TNFR

TNFR
  • 文章类型: Journal Article
    激动剂抗体正被用于从神经退行性疾病到癌症的治疗应用。对于肿瘤坏死因子(TNF)受体超家族,三个或更多受体的高阶聚类是它们激活的关键,这可以使用识别两个独特表位的抗体来实现。然而,双表位的产生(即,双特异性)抗体通常需要动物免疫,并且费力且不可预测。这里,我们报道了一种鉴定能有效激活TNF受体的双表位抗体的简单方法,无需额外的动物免疫.我们的方法使用现有的,受体特异性IgG,缺乏内在的激动剂活性,来阻断它们相应的表位,然后选择结合可接近表位的单链抗体。将选择的抗体与IgG的轻链融合以产生人四价抗体。我们通过将针对OX40和CD137(4-1BB)的几种临床阶段抗体转化为具有有效激动剂活性的双表位抗体来强调这种方法的广泛用途。
    Agonist antibodies are being pursued for therapeutic applications ranging from neurodegenerative diseases to cancer. For the tumor necrosis factor (TNF) receptor superfamily, higher-order clustering of three or more receptors is key to their activation, which can be achieved using antibodies that recognize two unique epitopes. However, the generation of biepitopic (i.e., biparatopic) antibodies typically requires animal immunization and is laborious and unpredictable. Here, we report a simple method for identifying biepitopic antibodies that potently activate TNF receptors without the need for additional animal immunization. Our approach uses existing, receptor-specific IgGs, which lack intrinsic agonist activity, to block their corresponding epitopes, then selects single-chain antibodies that bind accessible epitopes. The selected antibodies are fused to the light chains of IgGs to generate human tetravalent antibodies. We highlight the broad utility of this approach by converting several clinical-stage antibodies against OX40 and CD137 (4-1BB) into biepitopic antibodies with potent agonist 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
    肿瘤坏死因子(TNF)是一种重要的细胞因子,可以通过结合肿瘤坏死因子受体(TNFR)来调节多种细胞反应。我们研究了中华绒螯蟹的TNF是否可以调节血细胞的增殖。结果表明,EsTNF和EsTNFR在所有受试组织中均呈组成型表达,包括心脏,肝胰腺,肌肉,ill,胃,肠子,和血细胞。我们发现血细胞中存在低水平的EsTNF和EsTNFR转录物。受金黄色葡萄球菌或副溶血性弧菌刺激后,血细胞中的基因表达水平显着增加。我们还发现一些与细胞增殖相关的基因在脉冲rEsTNF刺激的血细胞中表达水平高于对照组。我们还用RNAi技术击倒了EsTNFR基因。结果表明,当TNF不结合TNFR时,与对照组相比,这些与细胞增殖相关的基因的表达水平明显下调。我们使用Edu技术重复上述实验,结果相似。与对照组相比,rEsTNF刺激的血细胞表现出更显著的增殖,敲除EsTNFR基因后增殖率显著下调。因此,我们表明,TNF结合TNFR可以影响E.sinensis血细胞的增殖,这可能表现为影响一些增殖相关基因的表达。
    Tumor necrosis factor (TNF) is an important cytokine that can regulate a variety of cellular responses by binding tumor necrosis factor receptor (TNFR). We studied whether the TNF of Eriocheir sinensis can regulate hemocyte proliferation. The results showed that the EsTNF and EsTNFR were constitutively expressed in all tested tissues, including the heart, hepatopancreas, muscles, gills, stomachs, intestines, and hemocytes. We found that low levels of EsTNF and EsTNFR transcripts were present in hemocytes. The gene expression levels were significantly increased in the hemocytes after being stimulated by Staphylococcus aureus or Vibrio parahaemolyticus. We also found some genes related to cell proliferation were expressed at a higher level in pulsing rTNF-stimulated hemocytes compared with the control group. We also knocked down the EsTNFR gene with RNAi technology. The results showed that the expression level of these genes related to cell proliferation was significantly down-regulated compared with the control group when the TNF does not bind TNFR. We used Edu technology to repeat the above experiments and the results were similar. Compared with the control group, the hemocytes stimulated by rTNF showed more significant proliferation, and the proliferation rate was significantly down-regulated after knocking down the EsTNFR gene. Therefore, we indicate that TNF binding TNFR can affect the proliferation of E. sinensis hemocytes, which might be manifested by affecting the expression of some proliferation-related genes.
