AXL

Axl
  • 文章类型: Journal Article
    激活EGFR(表皮生长因子受体)突变可以被特异性酪氨酸激酶抑制剂(TKIs)抑制,改变了肺癌治疗的前景。然而,由于次级突变和旁路受体,如AXL(AXL受体酪氨酸激酶),耐药性最终出现在大多数使用第一种药物治疗的患者中,第二-,或第三代TKI(例如,奥希替尼)。为了抑制AXL和对奥希替尼的耐药性,我们比较了两种抗AXL药物,抗体(mAb654)和TKI(bemcentinib)。虽然没有一对奥希替尼和抗AXL药物能够预防复发,三胞胎联合奥希替尼,西妥昔单抗(抗EGFR抗体),和任何一种抗AXL药物最初都是有效的。然而,更长的监测揭示了含mAb654的三联体的优越性,可能是由于受体内吞作用的诱导,激活免疫机制,或禁用内在变异器。因此,我们构建了一种同时参与AXL和EGFR的双特异性抗体。当与奥希替尼合用时,双特异性抗体持续抑制肿瘤复发,这需要临床试验。
    Activating EGFR (epidermal growth factor receptor) mutations can be inhibited by specific tyrosine kinase inhibitors (TKIs), which have changed the landscape of lung cancer therapy. However, due to secondary mutations and bypass receptors, such as AXL (AXL receptor tyrosine kinase), drug resistance eventually emerges in most patients treated with the first-, second-, or third-generation TKIs (e.g., osimertinib). To inhibit AXL and resistance to osimertinib, we compare two anti-AXL drugs, an antibody (mAb654) and a TKI (bemcentinib). While no pair of osimertinib and an anti-AXL drug is able to prevent relapses, triplets combining osimertinib, cetuximab (an anti-EGFR antibody), and either anti-AXL drug are initially effective. However, longer monitoring uncovers superiority of the mAb654-containing triplet, possibly due to induction of receptor endocytosis, activation of immune mechanisms, or disabling intrinsic mutators. Hence, we constructed a bispecific antibody that engages both AXL and EGFR. When combined with osimertinib, the bispecific antibody consistently inhibits tumor relapses, which warrants clinical trials.
    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
    蛛网膜下腔出血(SAH)后的白质损伤(WMI)通常会导致患者预后不良。先前的研究表明,SAH后的小胶质细胞M1极化导致淀粉样前体蛋白(APP)的积累和髓鞘碱性蛋白(MBP)的降解,从而催化WMI的恶化。因此,向M2表型(神经保护状态)转变小胶质细胞极化代表了逆转WMI的潜在治疗方法。SIRT2基因在神经疾病如神经变性和缺血性中风中至关重要。然而,其在SAH中的功能和潜在机制,特别是它如何影响小胶质细胞功能以改善WMI,仍然不清楚。我们的调查显示,在SAH后,SIRT2表达时间增加,主要在大脑体区域,与小胶质细胞有显著的共定位。然而,在施用SIRT2抑制剂AK-7后,观察到大鼠小胶质细胞极化向M2表型的转变以及短期和长期神经元功能的改善.机械上,CO-IP实验证实SIRT2可以与TAM受体家族中的受体酪氨酸激酶Axl相互作用,并充当脱乙酰酶来调节Axl的脱乙酰化。同时,AK-7对SIRT2的抑制可导致Axl的表达增加和抗炎通路PI3K/Akt信号通路的激活,它调节小胶质细胞M2极化,从而减少WMI。然而,当通过将shAxl病毒注射到侧脑室中抑制Axl表达时,下游信号通路被显著抑制。救援实验还证实,PI3K抑制剂可以逆转AK-7的神经保护作用。这些数据表明,SIRT2通过Axl/PI3K/AKT途径影响小胶质细胞极化来影响WMI,AK-7可以作为改善SAH患者神经功能的有效治疗药物。
    White matter injury (WMI) subsequent to subarachnoid hemorrhage (SAH) frequently leads to an unfavorable patient prognosis. Previous studies have indicated that microglial M1 polarization following SAH results in the accumulation of amyloid precursor protein (APP) and degradation of myelin basic protein (MBP), thereby catalyzing the exacerbation of WMI. Consequently, transitioning microglial polarization towards the M2 phenotype (neuroprotective state) represents a potential therapeutic approach for reversing WMI. The SIRT2 gene is pivotal in neurological disorders such as neurodegeneration and ischemic stroke. However, its function and underlying mechanisms in SAH, particularly how it influences microglial function to ameliorate WMI, remain unclear. Our investigations revealed that in post-SAH, there was a temporal increase in SIRT2 expression, predominantly in the cerebral corpus callosum