Interleukin-15

白细胞介素 - 15
  • 文章类型: Journal Article
    代谢紊乱在现代社会中非常普遍。运动模拟物被定义为可以产生健身有益效果的药理学化合物。最近,人们越来越关注丁香酚和瞬时受体电位香草酸1(TRPV1)在改善代谢健康方面的作用.这项研究的目的是调查丁香酚是否通过激活TRPV1来充当运动模拟物。这里,我们发现丁香酚能提高耐力,导致了快到慢的肌肉纤维的转化,并促进小鼠白色脂肪褐变和脂肪分解。机械上,丁香酚通过激活TRPV1介导的CaN信号通路促进肌纤维型转化。随后,我们将IL-15鉴定为受活化T细胞胞浆1(NFATc1)信号通路的CaN/核因子调控的肌细胞因子。此外,我们发现TRPV1介导的CaN/NFATc1信号,丁香酚激活,C2C12肌管中控制的IL-15水平。我们的结果表明,丁香酚可以作为一种运动模拟物,通过激活TRPV1介导的CaN信号通路来改善代谢健康。
    Metabolic disorders are highly prevalent in modern society. Exercise mimetics are defined as pharmacological compounds that can produce the beneficial effects of fitness. Recently, there has been increased interest in the role of eugenol and transient receptor potential vanilloid 1 (TRPV1) in improving metabolic health. The aim of this study was to investigate whether eugenol acts as an exercise mimetic by activating TRPV1. Here, we showed that eugenol improved endurance capacity, caused the conversion of fast-to-slow muscle fibers, and promoted white fat browning and lipolysis in mice. Mechanistically, eugenol promoted muscle fiber-type transformation by activating TRPV1-mediated CaN signaling pathway. Subsequently, we identified IL-15 as a myokine that is regulated by the CaN/nuclear factor of activated T cells cytoplasmic 1 (NFATc1) signaling pathway. Moreover, we found that TRPV1-mediated CaN/NFATc1 signaling, activated by eugenol, controlled IL-15 levels in C2C12 myotubes. Our results suggest that eugenol may act as an exercise mimetic to improve metabolic health via activating the TRPV1-mediated CaN signaling pathway.
    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
    自然杀伤(NK)细胞在抵抗病毒感染和肿瘤的免疫反应中起着至关重要的作用。然而,需要进一步研究以更好地了解决定NK细胞命运和功能的关键分子.在这项研究中,关于Hippo激酶Mst1和Mst2作为新的调节因子参与维持小鼠NK细胞稳态,我们取得了重要发现.NK细胞中存在高Mst1和Mst2(Mst1/2)活性对其正常发育至关重要,在规范的河马信号独立模式下生存和功能。机械上,Mst1/2通过调节增殖和线粒体代谢过程诱导细胞静止,从而保证NK细胞的发育和存活。此外,Mst1/2有效地感知IL-15信号传导并促进pSTAT3-TCF1的激活,这有助于NK细胞稳态。总的来说,我们的研究强调了Mst1/2作为代谢重编程和转录调节的关键调节因子对小鼠NK细胞存活和功能的关键作用,强调NK细胞发育和功能成熟过程中细胞静止的重要性。
    Natural killer (NK) cells play a crucial role in immune response against viral infections and tumors. However, further investigation is needed to better understand the key molecules responsible for determining the fate and function of NK cells. In this study, we made an important discovery regarding the involvement of the Hippo kinases Mst1 and Mst2 as novel regulators in maintaining mouse NK cell homeostasis. The presence of high Mst1 and Mst2 (Mst1/2) activity in NK cells is essential for their proper development, survival and function in a canonical Hippo signaling independent mode. Mechanistically, Mst1/2 induce cellular quiescence by regulating the processes of proliferation and mitochondrial metabolism, thereby ensuring the development and survival of NK cells. Furthermore, Mst1/2 effectively sense IL-15 signaling and facilitate the activation of pSTAT3-TCF1, which contributes to NK cell homeostasis. Overall, our investigation highlights the crucial role of Mst1/2 as key regulators in metabolic reprogramming and transcriptional regulation for mouse NK cell survival and function, emphasizing the significance of cellular quiescence during NK cell development and functional maturation.
