TAK1

TAK1
  • 文章类型: Journal Article
    系统性硬化症(SSc)是一种慢性自身免疫性疾病,其特征是皮肤和多个重要器官的纤维化,但SSc的免疫学发病机制尚不清楚。我们在这里显示miR-19b促进Th9细胞加剧SSc。具体来说,miR-19b和白介素(IL)-9在博来霉素诱导的小鼠实验性SSc中增加CD4+T细胞。抑制miR-19b可减少Th9细胞并改善疾病。机械上,转化生长因子β(TGF-β)加IL-4通过抑制NLRC3激活pSmad3-Ser213和TRAF6-K63泛素化。激活的TRAF6依次促进TGF-β激活的激酶1(TAK1)和核因子κB(NF-κB)p65的磷酸化,导致miR-19b的上调。值得注意的是,miR-19b通过直接抑制非典型E2F家族成员E2f8激活Il9基因表达。在SSc患者中,更高水平的IL9和MIR-19B与更严重的疾病进展相关。我们的发现揭示了miR-19b是Th9细胞介导的SSc发病机制的关键因素,并且应该对SSc患者具有临床意义。
    Systemic sclerosis (SSc) is a chronic autoimmune disease characterized by fibrosis of the skin and multiple vital organs, but the immunological pathogenesis of SSc remains unclear. We show here that miR-19b promotes Th9 cells that exacerbate SSc. Specifically, miR-19b and interleukin (IL)-9 increase in CD4+ T cells in experimental SSc in mice induced with bleomycin. Inhibiting miR-19b reduces Th9 cells and ameliorates the disease. Mechanistically, transforming growth factor beta (TGF-β) plus IL-4 activates pSmad3-Ser213 and TRAF6-K63 ubiquitination by suppressing NLRC3. Activated TRAF6 sequentially promotes TGF-β-activated kinase 1 (TAK1) and nuclear factor κB (NF-κB) p65 phosphorylation, leading to the upregulation of miR-19b. Notably, miR-19b activated Il9 gene expression by directly suppressing atypical E2F family member E2f8. In patients with SSc, higher levels of IL9 and MIR-19B correlate with worse disease progression. Our findings reveal miR-19b as a key factor in Th9 cell-mediated SSc pathogenesis and should have clinical implications for patients with SSc.
    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:本观察性研究探讨Pim-1在炎症信号通路中的调控机制。
    方法:使用THP-1、RAW264.7、BV2和Jurkat人T细胞系。
    方法:无。
    方法:用脂多糖(LPS)诱导炎症,其次是PIM1敲除。蛋白质印迹,免疫沉淀,免疫荧光,和RT-PCR测定用于评估PIM1敲低对LPS诱导的炎症的影响。
    结果:巨噬细胞样THP-1细胞中PIM1敲低抑制LPS诱导的促炎细胞因子的上调,诱导型一氧化氮合酶,环氧合酶-2,磷酸化Janus激酶,信号转导和转录激活因子3,细胞外信号调节激酶,c-Jun氨基末端激酶,p38和核因子κBp65(NF-κBp65)。它还抑制了NF-κB激酶α/β抑制剂的上调,并增强了NF-κBp65的核易位。此外,它抑制了NOD样受体家族pyrin结构域3(NLRP3)的上调,并抑制了LPS与三磷酸腺苷共同处理诱导的caspase-1的裂解。此外,p-转化生长因子-β激活激酶1(TAK1)与Pim-1相互作用。Pim激酶的所有三个成员(Pim-1,Pim-2和Pim-3)都是LPS介导的巨噬细胞炎症所必需的;然而,与Pim-1和Pim-3不同,Pim-2充当T细胞活性的负调节因子。
    结论:Pim-1在LPS诱导的炎症反应中与TAK1相互作用,并参与MAPK/NF-κB/NLRP3信号通路。此外,考虑到Pim-2在T细胞中的负调节作用,需要进一步深入研究它们各自的功能。
    OBJECTIVE: This observational study investigated the regulatory mechanism of Pim-1 in inflammatory signaling pathways.