area, with notable colocalization with microglia. However, following the administration of the SIRT2 inhibitor AK-7, a shift in microglial polarization towards the M2 phenotype and an improvement in both short-term and long-term neuronal functions in rats were observed. Mechanistically, CO-IP experiments confirmed that SIRT2 can interact with the receptor tyrosine kinase Axl within the TAM receptor family and act as a deacetylase to regulate the deacetylation of Axl. Concurrently, the inhibition of SIRT2 by AK-7 can lead to increased expression of Axl and activation of the anti-inflammatory pathway PI3K/Akt signaling pathway, which regulates microglial M2 polarization and consequently reduces WMI. However, when Axl expression was inhibited by the injection of the shAxl virus into the lateral ventricles, the downstream signaling pathways were significantly suppressed. Rescue experiments also confirmed that the neuroprotective effects of AK-7 can be reversed by PI3K inhibitors. These data suggest that SIRT2 influences WMI by affecting microglial polarization through the Axl/PI3K/AKT pathway, and that AK-7 could serve as an effective therapeutic drug for improving neurological functions in SAH patients.
    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
    我们先前报道,表皮生长因子受体酪氨酸激酶抑制剂(EGFR-TKI)奥希替尼和AXL抑制剂ONO-7475的联合治疗可有效预防高表达AXL的EGFR突变的非小细胞肺癌(NSCLC)细胞中耐药细胞的存活。然而,某些残留细胞预计最终会对这种联合疗法产生获得性抗性.在这项研究中,在高表达AXL的EGFR突变的NSCLC中,我们尝试从一线开始建立多药联合治疗,以克服对该联合治疗的耐药性.siRNA筛选试验显示,在奥希替尼-ONO-7475组合存在下,成纤维细胞生长因子受体1(FGFR1)敲低诱导细胞活力的显著抑制,它通过c-Myc途径上调FGF2来激活FGFR1。基于细胞的检测表明,奥希替尼三联疗法,与奥希替尼-ONO-7475治疗相比,ONO-7475和FGFR抑制剂BGJ398通过增加促凋亡因子Bim的表达和降低细胞活力而显著增加细胞凋亡。异种移植模型显示三联疗法显著抑制肿瘤再生长。额外的初始FGFR1抑制的新治疗策略可能在抑制耐葡聚糖和ONO-7475细胞的出现方面非常有效。
    We previously reported that combined therapy with epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) osimertinib and AXL inhibitor ONO-7475 is effective in preventing the survival of drug-tolerant cells in high-AXL-expressing EGFR-mutated non-small cell lung cancer (NSCLC) cells. Nevertheless, certain residual cells are anticipated to eventually develop acquired resistance to this combination therapy. In this study, we attempted to establish a multidrug combination therapy from the first-line setting to overcome resistance to this combination therapy in high-AXL-expressing EGFR-mutated NSCLC. siRNA screening assay showed that fibroblast growth factor receptor 1 (FGFR1) knockdown induced pronounced inhibition of cell viability in the presence of the osimertinib-ONO-7475 combination, which activates FGFR1 by upregulating FGF2 via the c-Myc pathway. Cell-based assays showed that triple therapy with osimertinib, ONO-7475, and the FGFR inhibitor BGJ398 significantly increased apoptosis by increasing expression of proapoptotic factor Bim and reduced cell viability compared with that observed for the osimertinib-ONO-7475 therapy. Xenograft models showed that triple therapy considerably suppressed tumor regrowth. A novel therapeutic strategy of additional initial FGFR1 inhibition may be highly effective in suppressing the emergence of osimertinib- and ONO-7475-resistant cells.