    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
    尽管CART疗法在血液恶性肿瘤中取得了显著成功,其在实体瘤中的疗效仍然有限。细胞因子工程化的CART细胞提供了一个有希望的途径,然而,它们的临床翻译受到与组成型细胞因子表达相关的风险的阻碍。在这个概念验证研究中,我们利用内源性IFN-γ启动子进行转基因IL-15的表达。我们证明IFN-γ表达受到TCR信号的严格调控。通过HDR介导的敲入将IRES-IL15引入IFN-γ基因的3'-UTR,我们证实IL-15的表达可以与IFN-γ以抗原刺激依赖性方式共表达。重要的是,转基因的插入不会损害内源性IFN-γ表达。体外和体内数据表明,由IFN-γ启动子驱动的IL-15显著提高了CAR-T细胞的抗肿瘤活性,提示IL-15表达的有效性。最后,作为我们临床翻译努力的一部分,我们开发了一种创新的双基因敲入方法。该方法使得能够使用单个AAV载体将CAR和IL-15基因同时整合到TRAC和IFN-γ基因座中。使用这种方法工程化以表达IL-15的CART细胞显示出增强的抗肿瘤功效。总的来说,我们的研究强调了利用内源性启动子在CART细胞中表达转基因细胞因子的可行性.
    Despite the remarkable success of chimeric antigen receptor (CAR) T therapy in hematological malignancies, its efficacy in solid tumors remains limited. Cytokine-engineered CAR T cells offer a promising avenue, yet their clinical translation is hindered by the risks associated with constitutive cytokine expression. In this proof-of-concept study, we leverage the endogenous interferon (IFN)-γ promoter for transgenic interleukin (IL)-15 expression. We demonstrate that IFN-γ expression is tightly regulated by T cell receptor signaling. By introducing an internal ribosome entry site IL15 into the 3\' UTR of the IFN-γ gene via homology directed repair-mediated knock-in, we confirm that IL-15 expression can co-express with IFN-γ in an antigen stimulation-dependent manner. Importantly, the insertion of transgenes does not compromise endogenous IFN-γ expression. In vitro and in vivo data demonstrate that IL-15 driven by the IFN-γ promoter dramatically improves CAR T cells\' antitumor activity, suggesting the effectiveness of IL-15 expression. Last, as a part of our efforts toward clinical translation, we have developed an innovative two-gene knock-in approach. This approach enables the simultaneous integration of CAR and IL-15 genes into TRAC and IFN-γ gene loci using a single AAV vector. CAR T cells engineered to express IL-15 using this approach demonstrate enhanced antitumor efficacy. Overall, our study underscores the feasibility of utilizing endogenous promoters for transgenic cytokines expression in CAR T cells.
