首页 > 最新文献

Signal Transduction and Targeted Therapy最新文献

英文 中文
Inhibition of RNA-binding proteins enhances immunotherapy in ovarian cancer. 抑制rna结合蛋白增强卵巢癌的免疫治疗。
IF 39.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-25 DOI: 10.1038/s41392-025-02515-1
Nadine Bley,Alexander Rausch,Simon Müller,Theresa Simon,Markus Glaß,Danny Misiak,Laura Schian,Lara Meret Peters,Mohammad Dipto,Ali Hmedat,Bianca Busch,Annekatrin Schott,Marcell Lederer,Alice Wedler,Robin Benedikt Rolnik,Hend Elrewany,Ehab Ghazy,Wolfgang Sippl,Martina Vetter,Markus Wallwiener,Stefan Hüttelmaier
High-grade serous ovarian cancer (HGSC) accounts for more than 70% of ovarian cancer-related deaths, yet therapeutic progress remains stagnant. Among the four molecular subtypes reported for HGSC, the C5 subtype is distinguished by high proliferation and immune evasion with an unfavorable MHC-I/PD-L1 ratio. However, the molecular drivers of this immune desert state remain largely undefined. Here, we identify RNA-binding proteins (RBPs) as key regulators of immune evasion in C5-HGSC through integrated single-cell and bulk RNA sequencing. We perform a targeted loss-of-function screen in C5-like cell models and find IGF2BP1 as a central mediator of immune evasion in vitro and in vivo. Mechanistically, IGF2BP1 abrogates interferon-gamma signaling by accelerating IRF1 protein degradation, thereby suppressing MHC-I presentation. We also discover that IGF2BP1 decouples PD-L1 expression from IRF1-dependent transcription and reshapes the immune receptor landscape to limit immune cell infiltration and T cell activation. Therapeutically, the small-molecule BTYNB effectively inhibits IGF2BP1 and synergizes with PD-1 blockade to overcome immune evasion in vivo. Multi-spectral imaging confirms these findings in human HGSC tissues and highlights the role of oncofetal RBPs as molecular drivers of the C5-HGSC subtype. This subtype-wide survey uncovers a previously unrecognized RBP-interferon regulatory axis and establishes RBP inhibition as a therapeutic strategy to enhance immune checkpoint therapy in immunologically cold ovarian tumors.
高级别浆液性卵巢癌(HGSC)占卵巢癌相关死亡的70%以上,但治疗进展仍然停滞不前。在报道的四种HGSC分子亚型中,C5亚型的特点是高增殖和免疫逃避,MHC-I/PD-L1比例不利。然而,这种免疫荒漠状态的分子驱动因素在很大程度上仍未确定。在这里,我们通过整合单细胞和大量RNA测序,确定RNA结合蛋白(rbp)是C5-HGSC中免疫逃避的关键调节因子。我们在c5样细胞模型中进行了靶向功能丧失筛选,发现IGF2BP1是体外和体内免疫逃避的中心介质。从机制上讲,IGF2BP1通过加速IRF1蛋白降解来消除干扰素- γ信号,从而抑制MHC-I的呈递。我们还发现IGF2BP1将PD-L1表达与irf1依赖性转录解耦,并重塑免疫受体景观,以限制免疫细胞浸润和T细胞活化。在治疗上,小分子BTYNB有效抑制IGF2BP1并与PD-1阻断协同克服体内免疫逃避。多光谱成像在人类HGSC组织中证实了这些发现,并强调了癌胎rbp作为C5-HGSC亚型的分子驱动因素的作用。这项亚型范围的调查揭示了一个以前未被认识到的RBP-干扰素调节轴,并建立了RBP抑制作为一种治疗策略来增强免疫冷卵巢肿瘤的免疫检查点治疗。
{"title":"Inhibition of RNA-binding proteins enhances immunotherapy in ovarian cancer.","authors":"Nadine Bley,Alexander Rausch,Simon Müller,Theresa Simon,Markus Glaß,Danny Misiak,Laura Schian,Lara Meret Peters,Mohammad Dipto,Ali Hmedat,Bianca Busch,Annekatrin Schott,Marcell Lederer,Alice Wedler,Robin Benedikt Rolnik,Hend Elrewany,Ehab Ghazy,Wolfgang Sippl,Martina Vetter,Markus Wallwiener,Stefan Hüttelmaier","doi":"10.1038/s41392-025-02515-1","DOIUrl":"https://doi.org/10.1038/s41392-025-02515-1","url":null,"abstract":"High-grade serous ovarian cancer (HGSC) accounts for more than 70% of ovarian cancer-related deaths, yet therapeutic progress remains stagnant. Among the four molecular subtypes reported for HGSC, the C5 subtype is distinguished by high proliferation and immune evasion with an unfavorable MHC-I/PD-L1 ratio. However, the molecular drivers of this immune desert state remain largely undefined. Here, we identify RNA-binding proteins (RBPs) as key regulators of immune evasion in C5-HGSC through integrated