    METHODS: THP-1, RAW 264.7, BV2, and Jurkat human T cell lines were used.
    METHODS: None.
    METHODS: Lipopolysaccharide (LPS) was used to induce inflammation, followed by PIM1 knockdown. Western blot, immunoprecipitation, immunofluorescence, and RT-PCR assays were used to assess the effect of PIM1 knockdown on LPS-induced inflammation.
    RESULTS: PIM1 knockdown in macrophage-like THP-1 cells suppressed LPS-induced upregulation of pro-inflammatory cytokines, inducible nitric oxide synthase, cyclooxygenase-2, phosphorylated Janus kinase, signal transducer and activator of transcription 3, extracellular signal-regulated kinase, c-Jun N-terminal kinase, p38, and nuclear factor kappa B p65 (NF-κB p65). It also suppressed upregulation of inhibitor of NF-κB kinase α/β and enhanced the nuclear translocation of NF-κB p65. Moreover, it inhibited the upregulation of Nod-like receptor family pyrin domain-containing 3 (NLRP3) and cleavage of caspase-1 induced by co-treatment of LPS with adenosine triphosphate. Additionally, p-transforming growth factor-β-activated kinase 1 (TAK1) interacted with Pim-1. All three members of Pim kinases (Pim-1, Pim-2, and Pim-3) were required for LPS-mediated inflammation in macrophages; however, unlike Pim-1 and Pim-3, Pim-2 functioned as a negative regulator of T cell activity.
    CONCLUSIONS: Pim-1 interacts with TAK1 in LPS-induced inflammatory responses and is involved in MAPK/NF-κB/NLRP3 signaling pathways. Additionally, considering the negative regulatory role of Pim-2 in T cells, further in-depth studies on their respective functions are needed.
    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
    许多DNA病毒开发了各种策略来抑制细胞死亡以促进其复制。然而,是否甲型流感病毒(IAV),一种快速复制的RNA病毒,减弱细胞死亡仍然未知。这里,我们报道IAV感染诱导小鼠肺泡上皮细胞系(LET1)和小鼠成纤维细胞瘤细胞系(L929)中TAK1磷酸化。TAK1特异性抑制剂5Z-7-Oxzeneonal(5Z)和TAK1敲除显着增强IAV诱导的细胞凋亡,PARP增加证明了这一点,caspase-8和caspase-3切割。TAK1抑制也增加坏死,如RIPK1S166、RIPK3T231/S232和MLKLS345磷酸化增加所证明。机械上,TAK1激活IKK,磷酸化RIPK1S25并抑制其活化。TAK1还激活p38及其下游激酶MK2,使RIPK1S321磷酸化,但不影响RIPK1的激活。进一步的研究表明,RIPK1抑制剂Nec-1和RIPK1敲除可消除IAV诱导的细胞凋亡和坏死;野生型而非激酶死亡(KD)-RIPK1的再表达可恢复IAV诱导的细胞死亡。ZBP1敲除消除IAV诱导的细胞死亡,而RIPK3敲除抑制IAV诱导的坏死,但不抑制细胞凋亡。5Z治疗可增强IAV诱导的细胞死亡,并略微降低H1N1病毒感染小鼠肺部的炎症反应,并延长IAV感染小鼠的生存期。我们的研究提供了IAV激活TAK1以抑制RIPK1依赖性细胞凋亡和坏死的证据,并且RIPK3是IAV诱导的坏死而不是上皮细胞凋亡所必需的。
    Many DNA viruses develop various strategies to inhibit cell death to facilitate their replication. However, whether influenza A virus (IAV), a fast-replicating RNA virus, attenuates cell death remains unknown. Here, we report that IAV infection induces TAK1 phosphorylation in a murine alveolar epithelial cell line (LET1) and a murine fibroblastoma cell line (L929). The TAK1-specific inhibitor 5Z-7-Oxzeneonal (5Z) and TAK1 knockout significantly enhance IAV-induced apoptosis, as evidenced by increased PARP, caspase-8, and caspase-3 cleavage. TAK1 inhibition also increases necroptosis as evidenced by increased RIPK1S166, RIPK3T231/S232, and MLKLS345 phosphorylation. Mechanistically, TAK1 activates IKK, which phosphorylates RIPK1S25 and inhibits its activation. TAK1 also activates p38 and its downstream kinase MK2, which phosphorylates RIPK1S321 but does not affect RIPK1 activation. Further investigation revealed that the RIPK1 inhibitor Nec-1 and RIPK1 knockout abrogate IAV-induced apoptosis and necroptosis; re-expression of wild-type but not kinase-dead (KD)-RIPK1 restores IAV-induced cell death. ZBP1 knockout abrogates IAV-induced cell death, whereas RIPK3 knockout inhibits IAV-induced necroptosis but not apoptosis. 