    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
    在这项研究中,我们检查了使用各种刺激的儿童MOGAD患者(n=4,未经治疗的缓解样本)和健康对照(n=5)的CD4T细胞活化,了解抗原特异性和旁观者机制如何促进MOGAD中CD4T细胞活化。TNFα,发现IL6和MOG肽池通过使用免疫标记测量调节因子(A20,IκBα)和磷酸化亚基(磷酸-p65和磷酸-STAT3)的表达来激活NF-κB或STAT3途径。泼尼松龙逆转NF-κB和STAT3的激活,并增加A20和IκBα的表达。在某些CD4T细胞亚群中,TNFR阻断部分逆转NF-κB活化,但不影响STAT3激活。我们观察到响应于各种刺激的NF-κB和STAT3的激活在MOGAD(缓解)和HC中表现基本相同。IL6刺激导致MOGAD患者在75分钟时更高的STAT3磷酸化,特别是在中枢和效应记忆CD4T细胞中(具有未调整的p值)。这些发现提示MOGAD中NF-κB和STAT3途径的潜在治疗靶向。需要进一步的研究来验证延长STAT3磷酸化的重要性及其与IL6受体阻滞剂治疗反应的相关性。
    In this study, we examined CD4 T cell activation using various stimuli in pediatric MOGAD patients (n = 4, untreated remission samples) and healthy controls (n = 5), to understand how both antigen-specific and bystander mechanisms contribute to CD4 T cell activation in MOGAD. TNFα, IL6, and MOG peptide pool were found to activate NF-κB or STAT3 pathways by measuring the expression of regulators (A20, IκBα) and phosphorylated subunits (phospho-p65 and phospho-STAT3) using immunolabeling. Prednisolone reversed activation of both NF-κB and STAT3 and increased the expression of A20 and IκBα. TNFR blocking partially reversed NF-κB activation in certain CD4 T cell subsets, but did not effect STAT3 activation. We observed that activation of NF-κB and STAT3 in response to various stimuli behaves mostly same in MOGAD (remission) and HC. IL6 stimulation resulted in higher STAT3 phosphorylation in MOGAD patients at 75 min, specifically in central and effector memory CD4 T cells (with unadjusted p-values). These findings suggest the potential therapeutic targeting of NF-κB and STAT3 pathways in MOGAD. Further investigation is needed to validate the significance of extended STAT3 phosphorylation and its correlation with IL6 receptor blocker treatment response.
