tyrosine kinase

酪氨酸激酶
  • 文章类型: Journal Article
    以碘喹唑啉支架为基础合成了表皮生长因子受体(EGFR)T790M/血管内皮生长因子受体-2(VEGFR-2)的新型抑制剂,芳香,和/或脂族部分。在体外检查了新型衍生物对A549,HCT116,密歇根州癌症基金会-7(MCF-7)的抗癌活性,和HepG2细胞。应用分子建模来发现它们与VEGFR-2和EGFR活性位点两者的结合方向。化合物8d,8c,6d,和6c显示最高的细胞毒性,IC50=6.00、6.90、6.12和6.24µM,7.05、7.35、6.80和6.80µM,5.75、7.50、6.90和6.95µM,和6.55,7.88,7.44和7.10µM对A549,HepG2,HCT116和MCF-7细胞系,相应地。评价了极具活性的八种化合物6a-d和8a-d对正常VERO(正常非洲绿猴肾细胞)的细胞毒性。我们的化合物对正常VERO细胞表现出低毒性,IC50=45.66-51.83μM。此外,对所有化合物进行EGFRT790M和VEGFR-2酶的抑制试验.化合物6d对EGFRT790M活性有显著的抑制作用,8d,6c,和8c,IC50分别为0.35、0.42、0.48和0.50µM。此外,化合物8d对VEGFR-2活性有显著抑制作用,8c,6d,和6c,IC50分别为0.92、0.95、1.00和1.20µM。按计划,导数6d,8d,6c,和8c表现出对EGFRT790M/VEGFR-2两种活性的异常抑制。最后,在硅吸收中,分布,新陈代谢,对高活性的四种化合物6c进行了排泄和毒性(ADMET)研究,6d,8c,和8d比较,以厄洛替尼和索拉非尼为参考标准。
    Novel inhibitors of epidermal growth factor receptor (EGFR)T790M/vascular endothelial growth factor receptor-2 (VEGFR-2) were synthesized based on the iodoquinazoline scaffold linked to different heteroaromatic, aromatic, and/or aliphatic moieties. The novel derivatives were in vitro examined for anticancer activities against A549, HCT116, michigan cancer foundation-7 (MCF-7), and HepG2 cells. Molecular modeling was applied to discover their orientation of binding with both VEGFR-2 and EGFR active sites. Compounds 8d, 8c, 6d, and 6c indicated the highest cytotoxicity with IC50 = 6.00, 6.90, 6.12 and 6.24 µM, 7.05, 7.35, 6.80, and 6.80 µM, 5.75, 7.50, 6.90, and 6.95 µM, and 6.55, 7.88, 7.44, and 7.10 µM against the A549, HepG2, HCT116, and MCF-7 cell lines, correspondingly. The cytotoxicity against normal VERO (normal african green monkey kidney cells) of the extremely active eight compounds 6a-d and 8a-d was evaluated. Our compounds exhibited low toxicity concerning normal VERO cells with IC50 = 45.66-51.83 μM. Furthermore, inhibition assays for both the EGFRT790M and VEGFR-2 enzymes were done for all compounds. Remarkable inhibition of EGFRT790M activity was achieved with compounds 6d, 8d, 6c, and 8c at IC50 = 0.35, 0.42, 0.48, and 0.50 µM correspondingly. Moreover, remarkable inhibition of VEGFR-2 activity was achieved with compounds 8d, 8c, 6d, and 6c at IC50 = 0.92, 0.95, 1.00, and 1.20 µM correspondingly. As planned, derivatives 6d, 8d, 6c, and 8c presented exceptional inhibition of both EGFRT790M/VEGFR-2 activities. Finally, in silico absorption, distribution, metabolism, excretion and toxicity (ADMET) studies were made for the highly active four compounds 6c, 6d, 8c, and 8d in comparison with erlotinib and sorafenib as reference standards.