single-cell and bulk RNA sequencing. We perform a targeted loss-of-function screen in C5-like cell models and find IGF2BP1 as a central mediator of immune evasion in vitro and in vivo. Mechanistically, IGF2BP1 abrogates interferon-gamma signaling by accelerating IRF1 protein degradation, thereby suppressing MHC-I presentation. We also discover that IGF2BP1 decouples PD-L1 expression from IRF1-dependent transcription and reshapes the immune receptor landscape to limit immune cell infiltration and T cell activation. Therapeutically, the small-molecule BTYNB effectively inhibits IGF2BP1 and synergizes with PD-1 blockade to overcome immune evasion in vivo. Multi-spectral imaging confirms these findings in human HGSC tissues and highlights the role of oncofetal RBPs as molecular drivers of the C5-HGSC subtype. This subtype-wide survey uncovers a previously unrecognized RBP-interferon regulatory axis and establishes RBP inhibition as a therapeutic strategy to enhance immune checkpoint therapy in immunologically cold ovarian tumors.","PeriodicalId":21766,"journal":{"name":"Signal Transduction and Targeted Therapy","volume":"16 1","pages":"419"},"PeriodicalIF":39.3,"publicationDate":"2025-12-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145823852","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Engineered extracellular vesicles reprogram T cells by targeting PD-1 and PHB1 signaling in inflammatory bowel disease. 在炎症性肠病中,工程细胞外囊泡通过靶向PD-1和PHB1信号重编程T细胞。
IF 39.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-25 DOI: 10.1038/s41392-025-02516-0
Mi-Kyung Oh,Hyun Sung Park,Dong-Hoon Chae,Aaron Yu,Jae Han Park,Jiyoung Heo,Keonwoo Cho,Jiho Kim,Byeonghwi Lim,Jun-Mo Kim,Jordan E Axelrad,Kyung Ku Jang,Jong Pil Im,Seong-Joon Koh,Byung-Soo Kim,Kyung-Rok Yu
Current therapies for inflammatory bowel disease (IBD) often fail to achieve complete remission and are associated with systemic toxicity owing to their broad immunosuppressive effects. To overcome these limitations, we developed a bioengineered extracellular vesicle (EV) platform that modulates key immune signaling pathways to efficiently restore the T-cell balance in inflamed intestinal tissues. EVs derived from Wharton's jelly mesenchymal stem cells were engineered to display PD-L1 on their surface and encapsulate miR-27a-3p. Surface PD-L1 engages the PD-1 checkpoint in activated T cells, attenuating T-cell receptor signaling via SHP2-mediated dephosphorylation of ZAP70 and AKT. In parallel, miR-27a-3p suppresses prohibitin 1 (PHB1), a mitochondrial regulator of Th17 cell bioenergetics and inflammatory function, thereby reducing Th17 polarization and increasing the number of FOXP3⁺ regulatory T cells. These dual-targeting EVs preferentially localized to inflamed intestinal tissues via chemokine (CCR2/CXCR4) and PD-1-dependent mechanisms. In humanized mouse models of colitis, these EVs attenuated mucosal inflammation, suppressed effector T-cell responses, and preserved epithelial integrity. In IBD patient-derived colonoid cultures, PD-L1/miR-27a-3p EVs maintained epithelial viability and barrier integrity without inducing cytotoxicity or structural disruption. Transcriptomic and single-cell analyses revealed the downregulation of inflammatory and exhaustion signatures, along with the enrichment of regulatory subsets. Collectively, this study presents a cell-free immunotherapeutic approach that reprograms T cells in inflamed tissues through the PD-1 and mitochondrial signaling pathways while maintaining intestinal epithelial integrity, offering a promising therapeutic strategy for IBD and other T cell-driven inflammatory disorders.
