cGAS‐STING pathway

  • 文章类型: Journal Article
    这项研究通过使用高通量单细胞转录组测序和先进的生物信息学分析研究TRIM28的作用及其复杂的分子机制,深入研究了去势抵抗前列腺癌(CRPC)的未开发领域。我们的全面检查揭示了动态的TRIM28表达变化,特别是在CRPC内的免疫细胞如巨噬细胞和CD8+T细胞中。与TCGA数据的相关性分析强调了TRIM28与免疫检查点表达之间的联系,并强调了其对免疫细胞数量和功能的关键影响。使用TRIM28基因敲除小鼠模型,我们确定了差异表达的基因和富集的途径,揭示TRIM28在cGAS-STING途径中的潜在调控参与。体外,实验进一步说明,前列腺癌细胞中TRIM28基因敲除通过抑制M2巨噬细胞极化和增强CD8+T细胞活性而诱导了显著的抗肿瘤免疫作用。这一有影响力的发现在原位移植肿瘤模型中得到了验证,其中TRIM28基因敲除表现出肿瘤生长的减速,减少M2巨噬细胞的比例,增强CD8+T细胞的浸润。总之,这项研究阐明了迄今为止未知的TRIM28在CRPC中的抗肿瘤免疫作用,并通过cGAS-STING信号通路揭示了其潜在的调节机制。这些发现为CRPC的免疫前景提供了新的见解,为开发创新的治疗策略提供有希望的方向。
    This study delves into the unexplored realm of castration-resistant prostate cancer (CRPC) by investigating the role of TRIM28 and its intricate molecular mechanisms using high-throughput single-cell transcriptome sequencing and advanced bioinformatics analysis. Our comprehensive examination unveiled dynamic TRIM28 expression changes, particularly in immune cells such as macrophages and CD8+ T cells within CRPC. Correlation analyses with TCGA data highlighted the connection between TRIM28 and immune checkpoint expression and emphasized its pivotal influence on the quantity and functionality of immune cells. Using TRIM28 knockout mouse models, we identified differentially expressed genes and enriched pathways, unraveling the potential regulatory involvement of TRIM28 in the cGAS-STING pathway. In vitro, experiments further illuminated that TRIM28 knockout in prostate cancer cells induced a notable anti-tumor immune effect by inhibiting M2 macrophage polarization and enhancing CD8+ T cell activity. This impactful discovery was validated in an in situ transplant tumor model, where TRIM28 knockout exhibited a deceleration in tumor growth, reduced proportions of M2 macrophages, and enhanced infiltration of CD8+ T cells. In summary, this study elucidates the hitherto unknown anti-tumor immune role of TRIM28 in CRPC and unravels its potential regulatory mechanism via the cGAS-STING signaling pathway. These findings provide novel insights into the immune landscape of CRPC, offering promising directions for developing innovative therapeutic strategies.
