AKT inhibitors

AKT 抑制剂
  • 文章类型: Journal Article
    背景:抑制PI3K/AKT通路激活可能阻碍癌症的发生和进展。这项研究的目的是评估PI3K/AKT抑制剂的疗效和安全性,并确定最适合不同癌症类型的抑制剂。
    方法:截至2024年6月的电子数据库用于检查PI3K抑制剂的疗效和安全性(alpelisib,copanlisib,duvelisib,和idelalisib)和AKT抑制剂(capivasertib,ipatasertib和MK-2206)用于治疗癌症。通过随机效应配对和网络荟萃分析评估数据。如果不受限制地比较PI3K或AKT抑制剂与非PI3K/AKT对照,则随机对照试验和回顾性研究符合资格。
    结果:结果基于34篇已发表的文章和6项在线注册试验(6710名患者)的34项研究。根据成对荟萃分析,PI3K/AKT抑制剂显示非常有效,特别是治疗突变癌症,但安全性较差。根据我们的网络荟萃分析,PI3K/AKT抑制剂,尤其是AKT抑制剂Capivasertib,对治疗乳腺癌(BC)等实体癌有效。此外,PI3K抑制剂,尤其是idelalisib,可有效治疗血液系统癌症,如慢性淋巴细胞白血病(CLL)。
    结论:PI3K/AKT抑制剂对基因突变患者有效。对于实体癌,如BC,capivasertib是疗效和安全性。对于以CLL为代表的血液癌症,idelalisib是疗效和安全性。上述研究可用于为不同癌症类型的患者推荐合适的靶向治疗。
    BACKGROUND: Inhibiting PI3K/AKT pathway activation may hinder the occurrence and progression of cancer. The aim of this study was to evaluate the efficacy and safety of the PI3K/AKT inhibitors and determine the most appropriate inhibitor for different cancer types.
    METHODS: Electronic databases up to June 2024 were used to examine the efficacy and safety of PI3K inhibitors (alpelisib, copanlisib, duvelisib, and idelalisib) and AKT inhibitors (capivasertib, ipatasertib and MK-2206) for the treatment of cancer. Data was assessed with a random-effect pairwise and network meta-analysis. Randomized controlled trials and retrospective studies were eligible if they compared PI3K or AKT inhibitors with non-PI3K/AKT controls with no restriction.
    RESULTS: The results were based on 34 studies from 34 published articles and 6 online registration trials (6710 patients). According to pairwise meta-analysis, PI3K/AKT inhibitors showed to be highly effective, especially for treating mutant cancers, but had poor safety profiles. According to our network meta-analysis, PI3K/AKT inhibitors, especially the AKT inhibitor capivasertib, are effective for treating solid cancers such as breast cancer (BC). Moreover, PI3K inhibitors, especially idelalisib, were effective for treating hematologic cancers such as chronic lymphocytic leukemia (CLL).
    CONCLUSIONS: The PI3K/AKT inhibitors are effective in patients with genetic mutations. For solid cancers such as BC, capivasertib was efficacy and safety. For hematological cancers represented by CLL, idelalisib was efficacy and safety. The above studies can be used when recommending appropriate targeted therapies for patients with different cancer types.