5Z treatment enhances IAV-induced cell death and slightly reduces the inflammatory response in the lungs of H1N1 virus-infected mice and prolongs the survival of IAV-infected mice. Our study provides evidence that IAV activates TAK1 to suppress RIPK1-dependent apoptosis and necroptosis, and that RIPK3 is required for IAV-induced necroptosis but not apoptosis in epithelial cells.
    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
    转化生长因子β激活激酶1(TAK1)/c-JunN末端激酶(JNK)轴是必需的MAPK上游介质,并调节免疫信号通路。然而,目前尚不清楚TAK1/JNK轴是否在早期脊椎动物适应性免疫中发挥调节信号转导的强度。在这项研究中,通过对尼罗罗非鱼(Oreochromisniloticus)建模,我们研究了TAK1/JNK轴对淋巴细胞介导的适应性免疫应答的潜在调节功能.相对于其他脊椎动物中的对应物,OnTAK1和OnJNK均表现出高度保守的序列和结构。它们的mRNA在免疫相关组织中广泛表达,而脾淋巴细胞的磷酸化水平在感染后第4天显着增强。此外,在佛波醇12-肉豆蔻酸酯13-乙酸酯加离子霉素(PI)或PHA体外激活淋巴细胞后,OnTAK1和OnJNK的转录水平显着上调,伴随着磷酸化水平的显著增加。更重要的是,特异性抑制剂NG25对OnTAK1活性的抑制导致OnJNK磷酸化水平显著降低。此外,用特异性抑制剂SP600125阻断OnJNK的活性导致T细胞活化标志物(包括IFN-γ)的表达显着降低,PHA诱导的T细胞活化期间的CD122、IL-2和CD44。总之,这些发现表明,尼罗罗非鱼中保守的TAK1/JNK轴通过调节淋巴细胞的激活而参与适应性免疫反应。本研究丰富了目前对硬骨鱼适应性免疫的认识,为理解鱼类免疫调节机制提供了新的视角。
    The transforming growth factor beta-activated kinase 1 (TAK1)/c-Jun N-terminal kinase (JNK) axis is an essential MAPK upstream mediator and regulates immune signaling pathways. However, whether the TAK1/JNK axis harnesses the strength in regulation of signal transduction in early vertebrate adaptive immunity is unclear. In this study, by modeling on Nile tilapia (Oreochromis niloticus), we investigated the potential regulatory function of TAK1/JNK axis on lymphocyte-mediated adaptive immune response. Both OnTAK1 and OnJNK exhibited highly conserved sequences and structures relative to their counterparts in other vertebrates. Their mRNA was widely expressed in the immune-associated tissues, while phosphorylation levels in splenic lymphocytes were significantly enhanced on the 4th day post-infection by Edwardsiella piscicida. In addition, OnTAK1 and OnJNK were significantly up-regulated in transcriptional level after activation of lymphocytes in vitro by phorbol 12-myristate 13-acetate plus ionomycin (P + I) or PHA, accompanied by a predominant increase in phosphorylation level. More importantly, inhibition of OnTAK1 activity by specific inhibitor NG25 led to a significant decrease in the phosphorylation level of OnJNK. Furthermore, blocking the activity of OnJNK with specific inhibitor SP600125 resulted in a marked reduction in the expression of T-cell activation markers including IFN-γ, CD122, IL-2, and CD44 during PHA-induced T-cell activation. In summary, these findings indicated that the conserved TAK1/JNK axis in Nile tilapia was involved in adaptive immune responses by regulating the activation of lymphocytes. This study enriched the current knowledge of adaptive immunity in teleost and provided a new perspective for understanding the regulatory mechanism of fish immunity.
