的发现表明,由于宿主单核吞噬细胞系统通过Fcγ受体介导的吞噬清除,EV的细胞内递送效率达不到预期,补体受体以及非调理吞噬受体。在本文中,我们研究了一系列拮抗宿主吞噬机制的细菌毒力蛋白,旨在探索它们在工程电动汽车中抵消吞噬作用的潜力。特别强调由A组链球菌分泌的IdeS和铜绿假单胞菌分泌的ImpA。因为它们不仅抵消吞噬作用,而且还与癌细胞上高度上调的表面生物标志物αVβ3结合,或分别切割肿瘤生长和转移促进因子CD44。这表明细菌抗吞噬蛋白,在使用预加载或后加载策略装饰到电动汽车上之后,不仅可以通过逃避宿主吞噬作用来提高基于EV的药物递送效率,从而获得更好的治疗效果,而且还可以通过整合吞噬作用拮抗作用和癌症靶向或失活来进一步实现创新的基于EV的协同癌症治疗方法。
    Extracellular vesicles (EVs), characterized by low immunogenicity, high biocompatibility and targeting specificity along with excellent blood-brain barrier permeability, are increasingly recognized as promising drug delivery vehicles for treating a variety of diseases, such as cancer, inflammation and viral infection. However, recent findings demonstrate that the intracellular delivery efficiency of EVs fall short of expectations due to phagocytic clearance mediated by the host mononuclear phagocyte system through Fcγ receptors, complement receptors as well as non-opsonic phagocytic receptors. In this text, we investigate a range of bacterial virulence proteins that antagonize host phagocytic machinery, aiming to explore their potential in engineering EVs to counteract phagocytosis. Special emphasis is placed on IdeS secreted by Group A Streptococcus and ImpA secreted by Pseudomonas aeruginosa, as they not only counteract phagocytosis but also bind to highly upregulated surface biomarkers αVβ3 on cancer cells or cleave the tumor growth and metastasis-promoting factor CD44, respectively. This suggests that bacterial anti-phagocytic proteins, after decorated onto EVs using pre-loading or post-loading strategies, can not only improve EV-based drug delivery efficiency by evading host phagocytosis and thus achieve better therapeutic outcomes but also further enable an innovative synergistic EV-based cancer therapy approach by integrating both phagocytosis antagonism and cancer targeting or deactivation.
    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
    纳米级细胞-细胞外基质(ECM)配体的物理性质深刻影响生物过程,如附着力,运动性,和差异化。虽然细胞对静态配体的机械反应已得到充分研究,具有“适应性”特性的动态配体呈递对细胞机械转导的影响尚不清楚。利用可控的可扩散配体界面,我们证明,具有快速配体迁移的表面上的细胞可以通过激活整合素α5β1来募集配体,从而在早期粘附阶段导致更快的局灶性粘附生长和扩散。通过利用紫外光敏感的锚分子来触发配体的“动态到静态”转化,我们依次激活α5β1和αvβ3整合素,显著促进间充质干细胞的成骨分化。这项研究说明了如何操纵分子动力学可以直接影响干细胞的命运,这表明“顺序”控制的移动表面作为工程智能生物材料涂层的适应性平台的潜力。
    The physical properties of nanoscale cell-extracellular matrix (ECM) ligands profoundly impact biological processes, such as adhesion, motility, and differentiation. While the mechanoresponse of cells to static ligands is well-studied, the effect of dynamic ligand presentation with \"adaptive\" properties on cell mechanotransduction remains less understood. Utilizing a controllable diffusible ligand interface, we demonstrated that cells on surfaces with rapid ligand mobility could recruit ligands through activating integrin α5β1, leading to faster focal adhesion growth and spreading at the early adhesion stage. By leveraging UV-light-sensitive anchor molecules to trigger a \"dynamic to static\" transformation of ligands, we sequentially activated α5β1 and αvβ3 integrins, significantly promoting osteogenic differentiation of mesenchymal stem cells. This study illustrates how manipulating molecular dynamics can directly influence stem cell fate, suggesting the potential of \"sequentially\" controlled mobile surfaces as adaptable platforms for engineering smart biomaterial coatings.