    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
    所有新出现的传染病中有三分之一是媒介传播的,没有针对任何媒介传播病毒的许可抗病毒疗法。寨卡病毒和Usutu病毒是两种主要由蚊子传播的新兴黄病毒。这些病毒调节不同的宿主途径,包括PI3K/AKT/mTOR通路。这里,我们报道了两种AKT抑制剂对ZIKV和USUV复制的影响,不同哺乳动物和蚊子细胞系中的Miransertib(ARQ-092,变构抑制剂)和Capivasertib(AZD5363,竞争性抑制剂)。在哺乳动物细胞中,Miransertib对ZIKV和USUV的抑制作用强于Capivasertib,而Capivasertib在蚊子细胞中显示出更强的作用。这些发现表明AKT在黄病毒感染中起保守作用。在脊椎动物宿主和无脊椎动物载体中。然而,AKT的特定功能可能因宿主物种而异。这些发现表明,AKT可能在黄病毒感染中发挥保守作用。脊椎动物宿主和无脊椎动物载体。然而,AKT的特定功能可能因宿主物种而异。因此,需要更好地了解病毒与宿主的相互作用,以开发预防人类疾病的新疗法和控制昆虫媒介传播的新方法。
    One third of all emerging infectious diseases are vector-borne, with no licensed antiviral therapies available against any vector-borne viruses. Zika virus and Usutu virus are two emerging flaviviruses transmitted primarily by mosquitoes. These viruses modulate different host pathways, including the PI3K/AKT/mTOR pathway. Here, we report the effect on ZIKV and USUV replication of two AKT inhibitors, Miransertib (ARQ-092, allosteric inhibitor) and Capivasertib (AZD5363, competitive inhibitor) in different mammalian and mosquito cell lines. Miransertib showed a stronger inhibitory effect against ZIKV and USUV than Capivasertib in mammalian cells, while Capivasertib showed a stronger effect in mosquito cells. These findings indicate that AKT plays a conserved role in flavivirus infection, in both the vertebrate host and invertebrate vector. Nevertheless, the specific function of AKT may vary depending on the host species. These findings indicate that AKT may be playing a conserved role in flavivirus infection in both, the vertebrate host and the invertebrate vector. However, the specific function of AKT may vary depending on the host species. A better understanding of virus-host interactions is therefore required to develop new treatments to prevent human disease and new approaches to control transmission by insect vectors.
    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
    PI3K/AKT/mTOR信号通路是乳腺癌中最频繁激活的通路之一,并且在几种生理功能的调节中也起着核心作用。通过开发针对三种激酶的抑制剂(PI3K,AKT,和mTOR)。虽然已经开发了多种化合物,目前,只有三种抑制剂被批准在晚期ER阳性患者中靶向该途径,HER2阴性乳腺癌:依维莫司(mTOR抑制剂),alpelisib(PIK3CA抑制剂),和capivasertib(AKT抑制剂)。像大多数靶向抗癌药物一样,耐药性在临床上是一个主要问题,也是经常限制这些药物整体疗效的一个因素.耐药性可以根据其发展的时间范围分为内在或获得性耐药性。尽管内在抗性在特定治疗之前存在,获得性抵抗是由治疗引起的。大多数ER阳性的患者,HER2阴性晚期乳腺癌可能会在其癌症旅程的某个时候提供PI3K/AKT/mTOR通路的抑制剂。可用的选项取决于批准标准和癌症的突变状态。在这一大群患者中,大多数人可能会在某个时候产生抗药性,这使得这是一个感兴趣的领域,目前尚未满足的需求。在这里,我们回顾了针对PI3K/AKT/mTOR信号通路的药物耐药的常见机制,阐述当前的管理方法,并讨论正在进行的临床试验,试图减轻这一重大问题。我们特别强调需要对AKT1抑制剂耐药性进行更多研究。
    The PI3K/AKT/mTOR signalling pathway is one of the most frequently activated pathways in breast cancer and also plays a central role in the regulation of several physiologic functions. There are major efforts ongoing to exploit precision medicine by developing inhibitors that target the three kinases (PI3K, AKT, and mTOR). Although multiple compounds have been developed, at present, there are just three inhibitors approved to target this pathway in patients with advanced ER-positive, HER2-negative breast cancer: everolimus (mTOR inhibitor), alpelisib (PIK3CA inhibitor), and capivasertib (AKT inhibitor). Like most targeted cancer drugs, resistance poses a major problem in the clinical setting and is a factor that has frequently limited the overall efficacy of these agents. Drug resistance can be categorised into intrinsic or acquired resistance depending on the timeframe it has developed within. Whereas intrinsic resistance exists prior to a specific treatment, acquired resistance is induced by a therapy. The majority of patients with ER-positive, HER2-negative advanced breast cancer will likely be offered an inhibitor of the PI3K/AKT/mTOR pathway at some point in their cancer journey, with the options available depending on the approval criteria in place and the cancer\'s mutation status. Within this large cohort of patients, it is likely that most will develop resistance at some point, which makes this an area of interest and an unmet need at present. Herein, we review the common mechanisms of resistance to agents that target the PI3K/AKT/mTOR signalling pathway, elaborate on current management approaches, and discuss ongoing clinical trials attempting to mitigate this significant issue. We highlight the need for additional studies into AKT1 inhibitor resistance in particular.