    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
    星形胶质细胞有助于各种神经退行性疾病的慢性神经炎症,包括帕金森病(PD),最常见的运动障碍。然而,星形胶质细胞在神经炎症中的确切作用尚不完全清楚.在这里,我们发现G蛋白信号调节因子5(RGS5)通过增强星形细胞肿瘤坏死因子受体(TNFR)信号促进神经退行性过程.我们发现星形胶质细胞中Rgs5的选择性消融会抑制细胞因子的产生,从而减轻PD动物模型中的神经炎症反应和神经元存活,而Rgs5的过表达具有相反的作用。机械上,RGS5通过与受体TNFR2结合,将星形胶质细胞从神经保护特性转变为促炎特性。RGS5还通过与受体TNFR1相互作用来增强TNFR信号传导介导的促炎反应。此外,通过RGS5aa1-108或小分子化合物feshurin和butein中断RGS5/TNFR相互作用,抑制星形细胞细胞因子的产生。我们表明星形胶质细胞RGS5的转录受转录因子早期B细胞因子1的控制,其表达受RGS5调节的TNF信号传导的相互影响。因此,我们的研究表明,除了其在G蛋白偶联受体信号传导中的传统作用外,星形胶质细胞RGS5是TNF信号传导回路的关键调节剂,并激活星形胶质细胞,从而导致慢性神经炎症。星形胶质细胞RGS5/TNFR相互作用的阻断是神经炎症相关神经退行性疾病的潜在治疗策略。
    Astrocytes contribute to chronic neuroinflammation in a variety of neurodegenerative diseases, including Parkinson\'s disease (PD), the most common movement disorder. However, the precise role of astrocytes in neuroinflammation remains incompletely understood. Herein, we show that regulator of G-protein signaling 5 (RGS5) promotes neurodegenerative process through augmenting astrocytic tumor necrosis factor receptor (TNFR) signaling. We found that selective ablation of Rgs5 in astrocytes caused an inhibition in the production of cytokines resulting in mitigated neuroinflammatory response and neuronal survival in animal models of PD, whereas overexpression of Rgs5 had the opposite effects. Mechanistically, RGS5 switched astrocytes from neuroprotective to pro-inflammatory property via binding to the receptor TNFR2. RGS5 also augmented TNFR signaling-mediated pro-inflammatory response by interacting with the receptor TNFR1. Moreover, interrupting RGS5/TNFR interaction by either RGS5 aa 1-108 or small molecular compounds feshurin and butein, suppressed astrocytic cytokine production. We showed that the transcription of astrocytic RGS5 was controlled by transcription factor early B cell factor 1 whose expression was reciprocally influenced by RGS5-modulated TNF signaling. Thus, our study indicates that beyond its traditional role in G-protein coupled receptor signaling, astrocytic RGS5 is a key modulator of TNF signaling circuit with resultant activation of astrocytes thereby contributing to chronic neuroinflammation. Blockade of the astrocytic RGS5/TNFR interaction is a potential therapeutic strategy for neuroinflammation-associated neurodegenerative 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
    免疫细胞上的共刺激受体代表了免疫治疗的有吸引力的目标,因为这些分子可以增加个体保护性免疫细胞群的频率和它们的寿命。以及增强各种效应器功能。4-1BB,TNF受体超家族的成员,也称为CD137和TNFRSF9,是一种可在几种细胞类型上诱导的分子,包括T细胞和NK细胞。动物模型中的临床前研究已经验证了用激动剂试剂或其天然配体刺激4-1BB的观点可用于增强常规T细胞和NK细胞免疫以防止肿瘤生长和病毒感染。此外,刺激4-1BB可增强调节性T细胞功能,在抑制自身免疫的正确背景下可能有用.已经生产了两种针对4-1BB的人类激动剂抗体,并在癌症的临床试验中进行了测试。具有可变的结果,导致大量第二代抗体构建体的产生,包括双细节和多细节,希望优化活性和选择性。这里,我们回顾了迄今为止4-1BB激动的进展,讨论适当靶向免疫系统以引发所需活性的并发症,连同工程参与者的挑战,并强调了在传染病和自身免疫中操纵这种分子的未开发潜力。
    Costimulatory receptors on immune cells represent attractive targets for immunotherapy given that these molecules can increase the frequency of individual protective immune cell populations and their longevity, as well as enhance various effector functions. 4-1BB, a member of the TNF receptor superfamily, also known as CD137 and TNFRSF9, is one such molecule that is inducible on several cell types, including T cells and NK cells. Preclinical studies in animal models have validated the notion that stimulating 4-1BB with agonist reagents or its natural ligand could be useful to augment conventional T cell and NK cell immunity to protect against tumor growth and against viral infection. Additionally, stimulating 4-1BB can enhance regulatory T cell function and might be useful in the right context for suppressing autoimmunity. Two human agonist antibodies to 4-1BB have been produced and tested in clinical trials for cancer, with variable results, leading to the production of a wealth of second-generation antibody constructs, including bi- and multi-specifics, with the hope of optimizing activity and selectivity. Here, we review the progress to date in agonism of 4-1BB, discuss the complications in targeting the immune system appropriately to elicit the desired activity, together with challenges in engineering agonists, and highlight the untapped potential of manipulating this molecule in infectious disease and autoimmunity.
