c-Met

C - MET
  • 文章类型: Journal Article
    肝细胞生长因子(HGF)在多个系统中表达,介导多种生物学活性,如有丝分裂,运动性,和形态发生。越来越多的研究揭示了HGF在从产前到成年期的卵巢和睾丸生理中的表达模式和功能。HGF通过调节卵巢中卵泡膜细胞和颗粒细胞的功能来调节卵泡生成和类固醇生成。它还介导体细胞增殖和类固醇生成,从而影响男性的精子发生。除了对生殖系统的生理影响外,近年来,HGF在治疗男性和女性不育症的临床前研究中显示出优势。特别是在妇女与卵巢早衰。本文旨在总结HGF在生殖系统中的多效性功能,为其临床应用提供前景。
    Hepatocyte growth factor (HGF) is expressed in multiple systems and mediates a variety of biological activities, such as mitosis, motility, and morphogenesis. A growing number of studies have revealed the expression patterns and functions of HGF in ovarian and testicular physiology from the prenatal to the adult stage. HGF regulates folliculogenesis and steroidogenesis by modulating the functions of theca cells and granulosa cells in the ovary. It also mediates somatic cell proliferation and steroidogenesis, thereby affecting spermatogenesis in males. In addition to its physiological effects on the reproductive system, HGF has shown advantages in preclinical studies over recent years for the treatment of male and female infertility, particularly in women with premature ovarian insufficiency. This review aims to summarize the pleiotropic functions of HGF in the reproductive system and to provide prospects for its clinical application.
    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
    作为空气交换的场所,肺组织长期暴露于大量的外来病原体。因此,肺已经发展出精致而复杂的免疫系统。除了它们的物理和化学屏障作用,肺上皮细胞可通过表达Toll样受体(TLRs)和其他模式识别受体,随着细胞因子的分泌。新的证据表明,肺上皮细胞可以产生和分泌免疫球蛋白(Ig),包括IgM,IgA,或IgG,从而执行抗体功能。此外,已经发现恶性转化的肺上皮细胞产生高水平的Ig,主要是IgG,不能实现抗体的作用,而是进行促进肿瘤的活动。结构分析表明,由于独特的糖基化修饰,肺癌细胞产生的IgG的生物学活性与正常肺上皮细胞产生的Ig的生物学活性不同。具体来说,唾液酸化IgG(SIA-IgG),以在IgyCH1的Asn162位点的非传统N-糖基化修饰为特征,在肿瘤干细胞中高度表达。已经证明SIA-IgG依赖于这种独特的唾液酸化修饰来促进肿瘤发生。转移,和免疫逃避。目前的研究结果证明,肺上皮细胞产生的Ig具有多方面的生物学活性,包括生理条件下的免疫防御功能,同时在恶性转化过程中获得促进肿瘤的活性。这些见解作为新的生物标志物和靶标,具有诊断和治疗肺癌的潜力。
    As the locus for air exchange, lung tissue is perpetually exposed to a significant quantity of foreign pathogens. Consequently, lung has developed a refined and intricate immune system. Beyond their physical and chemical barrier roles, lung epithelial cells can contribute to immune defence through the expression of Toll-like receptors (TLRs) and other pattern recognition receptors, along with the secretion of cytokines. Emerging evidence demonstrates that lung epithelial cells can generate and secrete immunoglobulins (Igs), including IgM, IgA, or IgG, thus performing antibody function. Moreover, malignantly transformed lung epithelial cells have been discovered to produce high levels of Ig, predominantly IgG, which do not fulfill the role of antibodies, but instead carries out tumour-promoting activity. Structural analysis has indicated that the biological activity of IgG produced by lung cancer cells differs from that of Igs produced by normal lung epithelial cells due to the unique glycosylation modification. Specifically, the sialylated IgG (SIA-IgG), characterised by a non-traditional N-glycosylation modification at the Asn162 site of Igγ CH1, is highly expressed in tumour stem cells. It has been demonstrated that SIA-IgG relies on this unique sialylation modification to promote tumorigenesis, metastasis, and immune evasion. Current results have proven that the Ig produced by lung epithelial cells has multifaceted biological activities, including immune defence functions under physiological conditions, while acquiring tumour-promoting activity during malignant transformation. These insights possess potential for the diagnosis and treatment of lung cancer as novel biomarkers and targets.