    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
    拉泽替尼,一种新的第三代表皮生长因子受体酪氨酸激酶抑制剂(EGFR-TKI),在EGFR突变型肺癌中证明了显著的功效。然而,抗性通常发展,促使人们考虑治疗策略以克服最初的耐药机制。这项研究旨在阐明对lazertinib的适应性耐药,并倡导新型联合治疗,证明其作为EGFR突变阳性NSCLC的一线治疗在预防耐药方面的功效。我们发现AXL敲低在存在拉泽替尼的情况下显着抑制肺癌细胞的活力,表明AXL激活有助于拉泽替尼耐药。然而,Lazertinib和AXL抑制剂的组合长期培养导致残余细胞增殖并增加MCL-1表达水平,它是由转录因子YAP的核易位介导的。MCL-1或YAP抑制剂与lazertinib和AXL抑制剂组合的三联疗法显着降低细胞活力并增加凋亡率。这些结果表明,AXL和YAP/MCL-1信号有助于EGFR突变肺癌细胞的适应性拉泽替尼耐药,提示AXL和YAP/MCL-1的初始双重抑制可能是消除拉泽替尼耐药细胞的高效策略。
    Lazertinib, a novel third-generation epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI), demonstrates marked efficacy in EGFR-mutant lung cancer. However, resistance commonly develops, prompting consideration of therapeutic strategies to overcome initial drug resistance mechanisms. This study aimed to elucidate the adaptive resistance to lazertinib and advocate novel combination treatments that demonstrate efficacy in preventing resistance as a first-line treatment for EGFR mutation-positive NSCLC. We found that AXL knockdown significantly inhibited lung cancer cell viability in the presence of lazertinib, indicating that AXL activation contributes to lazertinib resistance. However, long-term culture with a combination of lazertinib and AXL inhibitors led to residual cell proliferation and increased the MCL-1 expression level, which was mediated by the nuclear translocation of the transcription factor YAP. Triple therapy with an MCL-1 or YAP inhibitor in combination with lazertinib and an AXL inhibitor significantly reduced cell viability and increased the apoptosis rate. These results demonstrate that AXL and YAP/MCL-1 signals contribute to adaptive lazertinib resistance in EGFR-mutant lung cancer cells, suggesting that the initial dual inhibition of AXL and YAP/MCL-1 might be a highly effective strategy in eliminating lazertinib-resistant cells.
    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
    病毒受体决定了病毒的组织嗜性,与病毒感染引起的临床结局有一定的关系,这对于识别病毒受体,了解病毒的感染机制和开发进入抑制剂具有重要意义。邻近标记(PL)是一种研究蛋白质-蛋白质相互作用的新技术,但它尚未应用于病毒受体或共受体的鉴定。这里,我们试图通过使用TurboID催化的PL来鉴定SARS-CoV-2的共受体。膜蛋白血管紧张素转换酶2(ACE2)用作诱饵并与TurboID缀合,构建了稳定表达ACE2-TurboID的A549细胞系。在生物素和ATP存在下,SARS-CoV-2假病毒与ACE2-TurboID稳定表达的细胞系孵育,这可以启动TurboID的催化活性,并用生物素标记相邻的内源性蛋白。随后,收获生物素化的蛋白质并通过质谱鉴定。我们鉴定了一种膜蛋白,AXL,已在功能上显示可介导SARS-CoV-2进入宿主细胞。我们的数据表明PL可用于鉴定病毒进入的共受体。
    Virus receptors determine the tissue tropism of viruses and have a certain relationship with the clinical outcomes caused by viral infection, which is of great importance for the identification of virus receptors to understand the infection mechanism of viruses and to develop entry inhibitor. Proximity labeling (PL) is a new technique for studying protein-protein interactions, but it has not yet been applied to the identification of virus receptors or co-receptors. Here, we attempt to identify co-receptor of SARS-CoV-2 by employing TurboID-catalyzed PL. The membrane protein angiotensin-converting enzyme 2 (ACE2) was employed as a bait and conjugated to TurboID, and a A549 cell line with stable expression of ACE2-TurboID was constructed. SARS-CoV-2 pseudovirus were incubated with ACE2-TurboID stably expressed cell lines in the presence of biotin and ATP, which could initiate the catalytic activity of TurboID and tag adjacent endogenous proteins with biotin. Subsequently, the biotinylated proteins were harvested and identified by mass spectrometry. We identified a membrane protein, AXL, that has been functionally shown to mediate SARS-CoV-2 entry into host cells. Our data suggest that PL could be used to identify co-receptors for virus entry.