    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
    原位癌疫苗接种是刺激保护性抗肿瘤免疫的有吸引力的策略。细胞毒性T淋巴细胞(CTL)是适应性免疫防御的主要介质,在抗肿瘤免疫反应和建立免疫记忆中起关键作用,因此对于原位疫苗产生系统性和持久的抗肿瘤功效极为重要。然而,实体瘤中致密的细胞外基质和缺氧严重阻碍了CTL的浸润和功能,最终损害原位癌疫苗的功效。为了解决这个问题,一种强大的原位癌疫苗,Au@MnO2纳米颗粒(AMOPs),开发了基于包覆有二氧化锰壳的金纳米粒子核。AMOP调节不利的肿瘤微环境(TME)以恢复CTL的浸润和功能,并有效诱导免疫原性细胞死亡。干扰素基因途径的Mn2+介导的刺激物可以被激活以进一步增强AMOPs的治疗功效。因此,AMOPs疫苗成功地引发了持久的抗肿瘤免疫,大大抑制了原发,经常性,和转移性肿瘤。这项研究不仅强调了恢复CTL对实体瘤的功效的重要性,而且在克服TME障碍以实现持续的抗肿瘤免疫方面取得了进展。
    In situ cancer vaccination is an attractive strategy that stimulates protective antitumor immunity. Cytotoxic T lymphocytes (CTLs) are major mediators of the adaptive immune defenses, with critical roles in antitumor immune response and establishing immune memory, and are consequently extremely important for in situ vaccines to generate systemic and lasting antitumor efficacy. However, the dense extracellular matrix and hypoxia in solid tumors severely impede the infiltration and function of CTLs, ultimately compromising the efficacy of in situ cancer vaccines. To address this issue, a robust in situ cancer vaccine, Au@MnO2 nanoparticles (AMOPs), based on a gold nanoparticle core coated with a manganese dioxide shell is developed. The AMOPs modulated the unfavorable tumor microenvironment (TME) to restore CTLs infiltration and function and efficiently induced immunogenic cell death. The Mn2+-mediated stimulator of the interferon genes pathway can be activated to further augment the therapeutic efficacy of the AMOPs. Thus, the AMOPs vaccine successfully elicited long-lasting antitumor immunity to considerably inhibit primary, recurrent, and metastatic tumors. This study not only highlights the importance of revitalizing CTLs efficacy against solid tumors but also makes progress toward overcoming TME barriers for sustained antitumor 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
    探索抑制雄激素受体(AR)活性的靶标是抑制去势抵抗性前列腺癌(CRPC)发展的有效策略。组蛋白去甲基酶JMJD2A活性的上调是增加CRPC中AR表达的重要因素。根据我们的研究,我们发现JMJD2A和AR之间的结合亲和力在CRPC中增加,而AR增强子区域的AR组蛋白甲基化水平降低,而H3K27ac水平升高。进一步的研究表明,组蛋白脱甲基酶JMJD2A的过表达增加了JMJD2A和AR之间的结合亲和力,AR组蛋白甲基化水平降低,在AR增强子区域上调H3K27ac,增强AR活性。相反,击倒JMJD2A有效地逆转了这些影响。此外,在CRPC,JMJD2A表达上调,肿瘤固有免疫cGAS-STING信号通路被抑制,肿瘤微环境改变了,AR表达上调。然而,敲除JMJD2A和抑制干扰素基因的环GMP-AMP合酶/刺激因子(cGAS-STING)信号通路均逆转了这些作用。总之,我们的研究表明,在CRPC中,JMJD2A可以直接结合AR并通过其去甲基化活性激活残留的AR增强剂,从而促进AR表达。此外,JMJD2A表达上调抑制肿瘤的先天性免疫cGAS-STING信号通路,导致抗肿瘤免疫功能下降,并进一步促进AR表达。
    Exploring targets for inhibiting androgen receptor (AR) activity is an effective strategy for suppressing the development of castration-resistant prostate cancer (CRPC). Upregulation of histone demethylase JMJD2A activity is an important factor in increasing AR expression in CRPC. Based on our research, we found that the binding affinity between JMJD2A and AR increases in CRPC, while the level of AR histone methylation decreases and the H3K27ac level increases in the AR enhancer region. Further investigations revealed that overexpression of the histone demethylase JMJD2A increased the binding affinity between JMJD2A and AR, decreased AR histone methylation levels, upregulated H3K27ac in the AR enhancer region, and increased AR activity. Conversely, knocking down JMJD2A effectively reversed these effects. Additionally, in CRPC, JMJD2A expression was upregulated, the tumor-intrinsic immune cGAS-STING signaling pathway was suppressed, the tumor microenvironment was altered, and AR expression was upregulated. However, both knocking down JMJD2A and inhibiting the cyclic GMP-AMP synthase/stimulator of interferon genes (cGAS-STING) signaling pathway reversed these effects. In summary, our study indicates that in CRPC, JMJD2A can directly bind to AR and activate residual AR enhancers through its demethylation activity, thereby promoting AR expression. Furthermore, upregulation of JMJD2A expression inhibits the innate immune cGAS-STING signaling pathway of the tumor, leading to a decrease in antitumor immune function, and further promoting AR expression.