    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
    嵌合抗原受体(CAR)T细胞已在血液恶性肿瘤中表现出突出的治疗成功。然而,由于肿瘤微环境(TME)中细胞毒性T和CAR-T细胞的浸润不足,它们对实体瘤的疗效仍然受到限制,与实体瘤患者预后不良相关的因素。为了克服这个限制,我们设计了CAR-T细胞分泌CXCL10和IL15(10×15CAR-T),维持T细胞活力并增强它们的募集,从而在体外放大CAR-T细胞的长期细胞毒性能力。在使用NUGC4-T21细胞的异种移植模型中,与使用第二代CAR-T细胞治疗的小鼠相比,接受10×15CAR-T细胞的小鼠表现出优异的肿瘤减少和延长的存活率.组织病理学评估表明,在10X15CAR-T细胞处理后,TME中细胞毒性T细胞积累显著增加。因此,这些CAR-T细胞中CXCL10和IL15的协同分泌增强了肿瘤组织内的T细胞募集和适应性,改善肿瘤控制。这种方法可能为在实体肿瘤治疗中推进CAR-T疗法提供有希望的策略。
    Chimeric antigen receptor (CAR) T cells have demonstrated outstanding therapeutic success in hematological malignancies. Yet, their efficacy against solid tumors remains constrained due to inadequate infiltration of cytotoxic T and CAR-T cells in the tumor microenvironment (TME), a factor correlated with poor prognosis in patients with solid tumors. To overcome this limitation, we engineered CAR-T cells to secrete CXCL10 and IL15 (10 × 15 CAR-T), which sustain T cell viability and enhance their recruitment, thereby amplifying the long-term cytotoxic capacity of CAR-T cells in vitro. In a xenograft model employing NUGC4-T21 cells, mice receiving 10 × 15 CAR-T cells showed superior tumor reduction and extended survival rates compared to those treated with second-generation CAR-T cells. Histopathological evaluations indicated a pronounced increase in cytotoxic T cell accumulation in the TME post 10 × 15 CAR-T cell treatment. Therefore, the synergistic secretion of CXCL10 and IL15 in these CAR-T cells enhances T cell recruitment and adaptability within tumor tissues, improving tumor control. This approach may offer a promising strategy for advancing CAR-T therapies in the treatment of solid tumors.
    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
    免疫疗法广泛用于癌症治疗;然而,只有一部分患者对此反应良好。已经做出了巨大的努力来鉴定将受益于免疫疗法的患者。成功的抗肿瘤免疫取决于完整的癌症免疫循环,尤其是持久的CD8+T细胞反应。干扰素(IFN)-α/β/IFN-γ/白介素(IL)-15途径已被报道参与CD8T细胞的发育。这些途径可以预测对免疫疗法的反应。在这里,我们旨在分析多个公共数据库,以调查IFN-α/β/IFN-γ/IL-15通路是否可用于预测免疫治疗应答.结果表明,IFN-α/β/IFN-γ/IL-15通路能有效预测免疫治疗反应,鸟苷酸结合蛋白1(GBP1)可以代表IFN-α/β/IFN-γ/IL-15途径。在公共和私人群体中,我们进一步证明GBP1可以有效预测免疫治疗的反应。功能上,GBP1主要在巨噬细胞中表达,并与参与T细胞迁移的趋化因子密切相关。因此,我们的研究全面调查了GBP1在免疫治疗中的潜在作用,它可以作为免疫疗法的新型生物标志物和药物开发的靶标。
    Immunotherapy is widely used in cancer treatment; however, only a subset of patients responds well to it. Significant efforts have been made to identify patients who will benefit from immunotherapy. Successful anti-tumor immunity depends on an intact cancer-immunity cycle, especially long-lasting CD8+ T-cell responses. Interferon (IFN)-α/β/IFN-γ/interleukin (IL)-15 pathways have been reported to be involved in the development of CD8+ T cells. And these pathways may predict responses to immunotherapy. Herein, we aimed to analyze multiple public databases to investigate whether IFN-α/β/IFN-γ/IL-15 pathways could be used to predict the response to immunotherapy. Results showed that IFN-α/β/IFN-γ/IL-15 pathways could efficiently predict immunotherapy response, and guanylate-binding protein 1 (GBP1) could represent the IFN-α/β/IFN-γ/IL-15 pathways. In public and private cohorts, we further demonstrated that GBP1 could efficiently predict the response to immunotherapy. Functionally, GBP1 was mainly expressed in macrophages and strongly correlated with chemokines involved in T-cell migration. Therefore, our study comprehensively investigated the potential role of GBP1 in immunotherapy, which could serve as a novel biomarker for immunotherapy and a target for drug development.