    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:体外发育异常蛋白A(EDA),皮肤特异性TNF配体,与其膜受体EDAR相互作用,触发皮肤附件形成中的EDA信号传导。EDA信号中的基因突变导致无汗性/无汗性外胚层发育不良(A/HED),影响皮肤附属物的形成,包括头发,牙齿,和几个外分泌腺。
    结果:我们报道EDA触发其受体EDAR从胞质区室到质膜的易位。我们使用蛋白质亲和纯化来显示EDA刺激后,EDAR与SNAP23-STX6-VAMP1/2/3囊泡运输复合物相关。我们发现EDA依赖性PKA激活对于这种关联至关重要。值得注意的是,两个HED连锁EDAR突变中的任何一个,T346M和R420W,预防EDA诱导的EDAR易位;并且EDA诱导的PKA激活和SNAP23都是皮肤附件模型中睑板腺(MG)生长所必需的。
    结论:总体而言,在一种新的调节机制中,EDA增加其自身受体EDAR的质膜易位,增强皮肤附件形成中的EDA-EDAR信号传导。我们的发现还提供了PKA和SNAP23作为HED干预的潜在目标。
    BACKGROUND: Ectodysplasin-A (EDA), a skin-specific TNF ligand, interacts with its membrane receptor EDAR to trigger EDA signaling in skin appendage formation. Gene mutations in EDA signaling cause Anhidrotic/Hypohidrotic Ectodermal Dysplasia (A/HED), which affects the formation of skin appendages including hair, teeth, and several exocrine glands.
    RESULTS: We report that EDA triggers the translocation of its receptor EDAR from a cytosolic compartment into the plasma membrane. We use protein affinity purification to show that upon EDA stimulation EDAR associates with SNAP23-STX6-VAMP1/2/3 vesicle trafficking complexes. We find that EDA-dependent PKA activation is critical for the association. Notably, either of two HED-linked EDAR mutations, T346M and R420W, prevents EDA-induced EDAR translocation; and both EDA-induced PKA activation and SNAP23 are required for Meibomian gland (MG) growth in a skin appendage model.
    CONCLUSIONS: Overall, in a novel regulatory mechanism, EDA increases plasma membrane translocation of its own receptor EDAR, augmenting EDA-EDAR signaling in skin appendage formation. Our findings also provide PKA and SNAP23 as potential targets for the intervention of HED.
    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
    肿瘤坏死因子超家族(TNFSF)及其受体(TNFRSF)是免疫系统的重要调节因子,介导增殖,生存,分化,和免疫细胞的功能。因此,它们的靶向免疫疗法很有吸引力,虽然到目前为止,开发不足。在这篇综述中,我们讨论了TNFRSF的共刺激成员在最佳免疫应答产生中的重要性,靶向这些受体进行免疫治疗的基本原理,在临床前研究中成功靶向它们,以及将这种成功转化为临床的挑战。讨论了当前可用药物的功效和局限性,以及旨在克服当前问题的下一代免疫刺激剂的开发。并利用这种受体类来提供有效的,持久和安全的药物为患者。
    The tumor necrosis factor superfamily (TNFSF) and their receptors (TNFRSF) are important regulators of the immune system, mediating proliferation, survival, differentiation, and function of immune cells. As a result, their targeting for immunotherapy is attractive, although to date, under-exploited. In this review we discuss the importance of co-stimulatory members of the TNFRSF in optimal immune response generation, the rationale behind targeting these receptors for immunotherapy, the success of targeting them in pre-clinical studies and the challenges in translating this success into the clinic. The efficacy and limitations of the currently available agents are discussed alongside the development of next generation immunostimulatory agents designed to overcome current issues, and capitalize on this receptor class to deliver potent, durable and safe drugs for patients.