    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
    手术切除仍然是恶性黑色素瘤的主流治疗方法。然而,伤口愈合和残留肿瘤转移的挑战构成了重大障碍,导致患者复发率高。在这里,通过海藻酸钠(SA)与锰掺杂的生物活性玻璃(BG-Mn)交联形成的生物活性可注射水凝胶(BG-Mngel)被开发为抗肿瘤免疫疗法和黑色素瘤术后伤口愈合的通用平台。在生物活性玻璃(BG)中掺入Mn2可以激活cGAS-STING免疫途径,以引发癌症免疫疗法的强大免疫反应。此外,在BG中掺杂Mn2使系统具有优异的光热性能,从而促进STING激活和逆转肿瘤免疫抑制微环境。BG表现出良好的血管生成能力和组织再生潜力,Mn2+促进体外细胞迁移。BG-Mngel与抗PD-1抗体(α-PD-1)联合治疗恶性黑色素瘤时,它显示出增强的抗肿瘤免疫应答和长期免疫记忆应答。值得注意的是,BG-Mngel在治疗全层伤口时可以上调与血管形成相关的基因的表达并促进皮肤组织再生。总的来说,BG-MnGel是一种有效的辅助疗法,可调节恶性黑色素瘤的肿瘤转移和伤口愈合。
    Surgical resection remains the mainstream treatment for malignant melanoma. However, challenges in wound healing and residual tumor metastasis pose significant hurdles, resulting in high recurrence rates in patients. Herein, a bioactive injectable hydrogel (BG-Mngel) formed by crosslinking sodium alginate (SA) with manganese-doped bioactive glass (BG-Mn) is developed as a versatile platform for anti-tumor immunotherapy and postoperative wound healing for melanoma. The incorporation of Mn2+ within bioactive glass (BG) can activate the cGAS-STING immune pathway to elicit robust immune response for cancer immunotherapy. Furthermore, doping Mn2+ in BG endows system with excellent photothermal properties, hence facilitating STING activation and reversing the tumor immune-suppressive microenvironment. BG exhibits favorable angiogenic capacity and tissue regenerative potential, and Mn2+ promotes cell migration in vitro. When combining BG-Mngel with anti-PD-1 antibody (α-PD-1) for the treatment of malignant melanoma, it shows enhanced anti-tumor immune response and long-term immune memory response. Remarkably, BG-Mngel can upregulate the expression of genes related to blood vessel formation and promote skin tissue regeneration when treating full-thickness wounds. Overall, BG-MnGel serves as an effective adjuvant therapy to regulate tumor metastasis and wound healing for malignant melanoma.
    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
    刺激环磷酸鸟苷(GMP)-磷酸腺苷(AMP)合酶(cGAS)-干扰素基因刺激物(STING)途径是细菌激活肿瘤免疫系统的关键策略。然而,有限的刺激能力对推进细菌免疫疗法提出了重大挑战.这里,一种腺苷5'-三磷酸(ATP)响应性锰(Mn)基细菌材料(E.大肠杆菌@PDMC-PEG(聚乙二醇)工程成功,表现出协同激活cGAS-STING途径的特殊能力。在肿瘤微环境中,其特征是ATP水平升高,这种生物混合材料降解,导致二价锰离子(Mn2+)的释放和随后的细菌暴露。这种组合协同激活cGAS-STING途径,因为Mn2+增强了cGAS对细菌分泌的细胞外DNA(eDNA)的敏感性。体内实验的结果表明,生物杂合材料大肠杆菌@PDMC-PEG和VNP20009@PDMC-PEG有效抑制小鼠皮下黑素瘤和兔原位肝癌的生长。本文提供了基于细菌的肿瘤免疫治疗的发展的有价值的见解。
    Stimulating the cyclic guanosine monophophate(GMP)-adenosine monophosphate (AMP) synthase (cGAS)-stimulator of interferon genes (STING) pathway is a crucial strategy by which bacteria activate the tumor immune system. However, the limited stimulation capability poses significant challenges in advancing bacterial immunotherapy. Here, an adenosine 5\'-triphosphate (ATP)-responsive manganese (Mn)-based bacterial material (E. coli@PDMC-PEG (polyethylene glycol)) is engineered successfully, which exhibits an exceptional ability to synergistically activate the cGAS-STING pathway. In the tumor microenvironment, which is characterized by elevated ATP levels, this biohybrid material degrades, resulting in the release of divalent manganese ions (Mn2+) and subsequent bacteria exposure. This combination synergistically activates the cGAS-STING pathway, as Mn2+ enhances the sensitivity of cGAS to the extracellular DNA (eDNA) secreted by the bacteria. The results of the in vivo experiments demonstrate that the biohybrid materials E. coli@PDMC-PEG and VNP20009@PDMC-PEG effectively inhibit the growth of subcutaneous melanoma in mice and in situ liver cancer in rabbits. Valuable insights for the development of bacteria-based tumor immunotherapy are provided here.