    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
    免疫细胞因子的临床应用受到剂量限制性毒性的严格限制。为了应对这一挑战,在此,我们提出了下一代免疫细胞因子的概念,涉及LH05的设计,LH05是一种肿瘤条件性抗PD-L1/白介素-15(IL-15)前药.LH05创新性地掩盖了具有空间位阻的IL-15,减轻IL-15的“细胞因子汇”效应并降低与野生型抗PD-L1/IL-15相关的全身毒性。此外,在肿瘤微环境内的特异性蛋白水解裂解,LH05释放活性IL-15超激动剂,发挥有效的抗肿瘤作用。机械上,LH05的抗肿瘤功效取决于通过刺激趋化因子CXCL9和CXCL10而增加的CD8+T细胞和自然杀伤细胞的浸润,从而将冷肿瘤转化为热肿瘤.此外,在晚期和转移性肿瘤模型中,肿瘤条件性抗PD-L1/IL-15可与溶瘤病毒或检查点阻断协同作用.我们的发现为下一代免疫细胞因子的开发提供了令人信服的概念证明,对当前免疫疗法的知识和策略做出了重大贡献。
    The clinical applications of immunocytokines are severely restricted by dose-limiting toxicities. To address this challenge, here we propose a next-generation immunocytokine concept involving the design of LH05, a tumor-conditional anti-PD-L1/interleukin-15 (IL-15) prodrug. LH05 innovatively masks IL-15 with steric hindrance, mitigating the \"cytokine sink\" effect of IL-15 and reducing systemic toxicities associated with wild-type anti-PD-L1/IL-15. Moreover, upon specific proteolytic cleavage within the tumor microenvironment, LH05 releases an active IL-15 superagonist, exerting potent antitumor effects. Mechanistically, the antitumor efficacy of LH05 depends on the increased infiltration of CD8+ T and natural killer cells by stimulating the chemokines CXCL9 and CXCL10, thereby converting cold tumors into hot tumors. Additionally, the tumor-conditional anti-PD-L1/IL-15 can synergize with an oncolytic virus or checkpoint blockade in advanced and metastatic tumor models. Our findings provide a compelling proof of concept for the development of next-generation immunocytokines, contributing significantly to current knowledge and strategies of immunotherapy.
    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
    真武唐(ZWT),传统的草药混合物,临床上已推荐用于治疗狼疮性肾炎(LN)。然而,其作用机制仍然未知。在这里,我们旨在定义ZWT对LN的影响的免疫学机制,并确定其是否影响肾组织驻留的记忆T(TRM)细胞。鼠LN是通过单次注射普利坦诱导的,而体外TRM细胞分化为IL-15/TGF-β。我们发现ZWT或霉酚酸酯治疗可通过降低24小时尿蛋白来显着改善LN小鼠的肾损伤。Scr和抗dsDNAAb。ZWT还改善了肾脏病理并减少了IgG和C3沉积。此外,ZWT下调肾Desmin表达。此外,它降低了LN小鼠肾脏中CD8TRM细胞的数量,同时降低了TNF-α和IFN-γ的表达。与体内结果一致,含ZWT的血清在体外抑制IL-15/TGF-β诱导的TRM细胞分化。机械上,抑制CD8+TRM细胞中STAT3和CD122(IL2/IL-15Rβ)的磷酸化表达。重要的是,ZWT减少了F4/80+CD11b+和CD86+的总数,但不是CD206+,LN小鼠肾脏中的巨噬细胞。有趣的是,ZWT在体内和体外抑制巨噬细胞中IL-15蛋白的表达。因此,我们提供了第一个证据,证明ZWT汤可通过抑制IL-15/IL-15R/STAT3信号传导而减少CD8+TRM细胞,从而改善LN的预后.