    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
    皮肤鳞状细胞癌(cSCC)是一种经常在狗中诊断的肿瘤类型。然而,研究集中在进一步研究其分子生物学,主要是帮助实施新疗法的生物标志物,仍然是文学中的恐慌。因此,本文评估了呈现不同细胞分化程度的犬cSCC中的免疫染色和表皮生长因子受体(HER1和HER2)的基因表达。选择了32(32)只犬cSCC,根据其细胞分化程度进行分类,并进行免疫组织化学研究以评估HER1和HER2免疫染色的强度和分布。此外,通过实时PCR研究HER1和HER2基因表达。在两种标记中均观察到膜和细胞质免疫染色。HER2在低分化的cSCC中普遍存在;两种标志物之间存在正蛋白表达相关性。平均HER1基因表达在中等分化中更高,而低分化cSCC中平均HER2基因表达较高。此外,标记之间存在基因表达相关性,无论细胞分化程度。因此,低分化犬cSCC中HER2蛋白免疫染色和基因表达较高,并且能够理解在这种表皮生长因子受体中观察到的增加与犬物种中这种肿瘤的细胞分化程度成正比。本研究的结果有助于更好地理解犬cSCC的分子生物学;然而,它是相关的研究其他标记,旨在研究信号通路。
    Cutaneous squamous cell carcinoma (cSCC) is a neoplasm type often diagnosed in dogs. However, studies focused on further investigating its molecular biology, mainly biomarkers to help implementing new therapies, remain scare in the literature. Thus, immunostaining and the gene expression of epidermal growth factor receptors (HER1 and HER2) in canine cSCC presenting different cell differentiation degrees were herein assessed. Thirty-two (32) canine cSCC were selected, classified based on to their cell differentiation degree and subjected to immunohistochemical study to assess HER1 and HER2 immunostaining intensity and distribution. In addition, HER1 and HER2 gene expression was investigated through real-time PCR. Membranous and cytoplasmic immunostaining were observed in both markers. HER2 prevailed in poorly differentiated cSCC; there was positive protein expression correlation between both markers. Mean HER1 gene expression was higher in moderately differentiated, whereas mean HER2 gene expression was higher in poorly differentiated cSCC. Moreover, there was gene expression correlation between markers, regardless of cell differentiation degree. Thus, HER2 protein immunostaining and gene expression were higher in poorly differentiated canine cSCC and it enabled understanding that increase observed in this epidermal growth factor receptor is proportional to this neoplasm\'s cell differentiation degree in canine species. Results in the current study helped better understanding canine cSCC\'s molecular biology; however, it is relevant studying other markers aiming to investigate signaling pathways.
    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
    肝细胞癌(HCC)是一种高度未满足的医疗需求的疾病,已成为全球性的健康问题。对HCC发病机制的不完全理解和相关临床前动物模型的数量有限,阻碍了HCC靶向治疗的发展。我们最近公布了一个以前未表征的YES激酶(由YES1编码)在HCC中依赖的致癌信号通路。为了对肝癌的这个子集进行建模,我们从表达激活的人YES的转基因小鼠的肝肿瘤中建立了一系列同系细胞系。所产生的细胞系(称为HepYF)被富集用于表达干细胞和祖细胞标志物。迅速扩散,具有较高的SRC家族激酶(SFK)活性和激活的有丝分裂信号通路。转录组学分析表明,HepYF细胞是HCC最具侵袭性的增殖类别G3亚组的代表。HepYF细胞在原位植入同源宿主后形成快速生长的转移性肿瘤。索拉非尼或SFK抑制剂达沙替尼治疗显著抑制HepYF肿瘤的生长。新的HepYFHCC细胞系提供了相关的临床前模型来研究HCC的发病机理,并测试新的小分子抑制剂和免疫治疗方法。
    Hepatocellular carcinoma (HCC) is a disease of high unmet medical need that has become a global health problem. The development of targeted therapies for HCC has been hindered by the incomplete understanding of HCC pathogenesis and the limited number of relevant preclinical animal models. We recently unveiled a previously uncharacterized YES kinase (encoded by YES1)-dependent oncogenic signaling pathway in HCC. To model this subset of HCC, we established a series of syngeneic cell lines from liver tumors of transgenic mice expressing activated human YES. The resulting cell lines (referred to as HepYF) were enriched for expression of stem cell and progenitor markers, proliferated rapidly, and were characterized by high SRC family kinase (SFK) activity and activated mitogenic signaling pathways. Transcriptomic analysis indicated that HepYF cells are representative of the most aggressive proliferation class G3 subgroup of HCC. HepYF cells formed rapidly growing metastatic tumors upon orthotopic implantation into syngeneic hosts. Treatment with sorafenib or the SFK inhibitor dasatinib markedly inhibited the growth of HepYF tumors. The new HepYF HCC cell lines provide relevant preclinical models to study the pathogenesis of HCC and test novel small-molecule inhibitor and immunotherapy approaches.
