NF-kappaB-Inducing Kinase

  • 文章类型: Journal Article
    目的:结肠上皮细胞(CEC)增殖失调是结直肠癌发生发展的关键特征。我们表明NF-κB诱导激酶(NIK)通过协调CEC再生/分化通过非经典NF-κB信号传导减弱结直肠癌,这是经典NF-κB信号传导独特的。
    方法:初步研究评估了隐窝形态/功能,类器官产生,转录组概况,和微生物组。在施用氧化偶氮甲烷和葡聚糖硫酸钠(AOM/DSS)后,全身NIK敲除小鼠(Niki-/-)和条件敲除小鼠开始炎症和炎症诱导的肿瘤发生。
    结果:人类转录组数据显示非规范NF-κB信号传导失调。体外研究评估来自成熟,非分裂CECs和结肠干细胞(CSC)表现出增加的积累和发育迟缓,分别。Nk-/-细胞的转录组学分析揭示了与改变的分化再生相关的基因表达特征。当在体内评估时,在给予DSS的情况下,Nk-/-小鼠表现出更严重的结肠炎和以增加的致结肠微生物群为特征的改变的微生物组。在炎症诱导的肿瘤发生模型中,我们观察到在CEC中NIK被敲除的小鼠中肿瘤负荷和炎症增加(NikACEC)。有趣的是,当NIK在骨髓细胞中被有条件地敲除时(NikAMYE),这没有被概括。令人惊讶的是,骨髓细胞中经典途径的条件性敲除(RelAΔMYE)显示肿瘤负荷和炎症减少,并且在CECs中有条件敲除时没有显着变化(RelAΔCEC)。
    结论:非经典NF-κB信号调节异常以组织依赖性方式与结直肠癌的发生发展相关,并定义了NIK在调节胃肠道炎症和与结直肠癌相关的再生中的关键作用。
    OBJECTIVE: Dysregulated colonic epithelial cell (CEC) proliferation is a critical feature in the development of colorectal cancer. We show that NF-κB-inducing kinase (NIK) attenuates colorectal cancer through coordinating CEC regeneration/differentiation via noncanonical NF-κB signaling that is unique from canonical NF-kB signaling.
    METHODS: Initial studies evaluated crypt morphology/functionality, organoid generation, transcriptome profiles, and the microbiome. Inflammation and inflammation-induced tumorigenesis were initiated in whole-body NIK knockout mice (Nik-/-) and conditional-knockout mice following administration of azoxymethane and dextran sulfate sodium.
    RESULTS: Human transcriptomic data revealed dysregulated noncanonical NF-kB signaling. In vitro studies evaluating Nik-/- crypts and organoids derived from mature, nondividing CECs, and colonic stem cells exhibited increased accumulation and stunted growth, respectively. Transcriptomic analysis of Nik-/- cells revealed gene expression signatures associated with altered differentiation-regeneration. When assessed in vivo, Nik-/- mice exhibited more severe colitis with dextran sulfate sodium administration and an altered microbiome characterized by increased colitogenic microbiota. In the inflammation-induced tumorigenesis model, we observed both increased tumor burdens and inflammation in mice where NIK is knocked out in CECs (NikΔCEC). Interestingly, this was not recapitulated when NIK was conditionally knocked out in myeloid cells (NikΔMYE). Surprisingly, conditional knockout of the canonical pathway in myeloid cells (RelAΔMYE) revealed decreased tumor burden and inflammation and no significant changes when conditionally knocked out in CECs (RelAΔCEC).
    CONCLUSIONS: Dysregulated noncanonical NF-κB signaling is associated with the development of colorectal cancer in a tissue-dependent manner and defines a critical role for NIK in regulating gastrointestinal inflammation and regeneration associated with colorectal cancer.