    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
    背景:将AKT抑制剂纳入晚期或转移性乳腺癌的辅助治疗可改善临床结果。然而,应更好地评估AKT抑制剂的安全性,考虑到延长生存期和影响患者生活质量的可能性。我们的目的是评估在辅助治疗中添加AKT抑制剂如何影响治疗相关的不良事件。
    方法:我们用风险比(RRs)评估二元结果,95%置信区间(CI)。我们对所有端点使用了DerSimonian和Laird随机效应模型。使用I2统计学评估异质性。R,版本4.2.3用于统计分析。
    结果:总共7个RCT,包括1619例BC患者。显示出有利于AKT抑制剂不良反应发生的统计学意义的不良反应是腹泻(RR3.05;95%CI2.48-3.75;p<0.00001;I2=49%),高血糖(RR3.4;95%CI1.69-6.83;p=0.00058;I2=75%),恶心(RR1.69;95%CI1.34-2.13;p=0.000008;I2=42%),皮疹(RR2.79;95%CI1.49-5.23;p=0.0013;I2=82%),口腔炎(RR2.24;95%CI1.69-2.97;p<0.00001;I2=16%)和呕吐(RR2.99;95%CI1.85-4.86;p=0.00009;I2=42%)。脱发组之间没有显着差异(p=0.80),疲劳(p=0.087),和神经病变(p=0.363380)。
    结论:在辅助治疗中添加AKT抑制剂与治疗相关不良事件的增加相关。这些结果为评估AKT抑制剂治疗转移性BC患者的进一步临床试验提供了安全性信息。临床医生应密切监测患者与治疗相关的不良事件,以避免因这些早期治疗而导致的治疗中断和发病率。
    BACKGROUND: Incorporation of AKT inhibitors into adjuvant therapy for advanced or metastatic breast cancer has improved clinical outcomes. However, the safety of AKT inhibitors should be better evaluated, given the possibility of prolonging survival and impacting patient quality of life. Our aim was to assess how the addition of AKT inhibitors to adjuvant therapy affects treatment-related adverse events.
    METHODS: We evaluated binary outcomes with risk ratios (RRs), with 95% confidence intervals (CIs). We used DerSimonian and Laird random-effect models for all endpoints. Heterogeneity was assessed using I2 statistics. R, version 4.2.3, was used for statistical analyses.
    RESULTS: A total of seven RCTs comprising 1619 patients with BC. The adverse effects that show significance statistical favoring the occurrence of adverse effects in AKT inhibitor were diarrhea (RR 3.05; 95% CI 2.48-3.75; p < 0.00001; I2 = 49%), hyperglycemia (RR 3.4; 95% CI 1.69-6.83; p = 0.00058; I2 = 75%), nausea (RR 1.69; 95% CI 1.34-2.13; p = 0.000008; I2 = 42%), rash (RR 2.79; 95% CI 1.49-5.23; p = 0.0013; I2 = 82%), stomatitis (RR 2.24; 95% CI 1.69-2.97; p < 0.00001; I2 = 16%) and vomiting (RR 2.99; 95% CI 1.85-4.86; p = 0.00009; I2 = 42%). There was no significant difference between the groups for alopecia (p = 0.80), fatigue (p = 0.087), and neuropathy (p = 0.363380).