    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
    胃癌和胃食管连接部癌是全球肿瘤相关死亡的主要原因。尽管免疫治疗和分子靶向治疗的进展扩大了治疗选择,对于不可切除或转移性胃癌患者,他们的预后没有显著改变。少数患者,特别是那些PD-L1阳性的,HER-2阳性,或MSI高肿瘤,可能从晚期阶段的免疫检查点抑制剂和/或HER-2定向疗法中受益更多。然而,对于那些缺乏特定靶标和独特分子特征的人,常规化疗仍然是唯一推荐的有效和持久的治疗方案.在这次审查中,我们总结了各种信号通路的作用,并进一步研究了可用的靶标。然后,晚期胃癌II/III期临床试验的当前结果,随着现有生物标志物的优势和局限性,具体讨论。最后,当遇到重大挑战时,我们将提供我们对精准治疗模式的见解。
    Gastric cancer and gastroesophageal junction cancer represent the leading cause of tumor-related death worldwide. Although advances in immunotherapy and molecular targeted therapy have expanded treatment options, they have not significantly altered the prognosis for patients with unresectable or metastatic gastric cancer. A minority of patients, particularly those with PD-L1-positive, HER-2-positive, or MSI-high tumors, may benefit more from immune checkpoint inhibitors and/or HER-2-directed therapies in advanced stages. However, for those lacking specific targets and unique molecular features, conventional chemotherapy remains the only recommended effective and durable regimen. In this review, we summarize the roles of various signaling pathways and further investigate the available targets. Then, the current results of phase II/III clinical trials in advanced gastric cancer, along with the superiorities and limitations of the existing biomarkers, are specifically discussed. Finally, we will offer our insights in precision treatment pattern when encountering the substantial challenges.
    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
    分子工程能够创造具有新功能的适体,但前提是对其结构和识别机制的深刻理解。由于c-MET/HGF信号通路在肿瘤发展和侵袭中的关键作用,细胞间充质上皮转化因子(c-MET)引起了广泛关注。这项研究报道了构建与c-MET和其他特定蛋白质结合的新型嵌合适体的策略。c-MET被鉴定为DNA适体的分子靶标,HF3-58,通过细胞-SELEX选择。系统研究了HF3-58与c-MET的结合结构和机理,露出脚手架,认可,和冗余区域。通过分子工程设计,冗余区域被其他具有茎环结构的适体取代,产生新的嵌合适体,具有双特异性结合c-MET和特异性蛋白。嵌合双特异性适体HF-3b显示出介导T细胞与肿瘤细胞粘附的能力,提示在肿瘤免疫治疗中的前瞻性用途。这些发现表明适体HF3-58可以作为分子工程平台用于开发靶向c-MET的多种多功能配体。此外,全面了解适体的结合机制将为功能性适体的设计提供指导,显著扩大其潜在的应用。
    Molecular engineering enables the creation of aptamers with novel functions, but the prerequisite is a deep understanding of their structure and recognition mechanism. The cellular-mesenchymal epithelial transition factor (c-MET) is garnering significant attention due to the critical role of the c-MET/HGF signaling pathway in tumor development and invasion. This study reports a strategy for constructing novel chimeric aptamers that bind to both c-MET and other specific proteins. c-MET was identified to be the molecular target of a DNA aptamer, HF3-58, selected through cell-SELEX. The binding structure and mechanism of HF3-58 with c-MET were systematically studied, revealing the scaffold, recognition, and redundancy regions. Through molecular engineering design, the redundancy region was replaced with other aptamers possessing stem-loop structures, yielding novel chimeric aptamers with bispecificity for binding to c-MET and specific proteins. A chimeric bispecific aptamer HF-3b showed the ability to mediate the adhesion of T-cells to tumor cells, suggesting the prospective utility in tumor immunotherapy. These findings suggest that aptamer HF3-58 can serve as a molecular engineering platform for the development of diverse multifunctional ligands targeting c-MET. Moreover, comprehensive understanding of the binding mechanisms of aptamers will provide guidance for the design of functional aptamers, significantly expanding their potential applications.