    Zhen-Wu-Tang (ZWT), a conventional herbal mixture, has been recommended for treating lupus nephritis (LN) in clinic. However, its mechanisms of action remain unknown. Here we aimed to define the immunological mechanisms underlying the effects of ZWT on LN and to determine whether it affects renal tissue-resident memory T (TRM) cells. Murine LN was induced by a single injection of pristane, while in vitro TRM cells differentiated with IL-15/TGF-β. We found that ZWT or mycophenolate mofetil treatment significantly ameliorated kidney injury in LN mice by decreasing 24-h urine protein, Scr and anti-dsDNA Ab. ZWT also improved renal pathology and decreased IgG and C3 depositions. In addition, ZWT down-regulated renal Desmin expression. Moreover, it lowered the numbers of CD8+ TRM cells in kidney of mice with LN while decreasing their expression of TNF-α and IFN-γ. Consistent with in vivo results, ZWT-containing serum inhibited TRM cell differentiation induced by IL-15/TGF-β in vitro. Mechanistically, it suppressed phosphorylation of STAT3 and CD122 (IL2/IL-15Rβ)expression in CD8+ TRM cells. Importantly, ZWT reduced the number of total F4/80+CD11b+ and CD86+, but not CD206+, macrophages in the kidney of LN mice. Interestingly, ZWT suppressed IL-15 protein expression in macrophages in vivo and in vitro. Thus, we have provided the first evidence that ZWT decoction can be used to improve the outcome of LN by reducing CD8+ TRM cells via inhibition of IL-15/IL-15R /STAT3 signaling.
    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
    特发性重型再生障碍性贫血(SAA)是由T细胞诱导的造血干细胞和祖细胞(HSPC)破坏引起的骨髓衰竭疾病,然而,机制尚不清楚。我们对来自SAA患者和健康供体的PBMC和BMMCs进行了单细胞RNA测序,并鉴定了在骨髓中具有组织驻留表型(Trm)的CD8T细胞亚群,该细胞亚群表现出高IFN-γ和FasL表达,并且具有更高的能力通过FasL表达体外诱导HSPC的凋亡。IL-15Rα在单核细胞上呈递IL-15诱导CD8+Trm细胞,尤其是CD16+单核细胞,在SAA患者中增加。CD16+单核细胞有助于IL-15诱导的CD38+CXCR6+前Trm分化为CD8+Trm细胞,可以被CD38抑制剂78c抑制。我们的结果表明,IL-15诱导的CD8Trm细胞是介导SAA患者HSPC破坏的致病细胞,并且是未来治疗的治疗靶标。
    Idiopathic severe aplastic anemia (SAA) is a disease of bone marrow failure caused by T-cell-induced destruction of hematopoietic stem and progenitor cells (HSPCs), however the mechanism remains unclear. We performed single-cell RNA sequencing of PBMCs and BMMCs from SAA patients and healthy donors and identified a CD8+ T cell subset with a tissue residency phenotype (Trm) in bone marrow that exhibit high IFN-γ and FasL expression and have a higher ability to induce apoptosis in HSPCs in vitro through FasL expression. CD8+ Trm cells were induced by IL-15 presented by IL-15Rα on monocytes, especially CD16+ monocytes, which were increased in SAA patients. CD16+ monocytes contributed to IL-15-induced CD38+CXCR6+ pre-Trm differentiation into CD8+ Trm cells, which can be inhibited by the CD38 inhibitor 78c. Our results demonstrate that IL-15-induced CD8+ Trm cells are pathogenic cells that mediate HSPC destruction in SAA patients and are therapeutic targets for future treatments.