目前治疗炎症性肠病(IBD)的方法往往不能实现完全缓解,并且由于其广泛的免疫抑制作用而与全身毒性相关。为了克服这些限制,我们开发了一种生物工程细胞外囊泡(EV)平台,可以调节关键的免疫信号通路,有效地恢复炎症肠组织中的t细胞平衡。从Wharton’s jelly间充质干细胞中提取的ev被设计成在其表面显示PD-L1并包封miR-27a-3p。表面PD-L1参与活化T细胞中的PD-1检查点,通过shp2介导的ZAP70和AKT的去磷酸化减弱T细胞受体信号。与此同时,miR-27a-3p抑制抑制素1 (PHB1), PHB1是Th17细胞生物能量学和炎症功能的线粒体调节剂,从而减少Th17极化,增加FOXP3 +调节性T细胞的数量。这些双靶向ev通过趋化因子(CCR2/CXCR4)和pd -1依赖机制优先定位于炎症肠组织。在人源化结肠炎小鼠模型中,这些ev减轻了粘膜炎症,抑制了效应t细胞反应,并保持了上皮的完整性。在IBD患者衍生的结肠体培养中,PD-L1/miR-27a-3p ev维持上皮活力和屏障完整性,而不会诱导细胞毒性或结构破坏。转录组学和单细胞分析揭示了炎症和衰竭特征的下调,以及调节亚群的富集。总的来说,本研究提出了一种无细胞免疫治疗方法,通过PD-1和线粒体信号通路对炎症组织中的T细胞进行重编程,同时保持肠上皮的完整性,为IBD和其他T细胞驱动的炎症性疾病提供了一种有希望的治疗策略。
{"title":"Engineered extracellular vesicles reprogram T cells by targeting PD-1 and PHB1 signaling in inflammatory bowel disease.","authors":"Mi-Kyung Oh,Hyun Sung Park,Dong-Hoon Chae,Aaron Yu,Jae Han Park,Jiyoung Heo,Keonwoo Cho,Jiho Kim,Byeonghwi Lim,Jun-Mo Kim,Jordan E Axelrad,Kyung Ku Jang,Jong Pil Im,Seong-Joon Koh,Byung-Soo Kim,Kyung-Rok Yu","doi":"10.1038/s41392-025-02516-0","DOIUrl":"https://doi.org/10.1038/s41392-025-02516-0","url":null,"abstract":"Current therapies for inflammatory bowel disease (IBD) often fail to achieve complete remission and are associated with systemic toxicity owing to their broad immunosuppressive effects. To overcome these limitations, we developed a bioengineered extracellular vesicle (EV) platform that modulates key immune signaling pathways to efficiently restore the T-cell balance in inflamed intestinal tissues. EVs derived from Wharton's jelly mesenchymal stem cells were engineered to display PD-L1 on their surface and encapsulate miR-27a-3p. Surface PD-L1 engages the PD-1 checkpoint in activated T cells, attenuating T-cell receptor signaling via SHP2-mediated dephosphorylation of ZAP70 and AKT. In parallel, miR-27a-3p suppresses prohibitin 1 (PHB1), a mitochondrial regulator of Th17 cell bioenergetics and inflammatory function, thereby reducing Th17 polarization and increasing the number of FOXP3⁺ regulatory T cells. These dual-targeting EVs preferentially localized to inflamed intestinal tissues via chemokine (CCR2/CXCR4) and PD-1-dependent mechanisms. In humanized mouse models of colitis, these EVs attenuated mucosal inflammation, suppressed effector T-cell responses, and preserved epithelial integrity. In IBD patient-derived colonoid cultures, PD-L1/miR-27a-3p EVs maintained epithelial viability and barrier integrity without inducing cytotoxicity or structural disruption. Transcriptomic and single-cell analyses revealed the downregulation of inflammatory and exhaustion signatures, along with the enrichment of regulatory subsets. Collectively, this study presents a cell-free immunotherapeutic approach that reprograms T cells in inflamed tissues through the PD-1 and mitochondrial signaling pathways while maintaining intestinal epithelial integrity, offering a promising therapeutic strategy for IBD and other T cell-driven inflammatory disorders.","PeriodicalId":21766,"journal":{"name":"Signal Transduction and Targeted Therapy","volume":"1 1","pages":"418"},"PeriodicalIF":39.3,"publicationDate":"2025-12-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145823838","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting SKAP2 restores sperm motility and morphology through modulating mitochondrial organization and cytoskeletal remodeling. 以SKAP2为靶点,通过调节线粒体组织和细胞骨架重塑,恢复精子活力和形态。
IF 39.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-24 DOI: 10.1038/s41392-025-02513-3
Shiming Gan,Lin Yin,Jiaming Zhou,Sisi Li,Shumin Zhou,Xiaotong Yang,Rui Liu,Xu Fan,Yangyang Li,Zhendong Yao,Jingshou Chen,Peiran Hu,Wenjing Xiong,Yuan Yuan,Yujiao Wen,Youjiang Li,Ge Jin,Jianzhong Sheng,Yuzhen Gao,Hefeng Huang,Chen Zhang