    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
    这里,我们报告了chr14q32.31-32基因座的复发性局灶性缺失,包括TRAF3,NF-κB信号的负调节因子,新发弥漫性大B细胞淋巴瘤(DLBCL)(24/324例)。综合分析显示TRAF3拷贝数损失与NIK积累之间存在关联,中枢非规范(NC)NF-κB激酶,并增加NCNF-κB途径活性。因此,等基因DLBCL模型系统中的TRAF3基因消融导致NIK上调并增强NCNF-κB下游信号传导。TRAF3缺陷细胞中NIK的敲除或药理学抑制差异损害其增殖和存活,提示对NCNF-κB的获得性肿瘤成瘾。TRAF3消融还导致免疫抑制细胞因子IL-10的分泌加剧。TRAF3缺陷型DLBCL细胞与CD8+T细胞的共培养损害了颗粒酶B和干扰素(IFN)γ的诱导,在中和IL-10后恢复。我们的发现证实了TRAF3遗传改变与NCNF-κB激活之间的直接关系,并强调NIK是DLBCL定义的子集中的潜在治疗靶标。
    Here, we report recurrent focal deletions of the chr14q32.31-32 locus, including TRAF3, a negative regulator of NF-κB signaling, in de novo diffuse large B cell lymphoma (DLBCL) (24/324 cases). Integrative analysis revealed an association between TRAF3 copy number loss with accumulation of NIK, the central noncanonical (NC) NF-κB kinase, and increased NC NF-κB pathway activity. Accordingly, TRAF3 genetic ablation in isogenic DLBCL model systems caused upregulation of NIK and enhanced NC NF-κB downstream signaling. Knockdown or pharmacological inhibition of NIK in TRAF3-deficient cells differentially impaired their proliferation and survival, suggesting an acquired onco-addiction to NC NF-κB. TRAF3 ablation also led to exacerbated secretion of the immunosuppressive cytokine IL-10. Coculturing of TRAF3-deficient DLBCL cells with CD8+ T cells impaired the induction of Granzyme B and interferon (IFN) γ, which were restored following neutralization of IL-10. Our findings corroborate a direct relationship between TRAF3 genetic alterations and NC NF-κB activation, and highlight NIK as a potential therapeutic target in a defined subset of DLBCL.
    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
    炎症消退是预防慢性炎性疾病发展的重要过程。然而,调节银屑病炎症消退的机制尚不清楚.这里,我们报告ANKRD22是银屑病样炎症的内源性负协调器,因为ANKRD22缺陷型小鼠更容易受到IMQ诱导的银屑病样炎症的影响。机械上,ANKRD22缺陷导致TNFRII-NIK介导的非经典NF-κB信号通路过度激活,导致DC中IL-23的过度产生。这是由于ANKRD22是NIK的负反馈调节器,因为它在物理上与积累的NIK结合并协助降解。临床上,ANKRD22与IL23A表达和银屑病严重程度呈负相关。更重要的是,皮下施用携带ANKRD22过表达载体的AAV有效地加速了银屑病样皮肤炎症的消退。我们的发现表明ANKRD22是NIK的内源性监督者,负责牛皮癣的炎症消退,并且可以在银屑病治疗的背景下进行探索。
    Inflammation resolution is an essential process for preventing the development of chronic inflammatory diseases. However, the mechanisms that regulate inflammation resolution in psoriasis are not well understood. Here, we report that ANKRD22 is an endogenous negative orchestrator of psoriasiform inflammation because ANKRD22-deficient mice are more susceptible to IMQ-induced psoriasiform inflammation. Mechanistically, ANKRD22 deficiency leads to excessive activation of the TNFRII-NIK-mediated noncanonical NF-κB signaling pathway, resulting in the hyperproduction of IL-23 in DCs. This is due to ANKRD22 being a negative feedback regulator for NIK because it physically binds to and assists in the degradation of accumulated NIK. Clinically, ANKRD22 is negatively associated with IL-23A expression and psoriasis severity. Of greater significance, subcutaneous administration of an AAV carrying ANKRD22-overexpression vector effectively hastens the resolution of psoriasiform skin inflammation. Our findings suggest ANKRD22, an endogenous supervisor of NIK, is responsible for inflammation resolution in psoriasis, and may be explored in the context of psoriasis therapy.