    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
    关节痛是美国最常见的疼痛类型之一。在患有炎症性疾病如骨关节炎(OA)和痛风的患者的情况下,由于几种关键细胞因子的长期过度表达而导致的持续性炎症与关节内神经元超敏反应和损伤有关.最终,一部分患者出现慢性疼痛.关节痛的药物治疗包括使用镇痛药,如对乙酰氨基酚,非甾体抗炎药(NSAIDs),阿片类药物,抗抑郁药,以及关节内注射皮质类固醇和透明质酸。然而,NSAIDs是短效的,不能缓解剧烈的疼痛。阿片类药物通常在控制慢性疼痛方面无效,所有的治疗选择都会增加严重副作用的风险。
    我们探索了转化生长因子-β-活化激酶1(TAK1)在单碘乙酸盐(MIA)和尿酸单钠(MSU)关节疼痛模型中的治疗和镇痛作用,作为缓解慢性炎性疼痛的创新策略。机械性异常性疼痛(VonFrey),在接受选择性TAK1抑制剂的单独大鼠组中测量负重和组织学变化,HS-276,加巴喷丁或车辆。
    我们的数据支持,TAK1抑制有效地防止了MIA模型中机械性异常疼痛和差别性负重的发展。在痛风性关节炎的MSU模型中,HS-276治疗可显着减少雌性大鼠的机械性异常疼痛和膝关节水肿,但不是雄性老鼠.两种模型中受影响的关节的组织学评估表明,HS-276治疗可显着降低疾病引起的关节降解。
    我们的结果支持TAK1是炎性关节疾病如OA和痛风性关节炎的关键信号节点。选择性药理抑制显著减弱疾病的几个方面,包括关节退化和机械性疼痛。因此,TAK1是治疗疼痛性炎症性关节疾病的新治疗靶点。
    本文报道了TAK1在治疗OA和痛风等慢性炎症性关节疾病中的治疗潜力。使用选择性TAK1抑制剂,HS-276,我们显示了TAK1抑制在两种临床前鼠炎性关节痛模型中的治疗和镇痛作用。
    UNASSIGNED: Joint pain is one of the most commonly reported pain types in the United States. In the case of patients suffering from inflammatory diseases such as osteoarthritis (OA) and gout, persistent inflammation due to long-term overexpression of several key cytokines has been linked to neuronal hypersensitivity and damage within the joints. Ultimately, a subset of patients develop chronic pain. Pharmacologic treatment of joint pain involves the use of analgesics such as acetaminophen, non-steroidal anti-inflammatory drugs (NSAIDs), opioids, antidepressants, as well as intra-articular injections of corticosteroids and hyaluronic acid. However, NSAIDs are short-acting and fail to alleviate severe pain, opioids are generally ineffective at managing chronic pain, and all therapeutic options involve increased risks of serious side effects.
    UNASSIGNED: We explored the therapeutic and analgesic effects of transforming growth factor-β-activated kinase 1 (TAK1) inhibition in both the monoiodoacetate (MIA) and monosodium urate (MSU) models of joint pain as an innovative strategy for alleviating chronic inflammatory pain. Mechanical allodynia (Von Frey), weight-bearing and histological changes were measured in separate groups of rats receiving either the selective TAK1 inhibitor, HS-276, gabapentin or vehicle.