    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
    目的:本研究的目的是通过虚拟筛选鉴定抗EGFR/c-Met的双靶点抑制剂,动态仿真,和生物活性评价。这一努力旨在克服由L858R/T790M突变体诱导的药物抗性的挑战。
    方法:收集活性结构以构建药物分子组。接下来,将性质过滤应用于化合物库中的药物结构。然后通过虚拟筛选和聚类分析鉴定活性化合物。随后,我们对活性化合物进行了MTT抗肿瘤活性评估和激酶抑制试验,以确定最有希望的候选化合物.此外,对所选化合物进行AO染色和JC-1测定。最终,优选化合物与EGFR和c-Met蛋白进行分子对接和分子动力学模拟,分别。
    结果:T13074的IC50对于EGFRL858R/T790M激酶为2.446μM,对于c-Met激酶为7.401nM,强调其在克服EGFRL858R/T790M抗性方面的潜力。此外,T13074对H1975细胞的IC50为1.93μM。AO染色和JC-1测定的结果表明T13074以浓度依赖性方式诱导肿瘤细胞凋亡。值得注意的是,发现T13074与EGFR蛋白之间的结合能为-90.329±16.680kJ/mol,与c-Met蛋白的结合能为-139.935±17.414kJ/mol。
    结论:T13074在体内和体外均表现出优异的抗肿瘤活性,表明其作为双靶点EGFR/c-Met抑制剂的潜在效用。这表明其在克服由L858R/T790M突变诱导的EGFR抗性中的有希望的作用。
    OBJECTIVE: The objective of this study is to identify dual-target inhibitors against EGFR/c-Met through virtual screening, dynamic simulation, and biological activity evaluation. This endeavor is aimed at overcoming the challenge of drug resistance induced by L858R/T790M mutants.
    METHODS: Active structures were gathered to construct sets of drug molecules. Next, property filtering was applied to the drug structures within the compound library. Active compounds were then identified through virtual screening and cluster analysis. Subsequently, we conducted MTT antitumor activity evaluation and kinase inhibition assays for the active compounds to identify the most promising candidates. Furthermore, AO staining and JC-1 assays were performed on the selected compounds. Ultimately, the preferred compounds underwent molecular docking and molecular dynamics simulation with the EGFR and c-Met proteins, respectively.
    RESULTS: The IC50 of T13074 was determined as 2.446 μM for EGFRL858R/T790M kinase and 7.401 nM for c-Met kinase, underscoring its potential in overcoming EGFRL858R/T790M resistance. Additionally, T13074 exhibited an IC50 of 1.93 μM on the H1975 cell. Results from AO staining and JC-1 assays indicated that T13074 induced tumor cell apoptosis in a concentration-dependent manner. Notably, the binding energy between T13074 and EGFR protein was found to be -90.329 ± 16.680 kJ/mol, while the binding energy with c-Met protein was -139.935 ± 17.414 kJ/mol.
    CONCLUSIONS: T13074 exhibited outstanding antitumor activity both in vivo and in vitro, indicating its potential utility as a dual-target EGFR/c-Met inhibitor. This suggests its promising role in overcoming EGFR resistance induced by the L858R/T790M mutation.