    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
    三阴性乳腺癌(TNBC)是最具侵袭性的亚型,具有高转移和死亡率。鉴于缺乏可操作的靶标,如ER和HER2,TNBC仍然是一个未解决的治疗挑战。尽管有高CDK4/6表达水平,由于耐药的出现,在TNBC中抑制CDK4/6的功效受到限制。对CDK4/6抑制的抗性主要由RB1失活介导。由于我们的目标是克服对CDK4/6抑制的抗性,在这项研究中,我们主要使用不表达RB1的细胞系。在筛选CDK4/6抑制后的活化受体酪氨酸激酶(RTKs)后,我们确定了TAM(Tyro3,Axl,和MerTK)RTK是TNBC中至关重要的治疗漏洞。我们证明了用一种新的抑制剂靶向TAM受体,西拉巴替尼,TNBC对CDK4/6抑制剂显著敏感。在延长HER2抑制剂治疗后,HER2+乳腺癌抑制HER2表达,生理转化为TNBC样细胞。我们进一步表明,联合治疗对耐药HER2+乳腺癌也非常有效。在定量蛋白质组学和RNA-seq数据分析之后,我们将研究扩展到了TNBC的免疫分型。鉴于TAM受体在促进免疫抑制肿瘤微环境(TME)形成中的作用,我们进一步证明,CDK4/6抑制剂abemaciclib和sitravatinib的组合改变了TNBC的免疫格局,有利于免疫检查点阻断.总的来说,我们的研究提供了针对TNBC和潜在耐药HER2+乳腺癌的新型高效联合疗法,可迅速转移到临床.
    Triple-negative breast cancer (TNBC) is the most aggressive subtype with high metastasis and mortality rates. Given the lack of actionable targets such as ER and HER2, TNBC still remains an unmet therapeutic challenge. Despite harboring high CDK4/6 expression levels, the efficacy of CDK4/6 inhibition in TNBC has been limited due to the emergence of resistance. The resistance to CDK4/6 inhibition is mainly mediated by RB1 inactivation. Since our aim is to overcome resistance to CDK4/6 inhibition, in this study, we primarily used the cell lines that do not express RB1. Following a screening for activated receptor tyrosine kinases (RTKs) upon CDK4/6 inhibition, we identified the TAM (Tyro3, Axl, and MerTK) RTKs as a crucial therapeutic vulnerability in TNBC. We show that targeting the TAM receptors with a novel inhibitor, sitravatinib, significantly sensitizes TNBC to CDK4/6 inhibitors. Upon prolonged HER2 inhibitor treatment, HER2+ breast cancers suppress HER2 expression, physiologically transforming into TNBC-like cells. We further show that the combined treatment is highly effective against drug-resistant HER2+ breast cancer as well. Following quantitative proteomics and RNA-seq data analysis, we extended our study into the immunophenotyping of TNBC. Given the roles of the TAM receptors in promoting the creation of an immunosuppressive tumor microenvironment (TME), we further demonstrate that the combination of CDK4/6 inhibitor abemaciclib and sitravatinib modifies the immune landscape of TNBC to favor immune checkpoint blockade. Overall, our study offers a novel and highly effective combination therapy against TNBC and potentially treatment-resistant HER2+ breast cancer that can be rapidly moved to the clinic.