Sperm motility and morphology are indispensable for sperm-egg interaction and successful fertilization. However, the RNA splicing mechanisms in an m6A-dependent manner regulating spermiogenesis-related genes remain poorly defined, and targeted therapy strategies to restore impaired sperm motility and morphology are lacking. In this study, we identify heterogeneous nuclear ribonucleoprotein R (hnRNPR) as a critical m6A-dependent splicing mediator. Pathogenic mutations in HNRNPR cause sperm motility decline, morphological abnormality, and male infertility in both humans and mice. Mechanistically, Hnrnpr mutation disrupts m6A-dependent splicing of Skap2 pre-mRNA, thus impairing cytoskeletal structure and mitochondrial organization in sperm. Consistently, specific knockout of Skap2 in male germ cells displays sperm abnormalities, which phenocopy those observed in humans and mice with Hnrnpr mutants, unveiling a functional hnRNPR-SKAP2 axis. Leveraging these insights, we developed a therapeutic strategy to restore sperm motility and morphology, relying on extracellular vesicle-mediated SKAP2 delivery to enter the efferent ductules of the testicles, which could promote sperm cytoskeletal remodeling and mitochondrial organization. Notably, the co-culture of extracellular vesicle SKAP2 with human and mouse sperms also significantly enhanced the sperm motility. Altogether, these findings identify hnRNPR as a pivotal regulator of m6A-mediated Skap2 splicing during spermiogenesis and highlight extracellular vesicle SKAP2 as a promising therapeutic target for poor sperm quality and male infertility.
精子的运动和形态是精子与卵子相互作用和成功受精的必要条件。然而,以m6a依赖的方式调节精子发生相关基因的RNA剪接机制仍然不明确,并且缺乏靶向治疗策略来恢复受损精子的运动和形态。在这项研究中,我们发现异质核核糖核蛋白R (hnRNPR)是一个关键的m6a依赖性剪接介质。HNRNPR致病性突变导致人类和小鼠精子活力下降、形态异常和男性不育。在机制上,Hnrnpr突变破坏了Skap2前mrna的m6a依赖性剪接,从而损害了精子的细胞骨架结构和线粒体组织。在男性生殖细胞中,Skap2的特异性敲除会显示精子异常,这与在Hnrnpr突变的人类和小鼠中观察到的现象一致,揭示了一个功能性的Hnrnpr - Skap2轴。利用这些见解,我们开发了一种恢复精子活力和形态的治疗策略,依靠细胞外囊泡介导的SKAP2递送进入睾丸的输出小管,这可以促进精子细胞骨架重塑和线粒体组织。值得注意的是,细胞外囊泡SKAP2与人和小鼠精子共培养也显著增强了精子活力。总之,这些发现确定了hnRNPR是精子发生过程中m6a介导的Skap2剪接的关键调节因子,并强调了细胞外囊泡Skap2是治疗精子质量差和男性不育的有希望的治疗靶点。
{"title":"Targeting SKAP2 restores sperm motility and morphology through modulating mitochondrial organization and cytoskeletal remodeling.","authors":"Shiming Gan,Lin Yin,Jiaming Zhou,Sisi Li,Shumin Zhou,Xiaotong Yang,Rui Liu,Xu Fan,Yangyang Li,Zhendong Yao,Jingshou Chen,Peiran Hu,Wenjing Xiong,Yuan Yuan,Yujiao Wen,Youjiang Li,Ge Jin,Jianzhong Sheng,Yuzhen Gao,Hefeng Huang,Chen Zhang","doi":"10.1038/s41392-025-02513-3","DOIUrl":"https://doi.org/10.1038/s41392-025-02513-3","url":null,"abstract":"Sperm motility and morphology are indispensable for sperm-egg interaction and successful fertilization. However, the RNA splicing mechanisms in an m6A-dependent manner regulating spermiogenesis-related genes remain poorly defined, and targeted therapy strategies to restore impaired sperm motility and morphology are lacking. In this study, we identify heterogeneous nuclear ribonucleoprotein R (hnRNPR) as a critical m6A-dependent splicing mediator. Pathogenic mutations in HNRNPR cause sperm motility decline, morphological abnormality, and male infertility in both humans and mice. Mechanistically, Hnrnpr mutation disrupts m6A-dependent splicing of Skap2 pre-mRNA, thus impairing cytoskeletal structure and mitochondrial organization in sperm. Consistently, specific knockout of Skap2 in male germ cells displays sperm abnormalities, which phenocopy those observed in humans and mice with Hnrnpr mutants, unveiling a functional hnRNPR-SKAP2 axis. Leveraging these insights, we developed a therapeutic strategy to restore sperm motility and morphology, relying on extracellular vesicle-mediated SKAP2 delivery to enter the efferent ductules of the testicles, which could promote sperm cytoskeletal remodeling and mitochondrial organization. Notably, the