    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
    间充质上皮转化因子(c-Met)是一种与细胞增殖相关的受体酪氨酸激酶,生存,入侵,以及几种癌症的转移,包括结直肠癌(CRC),特别是在异常激活时。我们的研究策略性地设计了源自c-Met和抗体Onartuzumab之间相互作用的肽。通过利用循环策略,我们实现了显著增强的肽稳定性和亲和力。我们的体外评估证实,与线性对应物相比,环状肽HYNIC-cycOn表现出更高的亲和力(KD=83.5nM)和更高的特异性。通过体内实验,[99mTc]Tc-HYNIC-cycOn在非肿瘤细胞中显示出优异的肿瘤靶向能力和最小的吸收,如单光子发射计算机断层扫描所证实。值得注意的是,肿瘤与肌肉和肿瘤与肠道的比例,注射后1小时,分别为4.78±0.86和3.24±0.47。在原位CRC模型中观察到相当的比率,记录分别为4.94±0.32和3.88±0.41。总之,[99mTc]Tc-HYNIC-cycOn显示出作为临床应用的候选物的实质性希望。我们表明[99mTc]Tc-HYNIC-cycOn可以有效地靶向和观察体内表达c-Met的肿瘤,为在CRC中检测c-Met时提高诊断准确性提供了一种有前途的方法。
    The mesenchymal-epithelial transition factor (c-Met) is a receptor tyrosine kinase linked to the proliferation, survival, invasion, and metastasis of several types of cancers, including colorectal cancer (CRC), particularly when aberrantly activated. Our study strategically designs peptides derived from interactions between c-Met and the antibody Onartuzumab. By utilizing a cyclic strategy, we achieved significantly enhanced peptide stability and affinity. Our in vitro assessments confirmed that the cyclic peptide HYNIC-cycOn exhibited a higher affinity (KD = 83.5 nM) and greater specificity compared with its linear counterpart. Through in vivo experiments, [99mTc]Tc-HYNIC-cycOn displayed exceptional tumor-targeting capabilities and minimal absorption in nontumor cells, as confirmed by single-photon emission computed tomography. Notably, the ratios of tumor to muscle and tumor to intestine, 1 h postinjection, were 4.78 ± 0.86 and 3.24 ± 0.47, respectively. Comparable ratios were observed in orthotopic CRC models, recording 4.94 ± 0.32 and 3.88 ± 0.41, respectively. In summary, [99mTc]Tc-HYNIC-cycOn shows substantial promise as a candidate for clinical applications. We show that [99mTc]Tc-HYNIC-cycOn can effectively target and visualize c-Met-expressing tumors in vivo, providing a promising approach for enhancing diagnostic accuracy when detecting c-Met in CRC.
    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
    胰腺导管腺癌(PDAC)恶性程度极高,进展迅速。PDAC对当前治疗方法的总体响应率仍然不令人满意。因此,确定新的靶标并阐明与PDAC进展相关的潜在机制可能会提供额外的治疗策略.AHNAK2在多种肿瘤中异常表达并发挥促肿瘤发生作用。然而,AHNAK2在PDAC中的生物学作用尚不清楚。
    通过免疫组织化学(IHC)和定量实时聚合酶链反应(qRT-PCR)检测PDAC和配对的非肿瘤组织中AHNAK2的表达。进行慢病毒敲除以研究AHNAK2对胰腺癌细胞生物学功能的影响。采用皮下细胞来源的异种移植(CDX)模型和AHNAK2沉默的KPC自发小鼠模型,观察AHNAK2对肿瘤生长和预后的影响。使用蛋白质印迹评估响应于HGF处理的蛋白水平的c-MET的表达。
    我们的结果表明,AHNAK2在PDAC临床样本中高表达,并与不良预后相关。敲除能显著抑制AHNAK2的增殖,迁移,和胰腺癌细胞的侵袭。AHNAK2敲低或敲除导致具有PDAC的小鼠中的肿瘤生长抑制和延长的存活。此外,AHNAK2和c-MET表达水平在转录后水平显示出显著的正相关。机械上,AHNAK2通过阻止c-MET降解和持续激活HGF/c-MET信号通路促进肿瘤进展。
    总的来说,我们的研究表明,AHNAK2通过调节c-MET信号通路在PDAC进展中发挥重要作用,靶向AHNAK2可能是PDAC的有效治疗策略。
    UNASSIGNED: Pancreatic ductal adenocarcinoma (PDAC) is extremely malignant and rapidly progresses. The overall response rate of PDAC to current treatment methods is still unsatisfactory. Thus, identifying novel targets and clarifying the underlying mechanisms associated with PDAC progression may potentially offer additional treatment strategies. AHNAK2 is aberrantly expressed in a variety of tumors and exerts pro-tumorigenic effects. However, the biological role of AHNAK2 in PDAC remains poorly understood.