    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
    尽管治疗取得了显着进展,但许多肝细胞癌(HCC)患者对放疗的反应较差。迫切需要更深入地了解HCC对这种疗法的敏感性机制。已证明RECQL4在HCC患者的恶性细胞中上调。升高的RECQL4水平通过修复辐射诱导的双链DNA(dsDNA)片段降低HCC对放射治疗的敏感性。机械上,RECQL4对放疗的抑制作用是由于肿瘤微环境(TME)中树突状细胞和CD8+T细胞的募集减少。RECQL4通过抑制树突状细胞中的cGAS-STING途径来破坏辐射诱导的TME向杀肿瘤小生境的转化。敲除树突状细胞中的STING可以阻断RECQL4对HCC放射敏感性的影响。值得注意的是,在多个独立队列中,HCC中RECQL4的高表达与不良预后显著相关.总之,这项研究强调了HCC衍生的RECQL4如何通过DNA修复破坏树突状细胞中的cGAS-STING途径激活,从而降低HCC的放射敏感性。这些发现为HCC的临床治疗提供了新的视角。
    Many patients with hepatocellular carcinoma (HCC) respond poorly to radiotherapy despite remarkable advances in treatment. A deeper insight into the mechanism of sensitivity of HCC to this therapy is urgently required. It is demonstrated that RECQL4 is upregulated in the malignant cells of patients with HCC. Elevated RECQL4 levels reduce the sensitivity of HCC to radiotherapy by repairing radiation-induced double-stranded DNA (dsDNA) fragments. Mechanistically, the inhibitory effect of RECQL4 on radiotherapy is due to the reduced recruitment of dendritic cells and CD8+ T cells in the tumor microenvironment (TME). RECQL4 disrupts the radiation-induced transformation of the TME into a tumoricidal niche by inhibiting the cGAS-STING pathway in dendritic cells. Knocking out STING in dendritic cells can block the impact of RECQL4 on HCC radiosensitivity. Notably, high RECQL4 expressions in HCC is significantly associated with poor prognosis in multiple independent cohorts. In conclusion, this study highlights how HCC-derived RECQL4 disrupts cGAS-STING pathway activation in dendritic cells through DNA repair, thus reducing the radiosensitivity of HCC. These findings provide new perspectives on the clinical treatment of HCC.