    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
    胶质母细胞瘤(GBM)是一种高度侵袭性的脑癌,预后差,治疗选择有限。ALK和c-MET抑制剂Crizotinib已证明对新诊断的GBM具有临床前治疗潜力,虽然其功效受到血脑屏障穿透不良的限制。这里,我们确定克唑替尼为核因子-κB(NF-κB)诱导激酶的新型抑制剂,这是GBM生长和增殖的关键调节剂。我们进一步表明,克唑替尼与七甲胺花青染料的缀合,或近红外染料(IR-Crizotinib),与未结合的克唑替尼相比,体外减轻了神经胶质瘤细胞的增殖和存活。此外,我们观察到IR-Crizotinib在原位小鼠异种移植GBM肿瘤中的定位增加,导致体内肿瘤生长受损。总的来说,IR-Crizotinib表现出改善的颅内化疗递送和肿瘤定位,同时抑制NIK和非典型NF-κB信号,从而减少体外神经胶质瘤的生长,以及在体内,并增加临床前啮齿动物模型的存活率。
    Glioblastoma (GBM) is a highly aggressive form of brain cancer with a poor prognosis and limited treatment options. The ALK and c-MET inhibitor Crizotinib has demonstrated preclinical therapeutic potential for newly diagnosed GBM, although its efficacy is limited by poor penetration of the blood brain barrier. Here, we identify Crizotinib as a novel inhibitor of nuclear factor-κB (NF-κB)-inducing kinase, which is a key regulator of GBM growth and proliferation. We further show that the conjugation of Crizotinib to a heptamethine cyanine dye, or a near-infrared dye (IR-Crizotinib), attenuated glioma cell proliferation and survival in vitro to a greater extent than unconjugated Crizotinib. Moreover, we observed increased IR-Crizotinib localization to orthotopic mouse xenograft GBM tumors, which resulted in impaired tumor growth in vivo. Overall, IR-Crizotinib exhibited improved intracranial chemotherapeutic delivery and tumor localization with concurrent inhibition of NIK and noncanonical NF-κB signaling, thereby reducing glioma growth in vitro, as well as in vivo, and increasing survival in a preclinical rodent model.
    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
    由常染色体隐性遗传AIRE缺乏症引起的1型自身免疫性多内分泌病综合征(APS-1)患者会产生中和I型干扰素(IFN)1,2的自身抗体,从而易患危及生命的COVID-19肺炎3。在这里,我们报道了常染色体隐性遗传性NIK或RELB缺乏的患者,或常染色体显性遗传NF-κB2缺陷的特定类型,还具有针对I型IFN的中和自身抗体,并且患危及生命的COVID-19肺炎的风险更高。在常染色体显性遗传NF-κB2缺乏症患者中,这些自身抗体仅在与由于p100加工产生p52而导致的转录(p52活性)功能丧失(LOF)和由于未加工p100的积累而导致的调节(IκBδ活性)功能获得(GOF),因此增加了IκBδ的抑制活性(下文,p52LOF/IκBδGOF)。相比之下,针对I型IFN的中和自身抗体在NFKB2变体杂合子导致p100和p52单倍体不足的个体中未发现(以下,p52LOF/IκBδLOF)或p52的功能获得(以下,p52GOF/IκBδLOF)。与APS-1患者相反,患有NIK疾病的患者,RELB或NF-κB2具有非常少的组织特异性自身抗体。然而,它们的胸腺结构异常,几乎没有表达AIRE的髓质胸腺上皮细胞。替代NF-κB途径的人类先天性错误会损害表达AIRE的髓样胸腺上皮细胞的发育,从而产生针对I型IFN的自身抗体和对病毒性疾病的易感性。
    Patients with autoimmune polyendocrinopathy syndrome type 1 (APS-1) caused by autosomal recessive AIRE deficiency produce autoantibodies that neutralize type I interferons (IFNs)1,2, conferring a predisposition to life-threatening COVID-19 pneumonia3. Here we report that patients with autosomal recessive NIK or RELB deficiency, or a specific type of autosomal-dominant NF-κB2 deficiency, also have neutralizing autoantibodies against type I IFNs and are at higher risk of getting life-threatening COVID-19 pneumonia. In patients with autosomal-dominant NF-κB2 deficiency, these autoantibodies are found only in individuals who are heterozygous for variants associated with both transcription (p52 activity) loss of function (LOF) due to impaired p100 processing to generate p52, and regulatory (IκBδ activity) gain of function (GOF) due to the accumulation of unprocessed p100, therefore increasing the inhibitory activity of IκBδ (hereafter, p52LOF/IκBδGOF). By contrast, neutralizing autoantibodies against type I IFNs are not found in individuals who are heterozygous for NFKB2 variants causing haploinsufficiency of p100 and p52 (hereafter, p52LOF/IκBδLOF) or gain-of-function of p52 (hereafter, p52GOF/IκBδLOF). In contrast to patients with APS-1, patients with disorders of NIK, RELB or NF-κB2 have very few tissue-specific autoantibodies. However, their thymuses have an abnormal structure, with few AIRE-expressing medullary thymic epithelial cells. Human inborn errors of the alternative NF-κB pathway impair the development of AIRE-expressing medullary thymic epithelial cells, thereby underlying the production of autoantibodies against type I IFNs and predisposition to viral diseases.