    UNASSIGNED: The expression of AHNAK2 in PDAC and paired non-tumor tissues was detected by immunohistochemistry (IHC) and quantitative real-time polymerase chain reaction (qRT-PCR). Lentivirus knockdown was performed to investigate the impact of AHNAK2 on the biological function of pancreatic cancer cells. The subcutaneous cell-derived xenograft (CDX) model and the KPC spontaneous mouse model with AHNAK2 silencing were used to observe the effects of AHNAK2 on tumor growth and prognosis. The expression of c-MET at protein level in response to HGF treatment was assessed using western blot.
    UNASSIGNED: Our results demonstrated that AHNAK2 was highly expressed in PDAC clinical samples and associated with poor prognosis. Knockdown of AHNAK2 significantly inhibited the proliferation, migration, and invasion of pancreatic cancer cells. AHNAK2 knockdown or knockout resulted in tumor growth suppression and prolonged survival in mice with PDAC. In addition, AHNAK2 and c-MET expression levels showed a significant positive correlation at the post-transcriptional level. Mechanistically, AHNAK2 promoted tumor progression by preventing c-MET degradation and persistently activating the HGF/c-MET signaling pathway.
    UNASSIGNED: Overall, our study revealed that AHNAK2 plays an important role in PDAC progression by modulating the c-MET signaling pathway, and targeting AHNAK2 may be an effective therapeutic strategy for PDAC.
    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
    细胞间充质上皮转化因子(c-Met)是位于7q31基因座上的受体酪氨酸激酶(RTK),编码Met原癌基因,在调节细胞增殖中起关键作用,转移,分化,和凋亡通过各种信号通路。然而,它的异常激活和过表达与许多人类癌症有关。因此,c-Met是癌症治疗的有希望的靶标。然而,由于信号系统的复杂性以及不同蛋白质和酶的参与,选择性单靶向药物的抗癌作用受到限制。在抑制一条途径后,信号分子可以通过其他途径传播,导致单一靶向药物治疗效果不佳。同时阻断c-Met和另一个因子的双重抑制剂可以显着提高疗效并克服单靶标抑制剂的一些缺点,包括耐药性。在这次审查中,我们介绍了c-Met激酶以及c-Met与其他抗肿瘤靶点之间的协同作用。然后总结了基于c-Met治疗癌症的双靶点抑制剂,并详细讨论了它们的设计概念和结构-活性关系(SARs),为进一步开发基于c-Met的新型双重抑制剂提供了有价值的见解。
    The cellular-mesenchymal epithelial transition factor (c-Met) is a receptor tyrosine kinase (RTK) located on the 7q31 locus encoding the Met proto-oncogene and plays a critical role in regulating cell proliferation, metastasis, differentiation, and apoptosis through various signaling pathways. However, its aberrant activation and overexpression have been implicated in many human cancers. Therefore, c-Met is a promising target for cancer treatment. However, the anticancer effect of selective single-targeted drugs is limited due to the complexity of the signaling system and the involvement of different proteins and enzymes. After inhibiting one pathway, signal molecules can be transmitted through other pathways, resulting in poor efficacy of single-targeted drug therapy. Dual inhibitors that simultaneously block c-Met and another factor can significantly improve efficacy and overcome some of the shortcomings of single-target inhibitors, including drug resistance. In this review, We introduced c-Met kinase and the synergism between c-Met and other anti-tumor targets, then dual-target inhibitors based on c-Met for the treatment of cancers were summarized and their design concepts and structure-activity relationships (SARs) were discussed elaborately, providing a valuable insight for the further development of novel c-Met-based dual inhibitors.