    UNASSIGNED: Our data support that TAK1 inhibition effectively prevented the development of mechanical allodynia and differential weight-bearing in the MIA model. In the MSU model of gouty arthritis, treatment with HS-276 significantly reduced mechanical allodynia and knee edema in female rats, but not male rats. Histological evaluation of effected joints in both models showed that HS-276 treatment significantly reduced disease-induced degradation of the joint.
    UNASSIGNED: Our results support that TAK1 is a critical signaling node in inflammatory joint diseases such as OA and gouty arthritis. Selective pharmacological inhibition significantly attenuated several aspects of the disease, including joint degeneration and mechanical pain. Thus, TAK1 is a novel therapeutic target for the treatment of painful inflammatory joint diseases.
    UNASSIGNED: This article reports on the therapeutic potential of TAK1 in the treatment of chronic inflammatory joint diseases such as OA and gout. Using the selective TAK1 inhibitor, HS-276, we show the therapeutic and analgesic effects of TAK1 inhibition in two preclinical murine models of inflammatory joint pain.
    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
    椎间盘退变(IVDD)是一种常见的退行性疾病,与下腰痛(LBP)密切相关,带来巨大的经济和社会负担。在这项研究中,由于具有令人信服的抗炎和抗氧化功能,我们探索了植苗素(Hom)对IVDD的治疗效果。TNF-α用于体外模拟髓核(NP)细胞的炎症环境。我们证实Hom可以通过阻断NF-κB/MAPK信号通路减轻TNF-α诱导的炎症和ECM稳态紊乱。随后,我们筛选了Hom的结合靶标,证实Hom可以直接结合TAK1并抑制其磷酸化,从而下调炎症相关通路.通过针刺大鼠体内模型进一步验证了Hom对IVDD的治疗作用。总的来说,Hom是IVDD早期干预的一个有希望的小分子,具有巨大的临床转化价值。
    Intervertebral disc degeneration (IVDD) is a common degenerative disease which is closely related to low back pain (LBP) and brings huge economic and social burdens. In this study, we explored the therapeutic effects of Homoplantaginin (Hom) for IVDD due to its convincing anti-inflammatory and antioxidant functions. TNF-α was used to simulate the inflammatory environment for nucleus pulposus (NP) cells in vitro. We verified that Hom could alleviate the TNF-α-induced inflammation and disturbance of ECM homeostasis through blocking the NF-κB/MAPK signaling pathways. Subsequently, we screened the binding targets of Hom and confirmed that Hom could directly bind to TAK1 and inhibit its phosphorylation to down-regulate the inflammation-related pathways. The therapeutic effects of Hom on IVDD were further validated through a needle puncture rat model in vivo. Overall, Hom was a promising small molecule for IVDD early intervention, possessing huge clinical translational value.
    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
    急性肺损伤(ALI),在各种临床疾病中观察到的严重并发症,以广泛的炎症为特征,中性粒细胞浸润,和促炎细胞因子的产生。这项研究表明,最近鉴定的非编码RNAISIR及其人类同源基因AK131315在调节脂多糖(LPS)诱导的炎症反应中起作用。ISIR和AK131315增加了LPS刺激的巨噬细胞中炎性细胞因子的产生,在ALI动物模型中,外源性ISIR加重了LPS诱导的肺部炎症。机械上,ISIR通过增强TAK1激活促进LPS触发的NF-κB和MAPK信号传导和促炎细胞因子的转录。此外,观察到AK131315表达与革兰氏阴性菌引起的肺部感染之间存在显着相关性,这表明AK131315在细菌感染中起重要作用。总之,这些发现表明ISIR调节LPS诱导的炎症,AK131315参与细菌感染的发病机制.