    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
    The tumor necrosis factor alpha (TNF-α)-TNF-α receptor (TNFR) interaction plays a central role in the pathogenesis of various autoimmune diseases, particularly rheumatoid arthritis, and is therefore considered a key target for drug discovery. However, natural compounds that can specifically block the TNF-α-TNFR interaction are rarely reported. (-)-Epigallocatechin-3-gallate (EGCG) is the most active, abundant, and thoroughly investigated polyphenolic compound in green tea. However, the molecular mechanism by which EGCG ameliorates autoimmune arthritis remains to be elucidated. In the present study, we found that EGCG can directly bind to TNF-α, TNFR1, and TNFR2 with similar μM affinity and disrupt the interactions between TNF-α and TNFR1 and TNFR2, which inhibits TNF-α-induced L929 cell death, blocks TNF-α-induced NF-κB activation in 293-TNF-α response cell line, and eventually leads to inhibition of TNF-α-induced NF-κB signaling pathway in HFLS and MH7A cells. Thus, regular consumption of EGCG in green tea may represent a potential therapeutic agent for the treatment of TNF-α-associated diseases.
    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
    诱导肿瘤细胞特异性凋亡的能力代表了TNF受体(TNFR)家族成员CD40的最独特特征。最近对正常和恶性上皮细胞中由其膜呈递配体CD40L(mCD40L)触发的信号传导事件的研究已开始阐明CD40功能作用的精致背景和细胞类型特异性。这里,我们证明,与其他癌症相比,mCD40L在结直肠癌(CRC)细胞中触发了显着更快的凋亡,其具有夹带两个同时运行的信号轴的能力。CD40连接最初激活TNFR相关因子3(TRAF3),随后激活NADPH氧化酶(NOX)/凋亡信号调节激酶1(ASK1)信号传导和诱导活性氧(ROS)以介导p38/JNK和ROS依赖性细胞死亡。在这一点上,p38/JNK信号直接激活线粒体途径,并触发细胞内TNF相关的凋亡诱导配体(TRAIL)的快速诱导,该配体从内部区室发出信号以启动外源性caspase-10相关的凋亡,导致截短的Bid(tBid)激活的线粒体信号。p38和JNK对于直接的线粒体凋亡诱导和TRAIL/caspase-10/tBid途径都是必不可少的,但是它们的参与遵循功能层次和时间控制的相互作用,因为p38功能是JNK磷酸化所必需的。通过参与内在和外在途径,通过两个信号同时激活细胞凋亡,CD40可以加速CRC细胞死亡。我们的发现进一步揭示了CD40/mCD40L二元结构的多方面特性,新型TNFR串扰加速肿瘤细胞特异性死亡,并且可能对使用CD40作为治疗靶标有影响。
    The capacity to induce tumour-cell specific apoptosis represents the most unique feature of the TNF receptor (TNFR) family member CD40. Recent studies on the signalling events triggered by its membrane-presented ligand CD40L (mCD40L) in normal and malignant epithelial cells have started to unravel an exquisite context and cell type specificity for the functional effects of CD40. Here, we demonstrate that, in comparison to other carcinomas, mCD40L triggered strikingly more rapid apoptosis in colorectal carcinoma (CRC) cells, underpinned by its ability to entrain two concurrently operating signalling axes. CD40 ligation initially activates TNFR-associated factor 3 (TRAF3) and subsequently NADPH oxidase (NOX)/Apoptosis signal-regulating kinase 1 (ASK1)-signalling and induction of reactive oxygen species (ROS) to mediate p38/JNK- and ROS-dependent cell death. At that point, p38/JNK signalling directly activates the mitochondrial pathway, and triggers rapid induction of intracellular TNF-related apoptosis-inducing ligand (TRAIL) that signals from internal compartments to initiate extrinsic caspase-10-asscociated apoptosis, leading to truncated Bid (tBid)-activated mitochondrial signalling. p38 and JNK are essential both for direct mitochondrial apoptosis induction and the TRAIL/caspase-10/tBid pathway, but their involvement follows functional hierarchy and temporally controlled interplay, as p38 function is required for JNK phosphorylation. By engaging both intrinsic and extrinsic pathways to activate apoptosis via two signals simultaneously, CD40 can accelerate CRC cell death. Our findings further unravel the multi-faceted properties of the CD40/mCD40L dyad, highlighted by the novel TNFR crosstalk that accelerates tumour cell-specific death, and may have implications for the use of CD40 as a therapeutic target.