    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
    受体酪氨酸激酶(RTKs)的激酶结构域是高度保守的,然而,他们能够区分潜在的底物,以选择性地激活下游信号通路。在这项研究中,我们通过创建两个系列的嵌合RTK来测试催化域特异性的重要性。在一组中,胰岛素样生长因子I受体(IGF1R)的激酶结构域被胰岛素受体(IR)的激酶结构域取代,巨噬细胞刺激蛋白1受体/Ron(Ron)或Src。在另一组嵌合体中,表皮生长因子受体(EGFR)的激酶结构域同样被IR的激酶结构域所取代,罗恩,或Src。我们在哺乳动物细胞中表达了受体的野生型和嵌合形式。对于某些信令事件,例如识别IRS1,酪氨酸激酶催化结构域的身份似乎并不重要。相比之下,识别一些网站,如EGFR上的C末端自磷酸化位点,确实依赖于激酶结构域的身份。我们的数据还表明,当天然激酶结构域被Src取代时,配体依赖性丧失。这表明激酶结构域的身份对于适当的受体调节可能很重要。总的来说,结果与RTK信号的保真度取决于与底物的共定位和靶向的想法一致,以及激酶结构域的内在特异性。
    The kinase domains of receptor tyrosine kinases (RTKs) are highly conserved, yet they are able to discriminate among potential substrates to selectively activate downstream signaling pathways. In this study, we tested the importance of catalytic domain specificity by creating two series of chimeric RTKs. In one set, the kinase domain of insulin-like growth factor I receptor (IGF1R) was replaced by the kinase domains from insulin receptor (IR), macrophage stimulating protein 1 receptor/Ron (Ron) or Src. In the other set of chimeras, the kinase domain of epidermal growth factor receptor (EGFR) was similarly replaced by the kinase domains of IR, Ron, or Src. We expressed the wild-type and chimeric forms of the receptors in mammalian cells. For some signaling events, such as recognition of IRS1, the identity of the tyrosine kinase catalytic domain did not appear to be crucial. In contrast, recognition of some sites, such as the C-terminal autophosphorylation sites on EGFR, did depend on the identity of the kinase domain. Our data also showed that ligand dependence was lost when the native kinase domains were replaced by Src, suggesting that the identity of the kinase domains could be important for proper receptor regulation. Overall, the results are consistent with the idea that the fidelity of RTK signaling depends on co-localization and targeting with substrates, as well as on the intrinsic specificity of the kinase domain.
    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:复合粘着斑激酶(FAK)/Src和桩蛋白似乎在癌症的发病和进展中起关键作用。这项研究的目的是评估这些蛋白质在肾细胞癌(RCC)中的表达,考虑到免疫反应性评分(IRS),积极性和强度,并发现与患者临床特征的任何关联,组织学类型和其他病理特征暗示FAK/Src和桩蛋白在RCC中可能具有病理生理或预后作用。
    方法:2009年1月至2010年9月接受部分或根治性肾切除术的RCC患者符合这项回顾性横断面研究的条件。FAK的免疫组化表达,与各种临床病理特征相关,分析了福尔马林固定的石蜡包埋的肿瘤组织中的Src和paxillin蛋白。
    结果:在90名患者中,58例肾透明细胞癌,15有乳头状,11例具有发色性细胞,6例具有未分类的RCC。FAK,Src和paxillin表达率为55.6%,所有病例的32.2%和18.9%,分别。在单变量分析中,与透明细胞RCC相比,乳头状和发色组织学类型的患者更可能出现FAK阳性和IRS(p<0.005),Src阳性和IRS在T3期与T1期(p<0.005)中更常见,而在T3期与T2期(p=0.021)和3-4级与2级(p=0.013)的患者中,桩蛋白阳性的可能性更高。Paxillin-IRS与任何临床病理特征无关。在FAK和Src阳性和IRS的多因素分析中也再现了相同的关联,虽然发现paxillin阳性和IRS与女性相关(p=0.052,p=0.024),3-4年级高于2年级(p=0.022,p=0.020)。
    结论:我们的研究表明RCC显示FAK的免疫组织化学表达,Src和桩蛋白,这种表达与组织学类型有关,舞台和舞台/年级/性别,分别。这些发现暗示FAK/Src信号通路可能参与RCC中癌症的发病机制和进展。为抑制剂靶向治疗提供未来前景。
    OBJECTIVE: The complex focal adhesion kinase (FAK)/Src and paxillin seem to play a key role in the pathogenesis and progression of cancer. The aim of this study is to evaluate the expression of these proteins in renal cell carcinomas (RCCs), considering the immunoreactive score (IRS), the positivity and the intensity, and to find any association with patients\' clinical characteristics, histologic type and other pathological features that imply a possible pathophysiological or prognostic role of FAK/Src and paxillin in RCC.