    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
    EGFR酪氨酸激酶抑制剂(TKI)耐药是EGFR突变非小细胞肺癌(NSCLC)治疗的主要挑战。我们之前的工作表明,AXL的过表达通过上皮-间质转化(EMT)促进EGFR-TKI耐药的NSCLC患者的一个子集。与埃罗替尼耐药和敏感细胞相比,RP11-874J12.4在厄洛替尼耐药的NSCLC细胞(HCC827-ER3)中上调。有趣的是,RP11-874J12.4的表达与AXL呈正相关。此外,RP11-874J12.4在体外促进NSCLC细胞增殖和转移。机械上,RP11-874J12.4通过miR-34a-5p海绵促进AXL表达,据报道抑制AXLmRNA的翻译。同时,RP11-874J12.4在肺癌肿瘤中的表达高于癌旁组织,RP11-874J12.4高表达的患者临床预后较差。RP11-874J12.4的高表达可能是厄洛替尼耐药NSCLC患者的生物标志物。这些发现揭示了对NSCLC埃罗替尼耐药机制的新见解。它可能是诊断和治疗非小细胞肺癌的一个有希望的目标。
    EGFR tyrosine kinase inhibitor (TKI) resistance is a major challenge for EGFR-mutant non-small cell lung cancer (NSCLC) treatment. Our previous work revealed that overexpression of AXL promoted EGFR-TKI resistance through epithelial-mesenchymal transition (EMT) in a subset of NSCLC patients. Compared with erlotinib resistant and sensitive cells, RP11-874 J12.4 was upregulated in erlotinib-resistant NSCLC cells (HCC827-ER3). Interestingly, the expression of RP11-874 J12.4 positively correlated with AXL. Besides, RP11-874 J12.4 promotes NSCLC cell proliferation and metastasis in vitro. Mechanistically, RP11-874 J12.4 promoted AXL expression through sponge with miR-34a-5p, which was reported to inhibit the translation of AXL mRNA. Meanwhile, the expression of RP11-874 J12.4 in lung cancer tumors were higher than the adjacent tissue, and those patients with high expression of RP11-874 J12.4 showed a poor prognosis in clinical. High expression of RP11-874 J12.4 might be a biomarker for NSCLC patients with erlotinib resistance. These findings reveal a novel insight into the mechanism of erlotinib resistance in NSCLC, and it might be a promising target for the diagnosis and treatment of NSCLC.
    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
    由心室重构和功能障碍(例如心肌梗死后(MI))引起的心脏纤维化可导致心力衰竭。RNAN6-甲基腺苷(m6A)甲基化已被证明在许多疾病的发生和发展中起关键作用。在研究m6A阅读器YTHDF1在心脏纤维化中的生物学功能时,使用腺相关病毒9在小鼠心脏中敲除或过表达YTHDF1基因,并进行体内MI手术和体外转化生长因子-β(TGF-β)激活的心肌成纤维细胞建立纤维化模型。我们的结果表明,在小鼠心脏中沉默YTHDF1可以显着恢复受损的心功能并减轻心肌纤维化,而YTHDF1过表达可进一步增强心功能不全,加重心室病理重构和纤维化的发生。机械上,锌指BED型含有6个介导的YTHDF1基因启动子的转录功能。YTHDF1增强AXL翻译并激活TGF-β-Smad2/3信号通路,从而加重心功能不全和心肌纤维化的发生发展。始终如一,我们的数据表明YTHDF1参与激活,扩散,和迁移参与心脏纤维化的体外研究。我们的结果表明,YTHDF1可以作为心肌纤维化的潜在治疗靶标。
    Cardiac fibrosis caused by ventricular remodeling and dysfunction such as post-myocardial infarction (MI) can lead to heart failure. RNA N6-methyladenosine (m6A) methylation has been shown to play a pivotal role in the occurrence and development of many illnesses. In investigating the biological function of the m6A reader YTHDF1 in cardiac fibrosis, adeno-associated virus 9 was used to knock down or overexpress the YTHDF1 gene in mouse hearts, and MI surgery in vivo and transforming growth factor-β (TGF-β)-activated cardiac fibroblasts in vitro were performed to establish fibrosis models. Our results demonstrated that silencing YTHDF1 in mouse hearts can significantly restore impaired cardiac function and attenuate myocardial fibrosis, whereas YTHDF1 overexpression could further enhance cardiac dysfunction and aggravate the occurrence of ventricular pathological remodeling and fibrotic development. Mechanistically, zinc finger BED-type containing 6 mediated the transcriptional function of the YTHDF1 gene promoter. YTHDF1 augmented AXL translation and activated the TGF-β-Smad2/3 signaling pathway, thereby aggravating the occurrence and development of cardiac dysfunction and myocardial fibrosis. Consistently, our data indicated that YTHDF1 was involved in activation, proliferation, and migration to participate in cardiac fibrosis in vitro. Our results revealed that YTHDF1 could serve as a potential therapeutic target for myocardial fibrosis.