    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:结直肠癌(CRC)通常表现出对顺铂治疗的耐受性,但潜在的机制仍不清楚。在肿瘤微环境中,巨噬细胞在抵抗化疗的细胞毒性作用中发挥作用,通过参与细胞凋亡来清除化疗药物诱导的凋亡细胞。细胞外囊泡(EV)的参与,肿瘤微环境中的细胞间通信器,在调节有效细胞增殖以促进耐药性方面尚未得到彻底研究。
    方法:我们构建了表达GFP荧光的CRC细胞系(包括GFP-CT26和GFP-MC38),以通过流式细胞术分析检测巨噬细胞的胞出量。我们使用多步超速离心方法分离和纯化CRC分泌的EV,并通过电子显微镜和纳米流式细胞术鉴定它们。进行蛋白质组学分析以鉴定CRC-EV携带的蛋白质分子。使用CRISPR-Cas9构建MFGE8敲除CRC细胞系,并通过使用Western印迹的体外和体内实验验证其效果,免疫荧光,和流式细胞仪分析,证实这些电动汽车激活巨噬细胞αvβ3-Src-FAK-STAT3信号通路,从而促进红细胞增多。
    结果:在这项研究中,我们发现CRC衍生的EVs(CRC-EVs)可增强顺铂诱导的凋亡CRC细胞的巨噬细胞胞作用.癌症基因组图谱(TCGA)数据库的分析显示,在CRC患者中,efferocytosis相关基因MFGE8的高表达,提示预后较差。此外,基于质谱的蛋白质组分析在CRC-EV中鉴定出高丰度的MFGE8蛋白。利用CRISPR-Cas9基因编辑系统,我们产生了MFGE8敲除的CRC细胞,证明他们的电动汽车不能在体外和体内上调巨噬细胞的有效细胞增殖。此外,我们证明了在CRC-EVs中的MFGE8通过增加细胞表面αvβ3的表达来刺激巨噬细胞的细胞增殖,从而激活细胞内Src-FAK-STAT3信号通路。
    结论:因此,这项研究强调了携带MFGE8的CRC-EV激活巨噬细胞有效胞吞作用的机制.这种激活促进了顺铂诱导的凋亡CRC细胞的清除,促进CRC对顺铂的耐药。这些发现为化疗药物的潜在协同应用提供了新的见解,电动汽车抑制剂,和Efferocytosis拮抗剂用于CRC治疗。
    BACKGROUND: Colorectal cancer (CRC) commonly exhibits tolerance to cisplatin treatment, but the underlying mechanisms remain unclear. Within the tumor microenvironment, macrophages play a role in resisting the cytotoxic effects of chemotherapy by engaging in efferocytosis to clear apoptotic cells induced by chemotherapeutic agents. The involvement of extracellular vesicles (EVs), an intercellular communicator within the tumor microenvironment, in regulating the efferocytosis for the promotion of drug resistance has not been thoroughly investigated.
    METHODS: We constructed GFP fluorescent-expressing CRC cell lines (including GFP-CT26 and GFP-MC38) to detect macrophage efferocytosis through flow cytometric analysis. We isolated and purified CRC-secreted EVs using a multi-step ultracentrifugation method and identified them through electron microscopy and nanoflow cytometry. Proteomic analysis was conducted to identify the protein molecules carried by CRC-EVs. MFGE8 knockout CRC cell lines were constructed using CRISPR-Cas9, and their effects were validated through in vitro and in vivo experiments using Western blotting, immunofluorescence, and flow cytometric analysis, confirming that these EVs activate the macrophage αvβ3-Src-FAK-STAT3 signaling pathway, thereby promoting efferocytosis.
    RESULTS: In this study, we found that CRC-derived EVs (CRC-EVs) enhanced macrophage efferocytosis of cisplatin-induced apoptotic CRC cells. Analysis of The Cancer Genome Atlas (TCGA) database revealed a high expression of the efferocytosis-associated gene MFGE8 in CRC patients, suggesting a poorer prognosis. Additionally, mass spectrometry-based proteomic analysis identified a high abundance of MFGE8 protein in CRC-EVs. Utilizing CRISPR-Cas9 gene edition system, we generated MFGE8-knockout CRC cells, demonstrating that their EVs fail to upregulate macrophage efferocytosis in vitro and in vivo. Furthermore, we demonstrated that MFGE8 in CRC-EVs stimulated macrophage efferocytosis by increasing the expression of αvβ3 on the cell surface, thereby activating the intracellular Src-FAK-STAT3 signaling pathway.
    CONCLUSIONS: Therefore, this study highlighted a mechanism in CRC-EVs carrying MFGE8 activated the macrophage efferocytosis. This activation promoted the clearance of cisplatin-induced apoptotic CRC cells, contributing to CRC resistance against cisplatin. These findings provide novel insights into the potential synergistic application of chemotherapy drugs, EVs inhibitors, and efferocytosis antagonists for CRC treatment.