    METHODS: Patients with RCC who had undergone partial or radical nephrectomy from January 2009 to September 2010 were eligible for this retrospective cross-sectional study. The immunohistochemical expression of FAK, Src and paxillin proteins in formalin-fixed paraffin-embedded tumour tissue was analysed in association with various clinicopathological features.
    RESULTS: Out of ninety patients, 58 had clear cell renal carcinoma, 15 had papillary, 11 had chromophobe and six had unclassified RCC. FAK, Src and paxillin were expressed in 55.6%, 32.2% and 18.9% of all cases, respectively. In univariate analysis, FAK positivity and IRS were more likely in patients with papillary and chromophobe histologic type versus clear cell RCC (p<0.005), Src positivity and IRS presented more frequently in stage T3 versus T1 (p<0.005) and paxillin positivity was more likely in patients with stage T3 versus T2 (p=0.021) and grades 3-4 versus grade 2 (p=0.013). Paxillin-IRS was not associated with any clinicopathological features. The same associations were also reproduced in the multifactorial analysis for the FAK and Src positivity and IRS, while it was found that paxillin positivity and IRS were associated with the female gender (p=0.052, p=0.024), and were higher in grades 3-4 versus grade 2 (p=0.022, p=0.020).
    CONCLUSIONS: Our study suggests that RCC shows immunohistochemical expression of FAK, Src and paxillin proteins, and this expression varies in relation to the histologic type, the stage and the stage/grade/gender, respectively. These findings imply a possible involvement of the FAK/Src signalling pathway in the pathogenesis and progression of cancer in RCC, providing future perspectives for targeted therapies with inhibitors.
    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
    宫颈癌占女性因癌症死亡的大多数,主要在发展中国家。这种疾病背后的罪魁祸首是人乳头瘤病毒(HPV),占宫颈癌病例的90%以上。病毒株产生的蛋白质有利于细胞凋亡过程的敲除和子宫颈细胞的持续生长,从而导致肿瘤生长。促血管生成生长因子,如成纤维细胞生长因子(FGF),血管内皮生长因子(VEGF),血管生成素,和其他内皮生长因子(EGF),由肿瘤细胞和周围的微环境分泌,这进一步促进了癌症的发展。受体酪氨酸激酶的胞外结构域被配体(如VEGF和EGF)用于通过诱导受体二聚化来接合和激活它们,这有利于这些因素的级联影响。每个受体的酪氨酸激酶结构域相互磷酸化,激活受体并启动促进血管生成的信号级联,迁移,扩散,和内皮细胞的存活。癌细胞受益于其修饰的信号通路,导致致癌激活。在早期宫颈癌检测中,二线治疗策略涉及用VEGF和小分子酪氨酸激酶抑制剂(TKIs)阻断信号通路.这篇综述论文通过深入研究目前可用的抑制剂的细节,重点介绍了宫颈癌的起源和使用VEGF和酪氨酸激酶抑制剂来对抗它。Further,我们已经讨论了目前处于不同临床试验阶段的抑制剂分子。本文必将增进对宫颈癌及其治疗方法的了解,以及可以采取哪些进一步的干预措施来减轻目前作为发展中国家主要健康负担的疾病。
    Cervical cancer accounts for most deaths due to cancer in women, majorly in developing nations. The culprit behind this disease is the human papillomavirus (HPV) which accounts for more than 90% of cervical cancer cases. The viral strains produce proteins that favor the knocking down of the apoptosis process and continuous growth of cells in the cervix leading to tumor growth. Proangiogenic growth factors, such as fibroblast growth factor (FGF), vascular endothelial growth factor (VEGF), angiopoietins, and other endothelial growth factors (EGF), are secreted by tumor cells and