    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
    严重性,症状,COVID-19的结果被认为与病毒如何进入宿主细胞密切相关。该过程涉及血管紧张素转换酶2(ACE2)和酪氨酸蛋白激酶受体UFO(AXL)受体的关键作用。然而,关于ACE2和AXL的循环水平及其在COVID-19中的意义的研究有限。
    71个未感染个体的对照组也包括在研究中。根据《2019年冠状病毒病指南》(第10版),358例COVID-19患者被分为非重症病例和重症病例。采用酶联免疫吸附试验(ELISA)检测COVID-19感染后不同时间点血清ACE2/AXL水平,包括第0-7、8-15、31-179天和>180天。使用iFlash3000化学发光免疫分析仪评估COVID-19患者相同间隔的血清SARS-CoV-2IgG/IgM抗体。受试者工作特征(ROC)曲线用于评估生物标志物的诊断价值,并探讨了实验室参数与疾病进展之间的关联.
    与未感染组相比,COVID-19组的ACE2和AXL水平降低,SARS-COV-2IgG水平升高。AXL(AUC=0.774)对COVID-19的预测能力强于ACE2。在感染后的第一周,只有AXL水平在重度组和非重度组之间有统计学差异.第一周后,两组的ACE2和AXL水平存在差异。此外,在严重的COVID-19病例中,血清ACE2,AXL,SARS-COV-2IgM水平在第8-15天达到峰值,然后下降,而血清SARS-COV-2IgG水平持续上升,在下降前31-180天达到峰值。此外,与未感染组相比,感染组180天后AXL水平持续下降,SARS-COV-2IgG水平持续升高.
    血清ACE2和AXL水平与COVID-19严重程度相关。然而,AXL还可以在感染后的第一周提供临床恶化的早期预警。AXL似乎是预测COVID-19进展的潜在分子标志物。
    UNASSIGNED: The severity, symptoms, and outcome of COVID-19 is thought to be closely linked to how the virus enters host cells. This process involves the key roles of angiotensin-converting enzyme 2 (ACE2) and the Tyrosine protein kinase receptor UFO (AXL) receptors. However, there is limited research on the circulating levels of ACE2 and AXL and their implications in COVID-19.
    UNASSIGNED: A control group of 71 uninfected individuals was also included in the study. According to the Guidance for Corona Virus Disease 2019 (10th edition), a cohort of 358 COVID-19 patients were categorized into non-severe and severe cases. Serum ACE2/AXL levels in COVID-19 patients were detected by enzyme-linked immunosorbent assay (ELISA) at different time points post-COVID-19 infection, including days 0-7, 8-15, 31-179 and >180 days. Serum SARS-CoV-2 IgG/IgM antibodies in COVID-19 patients at the same intervals were assessed by using an iFlash 3000 Chemiluminescence Immunoassay Analyzer. The receiver operating characteristic (ROC) curves were used to assess the diagnostic value of the biological markers, and the association between laboratory parameters and illness progression were explored.
    UNASSIGNED: Compared with the uninfected group, the levels of ACE2 and AXL in the COVID-19 group were decreased, and the SARS-COV-2 IgG level was increased. AXL (AUC = 0.774) demonstrated a stronger predictive ability for COVID-19 than ACE2. In the first week after infection, only the level of AXL was statistically different between severe group and non-severe group. After first week, the levels of ACE2 and AXL were different in two groups. Moreover, in severe COVID-19 cases, the serum ACE2, AXL, and SARS-COV-2 IgM levels reached a peak during days 8-15 before declining, whereas serum SARS-COV-2 IgG levels continued to rise, reaching a peak at day 31-180 days before decreasing. In addition, the AXL level continued to decrease and the SARS-COV-2 IgG level continued to increase in the infected group after 180 days compared to the uninfected group.
    UNASSIGNED: The levels of serum ACE2 and AXL correlate with COVID-19 severity. However, AXL can also provide early warning of clinical deterioration in the first week after infection. AXL appears to be a superior potential molecular marker for predicting COVID-19 progression.