    CONCLUSIONS: The addition of AKT inhibitors to adjuvant therapy was associated with an increase in treatment-related adverse events. These results provide safety information for further clinical trials evaluating AKT inhibitor therapy for patients with metastatic BC. Clinicians should closely monitor patients for treatment-related adverse events to avoid discontinuation of therapy and morbidity caused by these early-stage therapies.
    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
    尽管开发了许多治疗策略,但血液恶性肿瘤仍然是治疗挑战。磷脂酰肌醇-3激酶(PI3K)/蛋白激酶B/哺乳动物雷帕霉素靶蛋白(PI3K/Akt/mTOR)信号通路在调节许多细胞功能中起着核心作用。包括细胞周期,扩散,静止,和长寿。因此,该通路的失调是癌变的特征。PI3K/Akt/mTOR信号的激活增强增殖,增长,以及癌细胞对化学疗法和免疫疗法的抗性。已经在各种类型的癌症中发现了该途径的过度激活,包括急性和慢性白血病。自2014年以来,PI3K/Akt/mTOR途径的抑制剂已用于白血病治疗,其中一些在临床试验中改善了治疗结果。最近,已经开发了新的PI3K/Akt/mTOR信号抑制剂,并在临床前和临床模型中进行了测试.在这次审查中,我们概述了PI3K/Akt/mTOR信号通路在血液恶性肿瘤细胞中的作用,并收集了该通路抑制剂的信息,这些抑制剂可能为白血病提供新的治疗机会.
    Blood malignancies remain a therapeutic challenge despite the development of numerous treatment strategies. The phosphatidylinositol-3 kinase (PI3K)/protein kinase B/mammalian target of rapamycin (PI3K/Akt/mTOR) signaling pathway plays a central role in regulating many cellular functions, including cell cycle, proliferation, quiescence, and longevity. Therefore, dysregulation of this pathway is a characteristic feature of carcinogenesis. Increased activation of PI3K/Akt/mTOR signaling enhances proliferation, growth, and resistance to chemo- and immunotherapy in cancer cells. Overactivation of the pathway has been found in various types of cancer, including acute and chronic leukemia. Inhibitors of the PI3K/Akt/mTOR pathway have been used in leukemia treatment since 2014, and some of them have improved treatment outcomes in clinical trials. Recently, new inhibitors of PI3K/Akt/mTOR signaling have been developed and tested both in preclinical and clinical models. In this review, we outline the role of the PI3K/Akt/mTOR signaling pathway in blood malignancies\' cells and gather information on the inhibitors of this pathway that might provide a novel therapeutic opportunity against leukemia.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

  • 文章类型: English Abstract
    BACKGROUND: An increasing understanding of the cellular processes involved in growth, metastasis and development of resistance enable the development of new treatment strategies for advanced prostate cancer.
    OBJECTIVE: Using selected examples, the aim of this report is to present current developments to the reader and to give an outlook on possible upcoming changes in the treatment of advanced prostate cancer.
    METHODS: Narrative report based on expert consensus, supported by a literature search in PubMed (MEDLINE) and the abstract databases of the American Society of Clinical Oncology (ASCO) and European Society of Medical Oncology (ESMO). Examples were selected to illustrate current developments without claiming completeness.
    RESULTS: The androgen receptor (AR) signal transduction pathway remains a focus of scientific interest. Androgen synthesis inhibitors and AR degraders are promising new approaches to overcome resistance mediated by AR mutations or splice variants. Inhibition of key switch sites of alternative signaling pathways such as AKT or CDK4/6 provide additional treatment options, including combinational strategies through a tight linkage with the AR signaling pathway. A better understanding of tumor microenvironment and immune response is required for novel immunotherapeutic strategies using bispecific T‑cell engagers (BiTEs) and chimeric antigen receptor (CAR) T cells.