    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
    嵌合抗原受体(CAR)-NK细胞不仅可以通过CAR分子识别抗原表达的癌细胞的能力,还可以通过NK细胞受体本身来消除肿瘤。这克服了CAR-T细胞的一些局限性,为更安全、更有效的现成细胞治疗铺路CAR-NK细胞。在这项研究中,CD70,淋巴瘤的泛靶点,构建了具有4-1BB共刺激结构域和IL-15的特异性第四代CAR,并通过Baboon包膜假型Lenti载体将其转导至脐带血来源的NK细胞.当与未转导的NK细胞和CD19特异性CAR-NK细胞相比时,CD70-CARNK细胞在体外和体内对CD19阴性B细胞淋巴瘤表现出优异的细胞毒性活性。重要的是,小鼠接受两种剂量的CD70-CARNK细胞显示有效根除肿瘤,伴有血浆IL-15浓度增加和CAR-NK细胞增殖和持久性增强。我们的研究表明,与单剂量CAR-NK细胞治疗B细胞淋巴瘤相比,重复给药的CAR-NK细胞疗法具有临床优势。
    UNASSIGNED: Chimeric antigen receptor (CAR) natural killer (NK) cells can eliminate tumors not only through the ability of the CAR molecule to recognize antigen-expressed cancer cells but also through NK-cell receptors themselves. This overcomes some of the limitations of CAR T cells, paving the way for CAR NK cells for safer and more effective off-the-shelf cellular therapy. In this study, CD70-specific (a pan-target of lymphoma) fourth-generation CAR with 4-1BB costimulatory domain and interleukin-15 (IL-15) was constructed and transduced into cord blood-derived NK cells by Baboon envelope pseudotyped lentiviral vector. CD70-CAR NK cells displayed superior cytotoxic activity in vitro and in vivo against CD19-negative B-cell lymphoma when compared with nontransduced NK cells and CD19-specific CAR NK cells. Importantly, mice that received 2 doses of CD70-CAR NK cells showed effective eradication of tumors, accompanied by increased concentration of plasma IL-15 and enhanced CAR NK cell proliferation and persistence. Our study suggests that repetitive administration-based CAR NK-cell therapy has clinical advantage compared with a single dose of CAR NK cells for the treatment of B-cell lymphoma.
    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
    近年来,癌症免疫疗法已成为一种有前途的临床治疗策略。不幸的是,复杂的免疫抑制肿瘤微环境(ITM)限制了免疫治疗的令人满意的抗肿瘤疗效。为了重塑ITM并减轻免疫逃避,我们构建了FA-PEG修饰的脂质体来递送质粒IL-15(pIL-15)和吉西他滨(GEM)(FPCL@pIL-15+FPGL),分别。取决于叶酸(FA)专门的靶向功能,FPCL@pIL-15(150nm)和FPGL(120nm)表现出对称的球形结构以及在肿瘤组织上的期望的渗透和积累。IL-15的转染表达通过与IL-15R结合有效促进自然杀伤(NK)细胞和CD8+T细胞的增殖和共活化。FPGL上调自然杀伤组2成员D配体(NKG2DLs)的表达,增强NK细胞的识别,以减轻免疫逃避,同时通过免疫原性细胞死亡(ICD)作用促进CD8+T细胞的活化。更重要的是,联合给药在皮下4T1肿瘤模型中实现了预期的抗肿瘤功效。实质上,我们证明了FPCL@pIL-15与FPGL的结合协同刺激和动员免疫系统以逆转ITM并触发抗肿瘤免疫反应,表明在免疫疗法中应用的巨大潜力。
    Cancer immunotherapy has emerged as a promising clinical treatment strategy in recent years. Unfortunately, the satisfactory antitumor therapeutic efficacy of immunotherapy is limited by intricate immunosuppressive tumor microenvironment (ITM). To remodel the ITM and alleviate the immune evasion, we constructed FA-PEG-modified liposomes to deliver plasmid IL-15 (pIL-15) and gemcitabine (GEM) (FPCL@pIL-15 + FPGL), respectively. The FPCL@pIL-15 (150 nm) and FPGL (120 nm) exhibited symmetrically spherical structures as well as desirable penetration and accumulation on tumor tissue depending on folic acid (FA) specialized targeting function. The transfected expression of IL-15 efficiently fosters the proliferation and co-activation of Natural killer (NK) cells and CD8+T cells through binding to IL-15R. FPGL upregulated the expression of Natural killer group 2 member D ligands (NKG2DLs) and reinforced recognition by NK cells to alleviate the immune evasion, and simultaneously promoted activation of CD8+T cells through immunogenic cell death (ICD) effects. More importantly, the combinatorial administration achieved intended anti-tumor efficacy in the subcutaneous 4T1 tumor model. In essence, we demonstrated that combining FPCL@pIL-15 with FPGL synergistically stimulates and mobilizes the immune system to reverse the ITM and trigger an anti-tumor immune response, indicating a tremendous potential for application in immunotherapy.
    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