the surrounding microenvironment, which further advances the development of cancer. The extracellular domain of receptor tyrosine kinases is employed by ligands (like VEGF and EGF) to engage and activate them by inducing receptor dimerization, which facilitates the cascade impact of these factors. The tyrosine kinase domains of each receptor autophosphorylate each other, activating the receptor and initiating signaling cascades that promote angiogenesis, migration, proliferation, and survival of endothelial cells. Cancer cells benefit from its modified signaling pathways, which cause oncogenic activation. Upon early cervical cancer detection, the second-line therapy strategy involves blocking the signaling pathways with VEGF and small molecule tyrosine kinase inhibitors (TKIs). This review paper highlights the genesis of cervical cancer and combating it using VEGF and tyrosine kinase inhibitors by delving into the details of the currently available inhibitors. Further, we have discussed the inhibitor molecules that are currently in various phases of clinical trials. This paper will surely enhance the understanding of cervical cancer and its treatment approaches and what further interventions can be done to alleviate the disease currently serving as a major health burden in the developing world.
    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
    海洋环境提供了含有潜在革命性生物活性化学物质的独特生物的丰富来源。这些生物中的一种是大白菜,一种被称为绿海藻的绿藻,Seagrapes,或者绿色鱼子酱.这种生物之所以脱颖而出,是因为它在医学上有很大的应用前景,特别是在癌症的研究中。通过利用计算模型(计算机模拟)和细胞实验室实验(体外),对绿色海藻中的化学成分进行了有效的分析,揭示其治疗非小细胞肺癌(NSCLC)的能力。这项研究特别强调阻断SRC,STAT3,PIK3CA,MAPK1,EGFR,和JAK1使用分子对接和体外。这些蛋白质在NSCLC的EGFR酪氨酸激酶抑制剂耐药途径中起着至关重要的作用。C.消旋C.racemosa提取物(CRE)中包含的化学Caulersin(C2)已被确定为对抗非小细胞肺癌(NSCLC)的有效药物,在计算机和体外。CRE和C2显示类似于奥希替尼(阳性对照/NSCLC药物)的抑制水平。
    The marine environment provides a rich source of distinct creatures containing potentially revolutionary bioactive chemicals. One of these organisms is Caulerpa racemosa, a type of green algae known as green seaweed, seagrapes, or green caviar. This organism stands out because it has great promise for use in medicine, especially in the study of cancer. Through the utilization of computational modeling (in silico) and cellular laboratory experiments (in vitro), the chemical components included in the green seaweed C. racemosa were effectively analyzed, uncovering its capability to treat non-small cell lung cancer (NSCLC). The study specifically emphasized blocking SRC, STAT3, PIK3CA, MAPK1, EGFR, and JAK1 using molecular docking and in vitro. These proteins play a crucial role in the EGFR Tyrosine Kinase Inhibitor Resistance pathway in NSCLC. The chemical Caulersin (C2) included in C. racemosa extract (CRE) has been identified as a potent and effective agent in fighting against non-small cell lung cancer (NSCLC), both in silico and in vitro. CRE and C2 showed a level of inhibition similar to that of osimertinib (positive control/NSCLC drug).