    CONCLUSIONS: New treatment strategies give hope that we will be able to intervene even more effectively in the course of disease of advanced prostate cancer in the future. However, a major challenge, especially for the implementation of targeted treatment approaches, is likely to be the heterogeneity of tumor progression not only inter- but also intrapersonally.
    UNASSIGNED: HINTERGRUND: Ein zunehmendes Verständnis für die zellulären Vorgänge bei Wachstum, Metastasierung und Resistenzentwicklung ermöglichen die Entwicklung neuer Behandlungsstrategien für das fortgeschrittene Prostatakarzinom.
    UNASSIGNED: Anhand ausgewählter Beispiele sollen dem Leser aktuelle Entwicklungen dargelegt und ein Ausblick auf einen möglichen bevorstehenden Wandel in der Therapie des fortgeschrittenen Prostatakarzinoms gegeben werden.
    METHODS: Narrativer Bericht basierend auf einem Expertenkonsens, unterstützt von einer Literaturrecherche in PubMed (MEDLINE) und den Abstract-Datenbanken von American Society of Clinical Oncology (ASCO) und European Society for Medical Oncology (ESMO). Ausgewählt wurden Beispiele zur Illustration der aktuellen Entwicklungen ohne den Anspruch auf Vollständigkeit.
    UNASSIGNED: Der Androgenrezeptor (AR)-Signaltransduktionsweg ist unverändert im Zentrum des wissenschaftlichen Interesses. Androgensyntheseinhibitoren und AR-Degrader sind neue vielversprechende Ansätze, um durch Mutationen oder Splicevarianten vermittelte Resistenzen zu überwinden. Die Hemmung zentraler Schaltstellen alternativer Signalwege wie AKT oder CDK4/6 (Cyclin-abhängige Kinasen) ermöglichen zusätzliche Behandlungsoptionen, auch als Kombinationsstrategien durch eine enge Vernetzung mit dem AR-Signalweg. Ein besseres Verständnis von Tumormikromilieu und Immunantwort ist Voraussetzung für neue immuntherapeutische Strategien mit bispezifischen T‑Zell-Engagern (BiTE) und chimären Antigenrezeptor (CAR)-T-Zellen.
    UNASSIGNED: Die neuen Behandlungsstrategien lassen hoffen, dass wir künftig noch effektiver in den Krankheitsverlauf des fortgeschrittenen Prostatakarzinoms eingreifen können. Eine große Herausforderung, insbesondere für die Umsetzung der zielgerichteten Therapieansätze, dürfte allerdings die Heterogenität des Tumorgeschehens nicht nur inter- sondern auch intrapersonell darstellen.
    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
    从头转移激素敏感型PC(mHSPC)占所有前列腺癌(PC)诊断的5-10%,但占PC相关死亡的近50%。自2015年以来,mHSPC的预后略有改善,这要归功于从一线开始引入新的激素药物和化疗联合雄激素剥夺治疗。这篇综述描述了当前从头mHSPC的治疗机会,重点关注在主要临床试验中确定的潜在分子生物标志物,这些生物标志物已经修改了护理标准,从头mHSPC的基因组特征,以及正在进行的主要试验,这些试验正在研究新的治疗方法以及在这种情况下生物标志物指导治疗的有效性。从头mHSPC的个性化治疗道路仍然漫长,考虑到随机临床试验,提供了当前治疗选择的基础,根据不一定反映所选治疗的生物学原理的临床标准对患者进行分层.mHSPC作为预测生物标志物的转录组学分析的作用需要进一步验证。还有待确定如何在mHSPC中检测到基因组变异,它们被认为是对去势抗性疾病的预测,可以在mHSPC设置中利用。
    De novo metastatic hormone-sensitive PC (mHSPC) accounts for 5-10% of all prostate cancer (PC) diagnoses but it is responsible for nearly 50% of PC-related deaths. Since 2015, the prognosis of mHSPC has slightly improved thanks to the introduction of new hormonal agents and chemotherapy combined with androgen deprivation therapy from the first-line setting. This review describes the current therapeutic opportunities for de novo mHSPC, focusing on potential molecular biomarkers identified in the main clinical trials that have modified the standard of care, the genomic features of de novo mHSPC, and the principal ongoing trials that are investigating new therapeutic approaches and the efficacy of a biomarker-guided treatment in this setting. The road toward personalized treatment for de novo mHSPC is still long, considering that the randomized clinical trials, which have furnished the basis of the current therapeutic options, stratified patients according to clinical criteria that did not necessarily reflect the biological rationale of the chosen therapy. The role of transcriptomic profiling of mHSPC as a predictive biomarker requires further validation, and it remains to be ascertained how the genomic variants detected in mHSPC, which are regarded as predictive in the castration-resistant disease, can be exploited in the mHSPC setting.