    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
    暂无摘要。
    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
    高级别胶质瘤的预后,如多形性胶质母细胞瘤(GBM),由于这些侵袭性癌症的高度侵袭性,它非常差。先前的工作表明,非典型NF-κB途径的TNF弱样因子(TWEAK)诱导以NF-κB诱导激酶(NIK)依赖性方式促进GBM细胞的侵袭性。虽然NIK活性主要在翻译后水平受到调控,我们在这里显示NIK(MAP3K14)在入侵细胞群体中的转录水平上调,在最具侵袭性的细胞中观察到最高的NIK表达。高诱导NIK基因表达的GBM细胞表现出集体侵袭的特征,促进邻近细胞的入侵。此外,我们证明E2F转录因子E2F4和E2F5直接调节NIK转录,并且是响应TWEAK促进GBM细胞侵袭所必需的。总的来说,我们的发现表明,NIK的转录诱导促进集体细胞迁移和侵袭,从而促进GBM发病机制。
    The prognosis of high-grade gliomas, such as glioblastoma multiforme (GBM), is extremely poor due to the highly invasive nature of these aggressive cancers. Previous work has demonstrated that TNF-weak like factor (TWEAK) induction of the noncanonical NF-κB pathway promotes the invasiveness of GBM cells in an NF-κB-inducing kinase (NIK)-dependent manner. While NIK activity is predominantly regulated at the posttranslational level, we show here that NIK (MAP3K14) is upregulated at the transcriptional level in invading cell populations, with the highest NIK expression observed in the most invasive cells. GBM cells with high induction of NIK gene expression demonstrate characteristics of collective invasion, facilitating invasion of neighboring cells. Furthermore, we demonstrate that the E2F transcription factors E2F4 and E2F5 directly regulate NIK transcription and are required to promote GBM cell invasion in response to TWEAK. Overall, our findings demonstrate that transcriptional induction of NIK facilitates collective cell migration and invasion, thereby promoting GBM pathogenesis.
    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
    NF-κB诱导激酶(NIK),这对于非经典NF-κB通路的激活至关重要,调节免疫的不同过程,发展,和疾病。尽管最近的研究已经阐明了NIK在适应性免疫细胞和癌细胞代谢中的重要功能,NIK在先天免疫细胞代谢驱动的炎症反应中的作用尚不清楚.在这项研究中,我们证明,小鼠NIK缺陷的骨髓源性巨噬细胞表现出线粒体依赖性代谢和氧化磷酸化的缺陷,这损害了维修的获得,抗炎表型。随后,NIK缺陷小鼠表现出以异常嗜酸性粒细胞为特征的骨髓细胞的偏斜,单核细胞,和血液中的巨噬细胞群,骨髓,和脂肪组织。此外,缺乏NIK的血液单核细胞显示对细菌LPS的高反应性和离体TNF-α产生升高。这些发现表明NIK控制着代谢重新布线,这对于平衡促炎和抗炎髓样免疫细胞功能至关重要。总的来说,我们的工作强调了NIK作为一种分子变阻器以前未被认识到的作用,它在先天免疫中精细调节免疫代谢,并提示代谢功能障碍可能是由NIK表达或活性异常引起的炎症性疾病的重要驱动因素。
    NF-κB-inducing kinase (NIK), which is essential for the activation of the noncanonical NF-κB pathway, regulates diverse processes in immunity, development, and disease. Although recent studies have elucidated important functions of NIK in adaptive immune cells and cancer cell metabolism, the role of NIK in metabolic-driven inflammatory responses in innate immune cells remains unclear. In this study, we demonstrate that murine NIK-deficient bone marrow-derived macrophages exhibit defects in mitochondrial-dependent metabolism and oxidative phosphorylation, which impair the acquisition of a prorepair, anti-inflammatory phenotype. Subsequently, NIK-deficient mice exhibit skewing of myeloid cells characterized by aberrant eosinophil, monocyte, and macrophage cell populations in the blood, bone marrow, and adipose tissue. Furthermore, NIK-deficient blood monocytes display hyperresponsiveness to bacterial LPS and elevated TNF-α production ex vivo. These findings suggest that NIK governs metabolic rewiring, which is critical for balancing proinflammatory and anti-inflammatory myeloid immune cell function. Overall, our work highlights a previously unrecognized role for NIK as a molecular rheostat that fine-tunes immunometabolism in innate immunity, and suggests that metabolic dysfunction may be an important driver of inflammatory diseases caused by aberrant NIK expression or activity.