    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
    造血细胞激酶(Hck),属于Src激酶家族的非受体酪氨酸激酶,与许多人类疾病的发病机理有着错综复杂的联系,与癌症有特别明显的关联。Hck不仅直接影响扩散,迁移,和癌细胞的凋亡,但也与JAK/STAT相互作用,MEK/ERK,PI3K/AKT,CXCL12/CXCR4等途径。Hck还影响肿瘤微环境以促进癌症的发作和进展。本文探讨了Hck在多种实体瘤中的功能作用和调控机制。此外,它探讨了Hck在血液恶性肿瘤中的意义。这篇综述总结了Hck抑制剂的研究现状,其中大多数处于研究的临床前阶段。值得注意的是,这些抑制剂主要用于白血病的治疗管理,它们的组合潜力为未来的研究指明了有希望的途径。总之,这篇综述强调了Hck在实体瘤中的作用机制的意义。这种见解对于理解有关Hck的当前研究趋势至关重要:针对Hck的靶向治疗在恶性肿瘤的诊断和治疗中显示出巨大的前景。进一步研究Hck在癌症中的作用,再加上特异性抑制剂的开发,有可能彻底改变癌症治疗方法。
    Hematopoietic cell kinase (Hck), a non-receptor tyrosine kinase belonging to the Src kinase family, is intricately linked to the pathogenesis of numerous human diseases, with a particularly pronounced association with cancer. Hck not only directly impacts the proliferation, migration, and apoptosis of cancer cells but also interacts with JAK/STAT, MEK/ERK, PI3K/AKT, CXCL12/CXCR4, and other pathways. Hck also influences the tumor microenvironment to facilitate the onset and progression of cancer. This paper delves into the functional role and regulatory mechanisms of Hck in various solid tumors. Additionally, it explores the implications of Hck in hematological malignancies. The review culminates with a summary of the current research status of Hck inhibitors, the majority of which are in the pre-clinical phase of investigation. Notably, these inhibitors are predominantly utilized in the therapeutic management of leukemia, with their combinatorial potential indicating promising avenues for future research. In conclusion, this review underscores the significance of the mechanism of Hck in solid tumors. This insight is crucial for comprehending the current research trends regarding Hck: targeted therapy against Hck shows great promise in both diagnosis and treatment of malignant tumors. Further investigation into the role of Hck in cancer, coupled with the development of specific inhibitors, has the potential to revolutionize approaches to cancer treatment.
    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
    本研究旨在探讨干扰素α-β受体亚基2(IFNAR2)和酪氨酸激酶2(TYK2)表达的可能预后意义。
    我们进行了一项包括COVID-19成年患者的回顾性研究。在任何干预之前收集所有血液样品。采用实时PCR技术检测54例和56例对照静脉血中IFNAR2和TYK2的表达。使用Delta-Ct方法评估IFNAR2和TYK2基因的转录量。
    我们的发现表明,需要氧气(O2)治疗的患者和不需要氧气(O2)治疗的患者之间IFNAR2和TYK2的基因表达水平没有显着差异(分别为p值=0.732和p值=0.629)。同样,IFNAR2和TYK2表达在住院少于7天的患者和住院7天或以上的患者之间没有显著差异(分别为p值=0.455和p值=0.626).我们还观察到IFNAR2表达与CRP之间的弱相关性(p值=0.045,r=0.192)。IFNAR2和TYK2转录本在COVID-19患者中的表达水平呈负相关(p值=0.044;偏相关系数=-0.283)。此外,与健康受试者相比,COVID-19组的IFNAR2和TYK2显著下调(p值分别=0.002和p值=0.028)。然而,根据COVID-19的严重程度,病例亚组之间的IFNAR2和TYK2表达均无显著差异。IFNAR2ΔΔCt(B=-0.184,95%CI:-0.524-0.157,p值=0.275)和TYK2ΔΔCt(B=0.114,95%CI:-0.268-0.496,p值=0.543)未发现是住院时间的显着预测因素。IFNAR2表达的曲线下面积(AUC)为0.655(p值=0.005,95%CI:0.554-0.757),表明它的判别价值差。
    我们无法明确评论IFNAR2和TYK2表达在COVID-19患者中的预后能力,需要更大规模的研究。这项研究的主要局限性包括缺乏纵向分析和样本量有限。
    UNASSIGNED: This study aimed to investigate the possible prognostic significance of interferon alpha-beta receptor subunit 2 (IFNAR2) and tyrosine kinase 2 (TYK2) expressions.