    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
    PI3K/AKT/mTOR(PAM)信号通路是真核细胞中高度保守的信号转导网络,可促进细胞存活,细胞生长,和细胞周期进程。PAM轴中转录因子的生长因子信号传导受到与其他几种信号传导途径的多重交叉相互作用的高度调节。信号转导的失调可能导致癌症的发展。PAM轴是人类癌症中最频繁激活的信号传导途径,并且通常涉及对抗癌疗法的抗性。该途径的成分的功能障碍,如PI3K的过度活跃,PTEN功能的丧失,和AKT的功能增益,是癌症治疗抵抗和疾病进展的臭名昭著的驱动因素。在这篇综述中,我们强调了癌症中PAM信号通路的主要失调,并讨论了PI3K的结果,AKT和mTOR抑制剂作为单一疗法并在临床试验中与其他抗肿瘤剂共同施用作为克服治疗抗性的策略。最后,PAM信号靶向治疗耐药的主要机制,还讨论了包括PAM信号在免疫学和免疫疗法中的应用。
    The PI3K/AKT/mTOR (PAM) signaling pathway is a highly conserved signal transduction network in eukaryotic cells that promotes cell survival, cell growth, and cell cycle progression. Growth factor signalling to transcription factors in the PAM axis is highly regulated by multiple cross-interactions with several other signaling pathways, and dysregulation of signal transduction can predispose to cancer development. The PAM axis is the most frequently activated signaling pathway in human cancer and is often implicated in resistance to anticancer therapies. Dysfunction of components of this pathway such as hyperactivity of PI3K, loss of function of PTEN, and gain-of-function of AKT, are notorious drivers of treatment resistance and disease progression in cancer. In this review we highlight the major dysregulations in the PAM signaling pathway in cancer, and discuss the results of PI3K, AKT and mTOR inhibitors as monotherapy and in co-administation with other antineoplastic agents in clinical trials as a strategy for overcoming treatment resistance. Finally, the major mechanisms of resistance to PAM signaling targeted therapies, including PAM signaling in immunology and immunotherapies are also discussed.
    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
    卵巢癌已成为妇科癌症相关死亡的最大原因。它通常在晚期诊断,没有有效的筛查策略。卵巢癌是一种高度异质性的疾病,可以细分为几个分子亚群。由于对参与癌变和肿瘤生长的分子途径有了更深入的了解,靶向药物已被批准或处于几个开发阶段。聚(ADP-核糖)聚合酶(PARP)抑制剂和抗血管内皮生长因子(VEGF)-A抗体是目前批准的两种最有效的卵巢癌靶向药物。随着贝伐单抗的成功,正在研究可以靶向替代血管生成途径的酪氨酸激酶抑制剂。此外,许多针对PI3激酶(PI3K)/AKT/哺乳动物雷帕霉素靶蛋白(mTOR)途径的治疗,正在开发或已经在临床研究中。MicroRNA也已成为卵巢癌治疗和临床诊断的新型生物标志物。这篇手稿回顾了分子,临床前和临床证据支持卵巢癌中生长依赖性途径的靶向,并评估与PARP抑制剂以外的靶向治疗相关的当前数据.