    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
    原理:AXL表达已被鉴定为急性髓细胞性白血病(AML)的预后因子,并且在约50%的AML患者中可检测到。在这项研究中,我们开发了AXL特异性单域抗体(sdAb),小鼠和人AXL蛋白的交叉反应,非侵入性成像和治疗表达AXL的癌细胞。方法:用小鼠和人AXL蛋白免疫羊驼诱导AXL特异性sdAb。使用ELISA对SdAb进行表征,流式细胞术,表面等离子体共振和AlphaFold2软件。选择先导化合物并用99mTc标记,用于使用SPECT/CT成像在人(THP-1细胞)或小鼠(C1498细胞)AML的小鼠模型中评价为诊断工具。为了治疗目的,将先导化合物与小鼠IgG2a-Fc尾融合,并进行体外功能测试,包括活力,人AML细胞系和原发性患者样本中的凋亡和增殖测定。使用这些体外模型,作为单一药物评估其抗肿瘤效果,并与标准护理药物维奈托克或阿糖胞苷联合使用。结果:基于其细胞结合潜能,交叉反应性,纳摩尔亲和力和GAS6/AXL阻断能力,我们选择sdAb20进行进一步评估。使用SPECT/CT成像,我们观察到99mTc-sdAb20在AXL阳性THP-1或C1498肿瘤小鼠中的肿瘤摄取。在THP-1异种移植物中,需要使用预注射冷sdAb20-Fc的优化方案来最大化肿瘤-背景信号。除了它的诊断价值,我们在体外使用sdAb20-Fc观察到肿瘤细胞增殖和活力的显著降低。此外,联合sdAb20-Fc和阿糖胞苷协同诱导人AML细胞系凋亡,而与维奈托克合用时,这些影响不太明显。结论:由于它们的诊断潜力,sdAb可用于筛选符合AXL靶向治疗条件的患者,并在治疗和疾病进展期间随访AXL表达。当与Fc结构域融合时,sdAb获得额外的治疗特性,可以导致治疗AXL阳性癌症患者的多药物方法。
    Rationale: AXL expression has been identified as a prognostic factor in acute myeloid leukemia (AML) and is detectable in approximately 50% of AML patients. In this study, we developed AXL-specific single domain antibodies (sdAbs), cross-reactive for both mouse and human AXL protein, to non-invasively image and treat AXL-expressing cancer cells. Methods: AXL-specific sdAbs were induced by immunizing an alpaca with mouse and human AXL proteins. SdAbs were characterized using ELISA, flow cytometry, surface plasmon resonance and the AlphaFold2 software. A lead compound was selected and labeled with 99mTc for evaluation as a diagnostic tool in mouse models of human (THP-1 cells) or mouse (C1498 cells) AML using SPECT/CT imaging. For therapeutic purposes, the lead compound was fused to a mouse IgG2a-Fc tail and in vitro functionality tests were performed including viability, apoptosis and proliferation assays in human AML cell lines and primary patient samples. Using these in vitro models, its anti-tumor effect was evaluated as a single agent, and in combination with standard of care agents venetoclax or cytarabine. Results: Based on its cell binding potential, cross-reactivity, nanomolar affinity and GAS6/AXL blocking capacity, we selected sdAb20 for further evaluation. Using SPECT/CT imaging, we observed tumor uptake of 99mTc-sdAb20 in mice with AXL-positive THP-1 or C1498 tumors. In THP-1 xenografts, an optimized protocol using pre-injection of cold sdAb20-Fc was required to maximize the tumor-to-background signal. Besides its diagnostic value, we observed a significant reduction in tumor cell proliferation and viability using sdAb20-Fc in vitro. Moreover, combining sdAb20-Fc and cytarabine synergistically induced apoptosis in human AML cell lines, while these effects were less clear when combined with venetoclax. Conclusions: Because of their diagnostic potential, sdAbs could be used to screen patients eligible for AXL-targeted therapy and to follow-up AXL expression during treatment and disease progression. When fused to an Fc-domain, sdAbs acquire additional therapeutic properties that can lead to a multidrug approach for the treatment of AXL-positive cancer patients.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号