    UNASSIGNED: We conducted a retrospective study including COVID-19 adult patients. All blood samples were collected before any interventions. The expressions of IFNAR2 and TYK2 were assessed using real-time PCR in venous blood samples of 54 cases and 56 controls. The transcript quantities of IFNAR2 and TYK2 genes were assessed using a Delta-Ct method.
    UNASSIGNED: Our findings show no significant differences in gene expression levels for IFNAR2 and TYK2 between patients who required oxygen (O2) therapy and those who did not (p-value = 0.732 and p-value = 0.629, respectively). Likewise, there were no significant differences in IFNAR2 and TYK2 expressions between patients hospitalized for less than 7 days and those hospitalized for 7 days or more (p-value = 0.455 and p-value = 0.626, respectively). We also observed a weak correlation between IFNAR2 expression and CRP (p-value = 0.045, r = 0.192). There was a negative correlation between the expression levels of IFNAR2 and TYK2 transcripts in COVID-19 patients (p-value = 0.044; partial correlation coefficient = -0.283). Additionally, IFNAR2 and TYK2 were significantly downregulated in the COVID-19 group compared to healthy subjects (p-value = 0.002 and p-value = 0.028, respectively). However, neither IFNAR2 nor TYK2 expression was significantly different between the case subgroups based on COVID-19 severity. The IFNAR2 ΔΔCt (B = -0.184, 95% CI: -0.524-0.157, p-value = 0.275) and the TYK2 ΔΔCt (B = 0.114, 95% CI: -0.268-0.496, p-value = 0.543) were not found to be significant predictors of hospitalization duration. The area under the curve (AUC) for IFNAR2 expression is 0.655 (p-value = 0.005, 95% CI: 0.554-0.757), suggesting its poor discriminative value.
    UNASSIGNED: We were unable to comment definitively on the prognostic power of IFNAR2 and TYK2 expressions in COVID-19 patients, and larger-scale studies are needed. The principal limitations of this study included the lack of longitudinal analysis and limited sample size.
    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
    白细胞介素-6(IL-6)类炎性细胞因子通过Janus酪氨酸激酶(JAK)/信号转导和转录激活因子(STAT)通路而促进胰腺导管腺癌(PDAC)的发生发展;在这个过程中,特定的细胞内信号介质的功能不太明确。在成年小鼠中使用配体控制和胰腺特异性敲除,我们在这项研究中证明,JAK1缺乏阻止KRASG12D诱导的胰腺肿瘤的形成,我们确定JAK1对于STAT3的组成型激活至关重要,STAT3的激活是PDAC的突出特征。我们将CCAAT/增强子结合蛋白δ(C/EBPδ)确定为JAK1信号的生物学相关下游靶标,在人类PDAC中上调。恢复C/EBPδ的表达足以恢复缺乏JAK1的癌细胞作为肿瘤球和异种移植小鼠的生长。总的来说,这项研究的结果表明,JAK1通过C/EBPδ执行炎症细胞因子的重要功能,并可能作为PDAC预防和治疗的分子靶标。
    Interleukin-6 (IL-6)-class inflammatory cytokines signal through the Janus tyrosine kinase (JAK)/signal transducer and activator of transcription (STAT) pathway and promote the development of pancreatic ductal adenocarcinoma (PDAC); however, the functions of specific intracellular signaling mediators in this process are less well defined. Using a ligand-controlled and pancreas-specific knockout in adult mice, we demonstrate in this study that JAK1 deficiency prevents the formation of KRASG12D-induced pancreatic tumors, and we establish that JAK1 is essential for the constitutive activation of STAT3, whose activation is a prominent characteristic of PDAC. We identify CCAAT/enhancer binding protein δ (C/EBPδ) as a biologically relevant downstream target of JAK1 signaling, which is upregulated in human PDAC. Reinstating the expression of C/EBPδ was sufficient to restore the growth of JAK1-deficient cancer cells as tumorspheres and in xenografted mice. Collectively, the findings of this study suggest that JAK1 executes important functions of inflammatory cytokines through C/EBPδ and may serve as a molecular target for PDAC prevention and treatment.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号