    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
    神经前体细胞的接合表达,发育下调8(NEDD8)至靶蛋白,称为Neddylation,参与许多细胞过程,并在各种病理疾病中异常。它与肝功能和衰竭的相关性仍然知之甚少。在这里,我们显示了NAE1的表达失调,NAE1是唯一的NEDD8E1酶的调节亚基,在人类急性肝衰竭中。在肝细胞中NAE1的胚胎和成人发作缺失导致肝细胞死亡,炎症,和纤维化,最终导致小鼠致命性肝损伤。肝Neddylation缺乏引发氧化应激,线粒体功能障碍,和肝细胞重编程,增强肝损伤。重要的是,NF-κB诱导激酶(NIK),丝氨酸/Thr激酶,是Neddylation底物。NIK的Neddylation促进其泛素化和降解。相反,抑制neddylation会导致异常的NIK激活,加重肝细胞损伤和炎症。N-乙酰半胱氨酸的给药,谷胱甘肽替代品和抗氧化剂,减轻成年雄性小鼠肝脏NAE1缺失引起的肝衰竭。因此,肝内化在维持出生后和成人肝脏稳态方面很重要,和确定的Neddylation靶标/途径提供了对治疗性干预急性肝功能衰竭的见解。
    The conjugation of neural precursor cell expressed, developmentally downregulated 8 (NEDD8) to target proteins, termed neddylation, participates in many cellular processes and is aberrant in various pathological diseases. Its relevance to liver function and failure remains poorly understood. Herein, we show dysregulated expression of NAE1, a regulatory subunit of the only NEDD8 E1 enzyme, in human acute liver failure. Embryonic- and adult-onset deletion of NAE1 in hepatocytes causes hepatocyte death, inflammation, and fibrosis, culminating in fatal liver injury in mice. Hepatic neddylation deficiency triggers oxidative stress, mitochondrial dysfunction, and hepatocyte reprogramming, potentiating liver injury. Importantly, NF-κB-inducing kinase (NIK), a serine/Thr kinase, is a neddylation substrate. Neddylation of NIK promotes its ubiquitination and degradation. Inhibition of neddylation conversely causes aberrant NIK activation, accentuating hepatocyte damage and inflammation. Administration of N-acetylcysteine, a glutathione surrogate and antioxidant, mitigates liver failure caused by hepatic NAE1 deletion in adult male mice. Therefore, hepatic neddylation is important in maintaining postnatal and adult liver homeostasis, and the identified neddylation targets/pathways provide insights into therapeutically intervening acute liver failure.
    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
    目标:非酒精性脂肪性肝病(NAFLD)的范围从脂肪变性到非酒精性脂肪性肝炎(NASH),通常通过很大程度上不确定的机制进展为肝细胞癌(HCC)。NASH和HCC依赖于炎症信号,其主要调节因子是NFκB转录因子家族,由规范和非规范途径激活。
    方法:这里,我们研究了NASH中的非经典NFκB诱导激酶(NIK/MAP3K14),NASH向HCC过渡,和DEN诱导的HCC。为此,我们在小鼠中进行饮食和化学干预,通过单核测序进行分析,基因表达和组织化学方法。最终,我们在人类患者样本中验证了我们的小鼠结果.
    结果:我们发现肝细胞特异性NIK缺乏症(NIKLKO)改善了NASH代谢并发症并减少了肝癌的发生,独立于其在NFκB途径中的作用。相反,肝NIK减弱肝保护性JAK2/STAT5信号传导,这是小鼠和人类NASH和NASH进展为HCC的先决条件。
    结论:我们的数据表明NIK介导的抑制性JAK2在633丝氨酸磷酸化,这可能适合于患者未来的治疗干预。
    Nonalcoholic fatty liver disease (NAFLD) ranges from steatosis to nonalcoholic steatohepatitis (NASH), which often progresses to hepatocellular carcinoma (HCC) through a largely undefined mechanism. NASH and HCC depend on inflammatory signaling, whose master regulator is the NFκB transcription factor family, activated by canonical and non-canonical pathways.
    Here, we investigated non-canonical NFκB-inducing kinase (NIK/MAP3K14) in metabolic NASH, NASH to HCC transition, and DEN-induced HCC. To this end, we performed dietary and chemical interventions in mice that were analyzed via single nucleus sequencing, gene expression and histochemical methods. Ultimately, we verified our mouse results in human patient samples.
    We revealed that hepatocyte-specific NIK deficiency (NIKLKO) ameliorated metabolic NASH complications and reduced hepatocarcinogenesis, independent of its role in the NFκB pathway. Instead, hepatic NIK attenuated hepatoprotective JAK2/STAT5 signaling that is a prerequisite for NASH and NASH to HCC progression in mice and humans.
    Our data suggest NIK-mediated inhibitory JAK2 phosphorylation at serine 633 that might be amenable for future therapeutic interventions in patients.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号