    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
    来自天然产物的小分子药物对于新的治疗发现至关重要。然而,他们的临床部署通常受到非特异性活动和严重不良反应的阻碍。本研究集中于3-氟-10-羟基-乙二胺(F-OH-Evo),一种有效的伊沃二胺衍生物,由于肿瘤选择性欠佳和细胞毒性增强,其发展受到限制。通过利用非凡的稳定性,特异性,和c(RGDFK)的αvβ3整联蛋白亲和力,通过将F-OH-Evo与cRGD结合合成了一种新型前药。这种创新的前药实质上增强了F-OH-Evo的肿瘤特异性靶向并提高了抗肿瘤活性。其中,化合物3c在体外对U87癌细胞表现出最佳的选择性抑制活性。它通过与αvβ3整合素结合选择性地进入U87细胞,在ROS和GSH的双重反应下释放母体分子以发挥对topoI的抑制活性。该结果强调了cRGD缀合的前药在靶向癌症治疗中的潜力。这种方法标志着在开发更安全,更有效的化疗药物方面取得了重大进展,强调前药策略在克服传统癌症治疗局限性方面的作用。
    Small molecule drugs sourced from natural products are pivotal for novel therapeutic discoveries. However, their clinical deployment is often impeded by non-specific activity and severe adverse effects. This study focused on 3-fluoro-10-hydroxy-Evodiamine (F-OH-Evo), a potent derivative of Evodiamine, whose development is curtailed due to suboptimal tumor selectivity and heightened cytotoxicity. By harnessing the remarkable stability, specificity, and αvβ3 integrin affinity of c(RGDFK), a novel prodrug by conjugating F-OH-Evo with cRGD was synthesized. This innovative prodrug substantially enhanced the tumor-specific targeting of F-OH-Evo and improved the anti-tumor activities. Among them, compound 3c demonstrated the best selective inhibitory activity toward U87 cancer cells in vitro. It selectively enterd U87 cells by binding to αvβ3 integrin, releasing the parent molecule under the dual response of ROS and GSH to exert inhibitory activity on topo I. The results highlight the potential of cRGD-conjugated prodrugs in targeted cancer therapy. This approach signifies a significant advancement in developing safer and more effective chemotherapy drugs, emphasizing the role of prodrug strategies in overcoming the limitations of traditional cancer treatments.
    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
    癌症是全球十大致命疾病之一,其中晚期转移癌死亡率最高。舒尼替尼和免疫检查点阻断剂通常用于治疗转移性肾癌,但疗效有限。因此,对于转移性肾癌,迫切需要开发新的靶向治疗方法.在这项研究中,我们设计了一种抗体融合蛋白,57103,同时靶向分化簇24(CD24),白细胞介素4受体(IL-4R),和整合素受体αvβ3和α5β1。体外实验表明57103显著抑制了细胞的增殖,迁移,入侵,菌落形成,和肾癌细胞的粘附能力,产生了全面而显著的抗肿瘤作用。此外,57103抑制血管生成,促进THP1来源的M0型巨噬细胞吞噬,并增强了外周血单核细胞和NK92MI-CD16a细胞的抗体依赖性细胞毒性。体内实验表明,在ACHN细胞异种移植裸鼠和MC38-hCD24荷瘤小鼠模型中,肿瘤生长受到显着抑制。免疫组织化学分析显示57103降低了肾癌细胞的增殖,并诱导了细胞凋亡。同时抑制血管生成。MC38-hPDL1和MC38-hCD24-hPDL1荷瘤小鼠模型进一步提供了将57103与PDL1拮抗剂阿特珠单抗组合的可能性。总之,57103是治疗转移性肾癌或PDL1过表达癌症的潜在候选药物。
    Cancer is one of the top 10 fatal diseases worldwide, among which advanced metastatic carcinoma has the highest mortality rate. Sunitinib and immune checkpoint blockers are commonly used to treat metastatic renal carcinoma with limited efficacy. Therefore, there is an urgent need to develop novel targeted therapies for metastatic renal cancer. In this study, we designed an antibody fusion protein, 57103, that simultaneously targeted the cluster of differentiation 24 (CD24), interleukin 4 receptor (IL-4R), and integrin receptors αvβ3 and α5β1. In vitro assays showed that 57103 significantly suppressed the proliferation, migration, invasion, colony formation, and adhesion abilities of renal cancer cells, resulting in a comprehensive and significant antitumor effect. Furthermore, 57103 inhibited angiogenesis, promoted THP1-derived M0-type macrophage phagocytosis, and enhanced the antibody-dependent cellular cytotoxicity of peripheral blood mononuclear and NK92MI-CD16a cells. In vivo experiments revealed significant inhibition of tumor growth in ACHN cell xenograft nude mice and an MC38-hCD24 tumor-bearing mouse model. Immunohistochemical analysis showed that 57103 decreased the proliferation and induced the apoptosis of renal cancer cells, while inhibiting angiogenesis. The MC38-hPDL1 and MC38-hCD24-hPDL1 tumor-bearing mouse models further offer the possibility of combining 57103 with the PDL1 antagonist atezolizumab. In conclusion, 57103 is a potential candidate drug for the treatment of metastatic renal carcinoma or PDL1-overexpressing cancer.