    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
    佐剂在疫苗开发中具有关键价值,因为它们作用于增强抗原的免疫原性和诱导持久的免疫。然而,只有少数佐剂被批准用于临床,这凸显了探索和开发新的佐剂以满足日益增长的疫苗接种需求的必要性。最近,新出现的证据表明,cGAS-STING途径通过产生I型干扰素反应来协调先天和适应性免疫。已经开发了许多cGAS-STING途径激动剂,并在用于治疗癌症或感染性疾病的临床前研究中进行了测试,获得了有希望的结果。作为佐剂,cGAS-STING激动剂已证明其在各种疾病中激活强大的防御免疫的潜力,包括COVID-19感染。这篇综述总结了cGAS-STING激动剂领域的最新发展,特别关注cGAS-STING激动剂作为疫苗接种佐剂的最新应用。还讨论了潜在的挑战,希望激发未来的研究兴趣,以进一步开发cGAS-STING作为疫苗佐剂。
    Adjuvants are of critical value in vaccine development as they act on enhancing immunogenicity of antigen and inducing long-lasting immunity. However, there are only a few adjuvants that have been approved for clinical use, which highlights the need for exploring and developing new adjuvants to meet the growing demand for vaccination. Recently, emerging evidence demonstrates that the cGAS-STING pathway orchestrates innate and adaptive immunity by generating type I interferon responses. Many cGAS-STING pathway agonists have been developed and tested in preclinical research for the treatment of cancer or infectious diseases with promising results. As adjuvants, cGAS-STING agonists have demonstrated their potential to activate robust defense immunity in various diseases, including COVID-19 infection. This review summarized the current developments in the field of cGAS-STING agonists with a special focus on the latest applications of cGAS-STING agonists as adjuvants in vaccination. Potential challenges were also discussed in the hope of sparking future research interests to further the development of cGAS-STING as vaccine adjuvants.
    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
    环磷酸鸟苷-磷酸腺苷合成酶(cGAS)/干扰素基因刺激因子(STING)信号通路一直是抗癌免疫的一个有希望的目标,但是合理地激活和增强肿瘤细胞中的这一途径至关重要。在这里,开发了谷胱甘肽敏感的ZnFe2O4基纳米系统,以编程方式启动和增强肿瘤细胞中的STING信号通路。将制备的ZnFe2O4纳米粒子包覆癌细胞膜(CCM),这使得纳米系统能够靶向肿瘤细胞。在肿瘤细胞中,ZnFe2O4纳米颗粒可以通过响应高水平的谷胱甘肽而分解,释放的Fe3+产生活性氧,诱导DNA漏入细胞质,刺激cGAS。然后Zn2促进cGAS-DNA相分离以增强cGAS酶活性。此外,作为抗有丝分裂剂的紫杉醇(PTX)的低剂量封装(ZnFe2O4-PTX@CCM)确保了cGAS/STING途径的持续激活。体外和体内结果证实,ZnFe2O4-PTX@CCM提高了cGAS/STING的活性,促进树突状细胞成熟,细胞毒性T淋巴细胞和自然杀伤细胞浸润增加,最终抑制肿瘤进展和术后复发。本研究为构建肿瘤免疫治疗的STING活化纳米系统提供了可行的参考。
    The cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)/stimulator of interferon genes (STING) signalling pathway has been a promising target for anticancer immunity, but rationally activating and enhancing this pathway in tumour cells is critical. Herein, a glutathione sensitive ZnFe2O4-based nanosystem is developed to programmatically initiate and enhance the STING signalling pathway in tumour cells. The prepared ZnFe2O4 nanoparticles were coated with cancer cell membrane (CCM), which enabled the nanosystem target tumour cells. In tumour cells, ZnFe2O4 nanoparticles could be disintegrated by responding to high level glutathione, and the released Fe3+ generated reactive oxygen species to induce the DNA leakage into the cytoplasm to stimulate cGAS. Then Zn2+ promoted cGAS-DNA phase separation to intensify the cGAS enzymatic activity. In addition, the low dose encapsulation of paclitaxel (PTX) acting as an antimitotic agent (ZnFe2O4-PTX@CCM) ensured the sustained activation of cGAS/STING pathway. The in vitro and in vivo results confirmed that ZnFe2O4-PTX@CCM elevated the cGAS/STING activity, promoted dendritic cell maturation, increased cytotoxic T lymphocyte and natural killer cells infiltration, eventually inhibiting the tumour progress and postoperative recurrence. This study provided feasible references on constructing STING activation nanosystem for tumour immunotherapy.
    导出

    更多引用

    收藏

    翻译标题摘要

    我要上传

       PDF(Pubmed)

公众号