    Ovarian cancer has become the largest cause of gynaecological cancer-related mortality. It is typically diagnosed at a late stage and has no effective screening strategy. Ovarian cancer is a highly heterogeneous disease that can be subdivided into several molecular subsets. As a result of a greater understanding of molecular pathways involved in carcinogenesis and tumor growth, targeted agents have been approved or are in several stages of development. Poly(ADP-ribose) polymerase (PARP) inhibitors and the anti-vascular endothelial growth factor (VEGF)-A antibodies are two types of approved and most effective targeted drugs for ovarian cancer at present. With the success of bevacizumab, tyrosine kinase inhibitors which could target alternate angiogenic pathways are being studied. Furthermore, many treatments targeting the PI3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) pathways, are being developed or are already in clinical studies. MicroRNAs have also become novel biomarkers for the therapy and clinical diagnosis of ovarian cancer. This manuscript reviews the molecular, preclinical and clinical evidence supporting the targeting of growth-dependent pathways in ovarian cancer and assesses current data related to targeted treatments beyond PARP 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
    Toll样受体8(TLR8)识别病毒和细菌来源的单链RNA,并介导人单核细胞和巨噬细胞分泌促炎细胞因子和I型干扰素。TLR8与其他内体TLRs一样,利用MyD88衔接蛋白启动来自内体的信号传导。这里,我们探讨了含Toll-白介素1受体结构域的衔接蛋白(TIRAP)在人原代单核细胞源性巨噬细胞(MDMs)TLR8信号调节中的潜在作用.要做到这一点,我们进行了TIRAP基因沉默,然后用合成配体或活细菌刺激细胞。通过定量PCR或Bioplex测定分析细胞因子基因的表达和分泌。分别,而转录因子的核易位通过免疫荧光和成像来解决,以及细胞分级分离和免疫印迹。免疫沉淀和Akt抑制剂也用于剖析信号传导机制。总的来说,我们显示TIRAP被招募到TLR8Mydosome信号传导复合物,其中TIRAP有助于Akt激酶激活和干扰素调节因子5(IRF5)的核易位。TIRAP对TLR8信号复合物的募集促进IRF5依赖性细胞因子IFNβ和IL-12p70的表达和分泌,在较小程度上,TNF.这些发现揭示了TIRAP在先天免疫防御中的新的非常规作用。
    Toll-like receptor 8 (TLR8) recognizes single-stranded RNA of viral and bacterial origin as well as mediates the secretion of pro-inflammatory cytokines and type I interferons by human monocytes and macrophages. TLR8, as other endosomal TLRs, utilizes the MyD88 adaptor protein for initiation of signaling from endosomes. Here, we addressed the potential role of the Toll-interleukin 1 receptor domain-containing adaptor protein (TIRAP) in the regulation of TLR8 signaling in human primary monocyte-derived macrophages (MDMs). To accomplish this, we performed TIRAP gene silencing, followed by the stimulation of cells with synthetic ligands or live bacteria. Cytokine-gene expression and secretion were analyzed by quantitative PCR or Bioplex assays, respectively, while nuclear translocation of transcription factors was addressed by immunofluorescence and imaging, as well as by cell fractionation and immunoblotting. Immunoprecipitation and Akt inhibitors were also used to dissect the signaling mechanisms. Overall, we show that TIRAP is recruited to the TLR8 Myddosome signaling complex, where TIRAP contributes to Akt-kinase activation and the nuclear translocation of interferon regulatory factor 5 (IRF5). Recruitment of TIRAP to the TLR8 signaling complex promotes the expression and secretion of the IRF5-dependent cytokines IFNβ and IL-12p70 as well as, to a lesser degree, TNF. These findings reveal a new and unconventional role of TIRAP in innate immune defense.
    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