    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:后纵韧带(OPLL)骨化,一种新出现的异位骨化病,导致脊髓压迫,导致运动和感觉功能障碍。OPLL的病因尚不清楚,但可能涉及整合素αVβ3调节成骨和血管生成的过程。在这项研究中,我们专注于整合素αVβ3在OPLL中的作用,并探索了c(RGDyk)肽作为有效和选择性的整合素αVβ3抑制剂在OPLL中抑制成骨和血管生成的潜在机制。
    方法:在手术中收集OPLL或对照韧带样本。对于OPLL样本,RNA测序结果显示整合素家族的激活,特别是整合素αVβ3。通过qPCR检测整合素αVβ3的表达,西方印迹,和免疫组织化学分析。使用荧光显微镜观察c(RGDyk)肽对来自OPLL患者的韧带成纤维细胞(LFs)和内皮细胞(ECs)的整合素αVβ3的靶向抑制作用。应用qPCR检测c(RGDyk)肽对致病性LFs骨化的影响,西方印迹。碱性磷酸酶染色或茜素红染色用于测试成骨能力。c(RGDyk)肽对血管生成的作用通过EC迁移和管形成测定来确定。通过micro-CT评估c(RGDyk)肽对异位骨形成的影响,组织学,免疫组织化学,和体内免疫荧光分析。
    结果:结果表明,用c(RGDyk)治疗后,LFs成骨分化明显下降。此外,c(RGDyk)肽抑制EC的迁移,从而阻止了成骨所需的营养支持。此外,c(RGDyk)肽抑制小鼠异位骨形成。机制分析表明,c(RGDyk)肽可以通过靶向整合素αVβ3并调节FAK/ERK途径来抑制OPLL中的成骨和血管生成。
    结论:因此,整合素αVβ3似乎是OPLL的新兴治疗靶点,和c(RGDyk)肽具有双重抑制作用,这对于OPLL的新治疗策略可能是有价值的。
    BACKGROUND: Ossification of the posterior longitudinal ligament (OPLL), an emerging heterotopic ossification disease, causes spinal cord compression, resulting in motor and sensory dysfunction. The etiology of OPLL remains unclear but may involve integrin αVβ3 regulating the process of osteogenesis and angiogenesis. In this study, we focused on the role of integrin αVβ3 in OPLL and explored the underlying mechanism by which the c(RGDyk) peptide acts as a potent and selective integrin αVβ3 inhibitor to inhibit osteogenesis and angiogenesis in OPLL.
    METHODS: OPLL or control ligament samples were collected in surgery. For OPLL samples, RNA-sequencing results revealed activation of the integrin family, particularly integrin αVβ3. Integrin αVβ3 expression was detected by qPCR, Western blotting, and immunohistochemical analysis. Fluorescence microscopy was used to observe the targeted inhibition of integrin αVβ3 by the c(RGDyk) peptide on ligaments fibroblasts (LFs) derived from patients with OPLL and endothelial cells (ECs). The effect of c(RGDyk) peptide on the ossification of pathogenic LFs was detected using qPCR, Western blotting. Alkaline phosphatase staining or alizarin red staining were used to test the osteogenic capability. The effect of the c(RGDyk) peptide on angiogenesis was determined by EC migration and tube formation assays. The effects of the c(RGDyk) peptide on heterotopic bone formation were evaluated by micro-CT, histological, immunohistochemical, and immunofluorescence analysis in vivo.
    RESULTS: The results indicated that after being treated with c(RGDyk), the osteogenic differentiation of LFs was significantly decreased. Moreover, the c(RGDyk) peptide inhibited the migration of ECs and thus prevented the nutritional support required for osteogenesis. Furthermore, the c(RGDyk) peptide inhibited ectopic bone formation in mice. Mechanistic analysis revealed that c(RGDyk) peptide could inhibit osteogenesis and angiogenesis in OPLL by targeting integrin αVβ3 and regulating the FAK/ERK pathway.
    CONCLUSIONS: Therefore, the integrin αVβ3 appears to be an emerging therapeutic target for OPLL, and the c(RGDyk) peptide has dual inhibitory effects that may be valuable for the new therapeutic strategy of OPLL.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号