    Acute lung injury (ALI), a critical complication observed in various clinical disorders, is characterized by widespread inflammation, neutrophil infiltration, and proinflammatory cytokine production. This study showed that the recently identified non-coding RNA ISIR and its human homolog gene AK131315 played a role in regulating lipopolysaccharide (LPS)-induced inflammatory responses. ISIR and AK131315 increased the production of inflammatory cytokines in LPS-stimulated macrophages, and exogenous ISIR aggravated LPS-induced lung inflammation in an animal model of ALI. Mechanistically, ISIR promoted LPS-triggered NF-κB and MAPK signaling and the transcription of proinflammatory cytokines by enhancing TAK1 activation. Furthermore, a significant correlation was observed between AK131315 expression and pulmonary infectious caused by Gram-negative bacteria, suggesting that AK131315 plays an important role in bacterial infections. Altogether, these findings indicate that ISIR regulates LPS-induced inflammation and AK131315 is involved in the pathogenesis of bacterial infections.
    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
    转化生长因子-β(TGF-β)活化激酶1(TAK1),也称为丝裂原活化蛋白激酶7(MAPK7),与TAK1结合蛋白(TAB1,TAB2和TAB3)形成关键的信号复合物,协调关键的生物过程,包括免疫反应,细胞生长,凋亡,和应激反应。刺激激活TAK1,如肿瘤坏死因子α(TNFα),白细胞介素-1β(IL-1β),和Toll样受体(TLRs),强调了它在细胞信号传导中的核心作用。鉴于TAK1结合蛋白(TAK1-TAB)复合物在细胞信号传导中的关键作用及其对各种生物过程的影响,这篇综述旨在了解泛素化如何彻底调节TAK1-TAB复合物。这种理解对于开发针对这种信号通路失调的疾病的靶向治疗至关重要。这项探索意义重大,因为它揭示了对活动的新见解,稳定性,以及建筑群的组装,强调其在疾病调制中的治疗潜力。
    Transforming growth factor-β (TGF-β) activated kinase 1 (TAK1), also named mitogen-activated protein kinase 7 (MAPK7), forms a pivotal signaling complex with TAK1-binding proteins (TAB1, TAB2, and TAB3), orchestrating critical biological processes, including immune responses, cell growth, apoptosis, and stress responses. Activation of TAK1 by stimuli, such as tumor necrosis factor α (TNFα), interleukin-1β (IL-1β), and Toll-like receptors (TLRs), underscores its central role in cellular signaling. Given the critical role of the TAK1-binding protein (TAK1-TAB) complex in cellular signaling and its impact on various biological processes, this review seeks to understand how ubiquitination thoroughly regulates the TAK1-TAB complex. This understanding is vital for developing targeted therapies for diseases where this signaling pathway is dysregulated. The exploration is significant as it unveils new insights into the activity, stability, and assembly of the complex, underscoring its therapeutic potential in disease modulation.
    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:先天免疫系统是宿主防御的第一道防线。转化生长因子-β激活激酶1(TAK1)是先天免疫的关键调节因子。细胞存活,和细胞稳态。因为它在免疫中的重要性,几种病原体已经进化为携带TAK1抑制剂。作为回应,宿主已经进化到感知TAK1抑制并诱导强烈的裂解细胞死亡,全角下垂,由RIPK1-PANoptosome介导。PANoptosis是由先天性免疫传感器启动并由胱天蛋白酶和RIPK驱动的独特的先天性免疫炎性溶解细胞死亡途径。虽然PANoptosis可能有利于清除病原体,过度激活与病理有关。因此,了解调节TAK1抑制剂(TAK1i)诱导的PANoptosis的分子机制对于我们理解RIPK1在健康和疾病中的作用至关重要.