    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
    促炎细胞因子肿瘤坏死因子(TNF)是肠道炎症性疾病的主要介导因子之一。影响紧密连接(TJ)蛋白的组成并导致上皮屏障的破坏。完整的肠屏障是强制性的,因为Peyer’s斑块的卵泡相关上皮代表了肠道免疫系统的第一道防线,并确保了来自肠腔的抗原的受控摄取。在目前的研究中,我们通过应用Ussing室技术分析了TNF对猪Peyer’s斑块卵泡相关上皮的详细影响。将Peyer的斑块和周围绒毛上皮的上皮组织标本固定在常规的Ussing室中,并与TNF孵育10小时。代表组织的上皮屏障功能,被记录下来。8小时后,在Peyer的贴片组织标本中检测到跨上皮电阻的降低,而绒毛上皮不受TNF的显著影响。随后对TJ蛋白表达的分子分析显示claudin-1和-4的显着减少,而claudin-2的增加。在邻近的绒毛上皮中,TJ蛋白表达无明显变化。在Peyer的斑块中也可以检测到TNF受体-2(TNFR-2)的强烈增加,与该受体在Peyer斑块中的主要作用一致。我们的发现与共聚焦激光扫描免疫荧光显微镜检测到的变化一致。在阻断实验中分析了经由肌球蛋白轻链激酶(MLCK)的TNF效应的调节。我们的详细分析首次表明TNF会影响猪Peyer's斑块的卵泡相关上皮的屏障功能,但对绒毛上皮没有影响。这些发现不仅揭示了两种结构之间上皮屏障功能的基本差异,而且Peyer的贴剂作为主要的粘膜免疫防御的意义。
    The proinflammatory cytokine tumor necrosis factor (TNF) has been described as one of the main mediators of intestinal inflammatory diseases, affecting the composition of tight junction (TJ) proteins and leading to a disruption of the epithelial barrier. An intact intestinal barrier is mandatory, because the follicle-associated epithelium of Peyer\'s patches represents the first defense line of the intestinal immune system and ensures a controlled uptake of antigens from the gut lumen. In the current study, we have analyzed the detailed effects of TNF on the follicle-associated epithelium of porcine Peyer\'s patches by applying the Ussing chamber technique. Epithelial tissue specimens of Peyer\'s patches and the surrounding villus epithelium were mounted into conventional Ussing chambers and incubated with TNF for 10 h. The transepithelial resistance, representing epithelial barrier function of the tissue, was recorded. A reduction of transepithelial resistance was detected after 8 h in Peyer\'s patch tissue specimens, whereas the villus epithelium was not significantly affected by TNF. Subsequent molecular analysis of TJ protein expression revealed a marked decrease of claudin-1 and -4, and an increase of claudin-2. In neighboring villus epithelium, no significant changes in the expression of TJ proteins could be shown. A strong increase of TNF receptor-2 (TNFR-2) could also be detected in Peyer\'s patches, in agreement with the major role of this receptor in Peyer\'s patches. Our findings were in accordance with changes detected by confocal laser scanning immunofluorescence microscopy. The regulation of TNF effects via myosin light chain kinase (MLCK) was analyzed in blocking experiments. Our detailed analysis is the first to show that TNF affects the barrier function of the follicle-associated epithelium of porcine Peyer\'s patches but has no effects on the villus epithelium. These findings reveal not only the basic differences of epithelial barrier function between the two structures, but also the significance of Peyer\'s patches as a primary mucosal immune defense.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号