    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:间充质干细胞(MSCs)移植是一种新开发的治疗急性肝衰竭(ALF)的策略。尽管如此,MSCs移植后存活率低,对受损组织归巢不良,限制了MSCs的临床应用。该研究评估了缺氧预处理(HPC)是否可以提高人羊膜间充质干细胞(hA-MSCs)的生物学活性。促进它们对ALF小鼠肝脏的归巢能力,影响肝组织修复。
    方法:流式细胞术,CCK8,Transwell,和蛋白质印迹试验进行,以评估低氧预处理对表型的影响,扩散,研究了hA-MSCs的迁移以及c-Met和CXCR4基因表达水平的变化。评价缺氧预处理hA-MSCs移植对D-氨基半乳糖(D-GalN)/LPS诱导的ALF归巢和修复的影响。通过添加c-Met阐明了机制,CXCR4特异性阻断剂(SU11274和AMD3100)。
    结果:缺氧预处理(1%氧气体积分数)后,hA-MSCs保持粘附和涡旋集落生长的形态学特征,并且显示高CD44、CD90和CD105表达水平和低CD31、CD34和CD45表达水平。hA-MSCs的低氧预处理显著增加其增殖和迁移,并高表达c-Met和CXCR4基因。体内和体外,c-Met特异性阻断剂SU11274抑制了这种促迁移作用.在急性肝衰竭小鼠模型中,HGF在肝脏中的表达水平明显高于血清中的表达水平,肺和肾.缺氧预处理hA-MSCs的移植显著改善了ALF小鼠的肝功能和存活率,增强了肝细胞的抗凋亡能力。c-Met特异性阻断剂SU11274抑制了低氧预处理的抗凋亡增强作用。观察到低氧hA-MSC施用比施用正常氧-hA-MSC后记录的肝脏中的荧光细胞显著增加。添加c-Met抑制剂SU11274后,与低氧预处理后记录的相比,肝荧光细胞数量显着减少,而c-Met抑制剂SU11274对正常氧-hA-MSCs的影响不显著。
    结论:低氧预处理对人羊膜间充质干细胞的形态和表型特征无影响,但是它可以促进它们的扩散,迁移,抗凋亡作用,和归巢率,提高急性肝衰竭的修复,这可能是由HGF/c-Met信号轴介导的。
    BACKGROUND: Transplantation of mesenchymal stem cells (MSCs) is a newly developed strategy for treating acute liver failure (ALF). Nonetheless, the low survival rate of MSCs after transplantation and their poor homing to damaged tissues limit the clinical application of MSCs. The research assessed whether hypoxic preconditioning (HPC) can improve the biological activity of human amniotic mesenchymal stem cells (hA-MSCs), promote their homing ability to the liver of mice with ALF, and influence liver tissue repair.
    METHODS: Flow cytometry, CCK8, Transwell, and Western blotting assays were conducted to assess the effects of hypoxic preconditioning on the phenotype, proliferation, and migration of hA-MSCs and the changes in the c-Met and CXCR4 gene expression levels were studied. To evaluate the effects of the transplantation of hypoxic preconditioning of hA-MSCs on the homing and repair of D-galactosamine (D-GalN)/LPS-induced ALF, the mechanism was elucidated by adding c-Met, CXCR4-specific blockers (SU11274 and AMD3100).