    结果:在这项研究中,通过分析基于细胞死亡的CRISPR筛选结果,我们确定了蛋白磷酸酶6(PP6)全酶成分是TAK1i诱导的PANoptosis的调节因子。PP6酶组分的损失,PPP6C,显著降低TAK1i诱导的PANoptosis。此外,PP6调节亚基PPP6R1,PPP6R2和PPP6R3在调节TAK1i诱导的PANoptosis中具有冗余作用,它们的联合消耗是阻断TAK1i诱导的细胞死亡所必需的。机械上,PPP6C及其调节亚基促进RIPK1的促死亡S166自磷酸化,并导致促存活S321磷酸化减少。
    结论:总体而言,我们的研究结果表明,在激活TAK1i诱导的磷酸酶PP6复合物中,RIPK1依赖的PANoptosis,这表明这种复合物可以在炎症条件下成为治疗目标。
    BACKGROUND: The innate immune system serves as the first line of host defense. Transforming growth factor-β-activated kinase 1 (TAK1) is a key regulator of innate immunity, cell survival, and cellular homeostasis. Because of its importance in immunity, several pathogens have evolved to carry TAK1 inhibitors. In response, hosts have evolved to sense TAK1 inhibition and induce robust lytic cell death, PANoptosis, mediated by the RIPK1-PANoptosome. PANoptosis is a unique innate immune inflammatory lytic cell death pathway initiated by an innate immune sensor and driven by caspases and RIPKs. While PANoptosis can be beneficial to clear pathogens, excess activation is linked to pathology. Therefore, understanding the molecular mechanisms regulating TAK1 inhibitor (TAK1i)-induced PANoptosis is central to our understanding of RIPK1 in health and disease.
    RESULTS: In this study, by analyzing results from a cell death-based CRISPR screen, we identified protein phosphatase 6 (PP6) holoenzyme components as regulators of TAK1i-induced PANoptosis. Loss of the PP6 enzymatic component, PPP6C, significantly reduced TAK1i-induced PANoptosis. Additionally, the PP6 regulatory subunits PPP6R1, PPP6R2, and PPP6R3 had redundant roles in regulating TAK1i-induced PANoptosis, and their combined depletion was required to block TAK1i-induced cell death. Mechanistically, PPP6C and its regulatory subunits promoted the pro-death S166 auto-phosphorylation of RIPK1 and led to a reduction in the pro-survival S321 phosphorylation.
    CONCLUSIONS: Overall, our findings demonstrate a key requirement for the phosphatase PP6 complex in the activation of TAK1i-induced, RIPK1-dependent PANoptosis, suggesting this complex could be therapeutically targeted in inflammatory conditions.
    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
    内质网(ER)形态的动态变化是维持细胞稳态的核心。微管(MT)通过与许多ER成形蛋白复合物协调,促进ER网络连续重塑为薄片和小管,尽管这个过程是如何被细胞外信号控制的仍然是未知的。在这里,我们报道了TAK1,一种对各种生长因子和细胞因子(包括TGF-β和TNF-α)有反应的激酶,通过激活αTAT1触发ER插管,αTAT1是一种MT乙酰化酶,可增强ER滑动。我们表明,这种TAK1/αTAT1依赖性ER重塑通过主动下调BOK来促进细胞存活,ER膜相关的促凋亡效应物。虽然BOK与IP3R复合时通常会受到保护而不会降解,在ER片到小管的转化过程中,它们的解离会迅速降解。这些发现证明了配体诱导的ER重塑的独特机制,并表明TAK1/αTAT1途径可能是ER应激和功能障碍的关键靶标。
    Dynamic changes in the endoplasmic reticulum (ER) morphology are central to maintaining cellular homeostasis. Microtubules (MT) facilitate the continuous remodeling of the ER network into sheets and tubules by coordinating with many ER-shaping protein complexes, although how this process is controlled by extracellular signals remains unknown. Here we report that TAK1, a kinase responsive to various growth factors and cytokines including TGF-β and TNF-α, triggers ER tubulation by activating αTAT1, an MT-acetylating enzyme that enhances ER-sliding. We show that this TAK1/αTAT1-dependent ER remodeling promotes cell survival by actively downregulating BOK, an ER membrane-associated proapoptotic effector. While BOK is normally protected from degradation when complexed with IP3R, it is rapidly degraded upon their dissociation during the ER sheets-to-tubules conversion. These findings demonstrate a distinct mechanism of ligand-induced ER remodeling and suggest that the TAK1/αTAT1 pathway may be a key target in ER stress and dysfunction.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号