    RESULTS: After hypoxia pretreatment (1% oxygen volume fraction), hA-MSCs maintained the morphological characteristics of adherence and vortex colony growth and showed high CD44, CD90, and CD105 and low CD31, CD34, and CD45 expression levels. Hypoxic preconditioning of hA-MSCs significantly increased their proliferation and migration and highly expressed the c-Met and CXCR4 genes. In vivo and in vitro, this migration-promoting effect was suppressed by the c-Met specific blocker SU11274. In the acute liver failure mouse model, the HGF expression level was considerably elevated in the liver than that in the serum, lungs and kidneys. The transplantation of hypoxic preconditioned hA-MSCs introduced a remarkable improvement in the liver function and survival rate of mice with ALF and enhanced the anti-apoptosis ability of liver cells. The anti-apoptotic enhancing effect of hypoxic preconditioning was suppressed by the c-Met specific blocker SU11274. Hypoxic hA-MSCs administration was observed to have considerably increased the fluorescent cells in the liver than that recorded after administering normal oxygen-hA-MSCs. The number of hepatic fluorescent cells decreased remarkably after adding the c-Met inhibitor SU11274, compared to that recorded after hypoxic pretreatment, whereas the effect of c-Met inhibitor SU11274 on normal oxygen-hA-MSCs was not significant.
    CONCLUSIONS: Hypoxic preconditioning depicted no impact on the morphology and phenotype features of the human amniotic mesenchymal stem cells, but it can promote their proliferation, migration, anti-apoptotic effect, and homing rate and improve the repair of acute liver failure, which might be mediated by the HGF/c-Met signaling axis.
    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
    免疫检查点蛋白阻断(ICB)已成为一种强大的免疫治疗方法,但抑制非癌细胞和正常组织的免疫相关不良事件(irAEs)仍然具有挑战性.以肿瘤微环境高度表达的分子为刺激,开发了可激活的ICB,但是考虑到刺激分子在整个肿瘤组织中的扩散,它们仍然缺乏精确性和效率。这里我们用双链DNA链组装PD-L1,被称为“安全捕获”,规范其对ICB的可及性。非癌细胞的安全捕获保持在“开启”状态,以防止ICB与PD-L1结合。癌细胞膜蛋白c-Met作为触发蛋白与安全捕获反应,选择性地暴露其用于ICB试剂的杂交区域。ICB试剂是一种可伸缩的DNA纳米串,具有重复的发夹结构单元,其收缩驱动PD-L1聚类,内吞引导降解。安全捕捉,甚至保持“安全开启”状态,通过DNA链置换反应从细胞膜上去除,以最大程度地减少其对非癌细胞的影响。该策略显示了仅在体外和体内针对癌细胞的选择性和有效的免疫治疗能力。并显示对正常组织中的irAE的有效抑制,因此将成为小鼠精确免疫疗法的一种有希望的方法。
    Immune checkpoint protein blockade (ICB) has emerged as a powerful immunotherapy approach, but suppressing immune-related adverse events (irAEs) for noncancerous cells and normal tissues remains challenging. Activatable ICB has been developed with tumor microenvironment highly-expressed molecules as stimuli, but they still lack precision and efficiency considering the diffusion of stimuli molecules in whole tumor tissue. Here we assemble PD-L1 with a duplex DNA strand, termed as \"safety catch\", to regulate its accessibility for ICB. The safety catch remains at \"on\" status for noncancerous cells to prevent ICB binding to PD-L1. Cancer cell membrane protein c-Met acts as a trigger protein to react with safety catch, which selectively exposes its hybridization region for ICB reagent. The ICB reagent is a retractable DNA nanostring with repeating hairpin-structural units, whose contraction drives PD-L1 clustering with endocytosis-guided degradation. The safety catch, even remained at \"safety on\" status, is removed from the cell membrane via a DNA strand displacement reaction to minimize its influence on noncancerous cells. This strategy demonstrates selective and potent immunotherapeutic capabilities only against cancer cells both in vitro and in vivo, and shows effective suppression of irAEs in normal tissues, therefore would become a promising approach for precise immunotherapy in mice.
    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