co-culture of extracellular vesicle SKAP2 with human and mouse sperms also significantly enhanced the sperm motility. Altogether, these findings identify hnRNPR as a pivotal regulator of m6A-mediated Skap2 splicing during spermiogenesis and highlight extracellular vesicle SKAP2 as a promising therapeutic target for poor sperm quality and male infertility.","PeriodicalId":21766,"journal":{"name":"Signal Transduction and Targeted Therapy","volume":"56 1","pages":"416"},"PeriodicalIF":39.3,"publicationDate":"2025-12-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145813412","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Correction: Evolving understanding of autoimmune mechanisms and new therapeutic strategies of autoimmune disorders. 纠正:对自身免疫机制的不断发展的理解和自身免疫疾病的新治疗策略。
IF 52.7 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-24 DOI: 10.1038/s41392-025-02525-z
Yi Song, Jian Li, Yuzhang Wu
{"title":"Correction: Evolving understanding of autoimmune mechanisms and new therapeutic strategies of autoimmune disorders.","authors":"Yi Song, Jian Li, Yuzhang Wu","doi":"10.1038/s41392-025-02525-z","DOIUrl":"10.1038/s41392-025-02525-z","url":null,"abstract":"","PeriodicalId":21766,"journal":{"name":"Signal Transduction and Targeted Therapy","volume":"10 1","pages":"426"},"PeriodicalIF":52.7,"publicationDate":"2025-12-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12738843/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145828142","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Lipocalin-2 drives brain metastatic progression through reciprocal tumor-microenvironment interactions in lung cancer. Lipocalin-2在肺癌中通过肿瘤-微环境相互作用驱动脑转移进展。
IF 39.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-24 DOI: 10.1038/s41392-025-02514-2
Yixiang Zhu,Jian Zhang,Danming He,Hongqing Cai,Yan He,Li Yuan,Sini Li,Yucheng Dong,Wei Zhuang,Zhijie Wang,Jianchun Duan,Xue Zhang,Zixiao Ma,Hua Bai,Jie Wang
Brain metastasis is a major contributor to mortality in patients with lung cancer. The unique microenvironment of the brain plays a critical role in the initiation and progression of brain metastases (BM), yet the molecular mechanisms underlying tumor-microenvironment interactions remain poorly understood. Here, we demonstrate that upregulation of lipocalin-2 (LCN2) in tumor cells promotes brain metastatic progression by orchestrating crosstalk among metastatic tumor cells, astrocytes, and macrophages. Brain metastatic tumor cells secrete LCN2, which binds to SLC22A17 on astrocytes, activating JAK2/STAT3 signaling and inducing astrocyte activation and chemokine secretion, thereby facilitating macrophage recruitment. In turn, macrophages secrete IL-1β, which further upregulates LCN2 expression in tumor cells. Prophylactic administration of the IL-1 receptor antagonist anakinra inhibits BM formation, whereas therapeutic administration alone is ineffective. However, treatment with the STAT3 inhibitor SH4-54, either alone or in combination with anakinra, significantly suppressed tumor growth in the BM. Furthermore, tumor-secreted LCN2 can bind to SLC22A17 on tumor cells, activating JAK2/STAT3 signaling and promoting VEGF-A expression and release, which enhances tumor neovascularization. Inhibition of this axis with SH4-54, bevacizumab, or their combination effectively reduces the tumor burden in BM-bearing mice. These findings underscore the central role of LCN2 in driving brain metastasis and highlight a potential therapeutic strategy for targeting brain metastatic lung cancer.
脑转移是肺癌患者死亡的主要原因。大脑独特的微环境在脑转移(BM)的发生和发展中起着至关重要的作用,然而肿瘤-微环境相互作用的分子机制仍然知之甚少。在这里,我们证明了肿瘤细胞中脂钙素-2 (LCN2)的上调通过在转移性肿瘤细胞、星形胶质细胞和巨噬细胞之间协调串扰来促进脑转移进展。脑转移肿瘤细胞分泌LCN2, LCN2与星形胶质细胞上的SLC22A17结合,激活JAK2/STAT3信号,诱导星形胶质细胞活化和趋化因子分泌,从而促进巨噬细胞募集。反过来,巨噬细胞分泌IL-1β,进一步上调肿瘤细胞中LCN2的表达。预防性给予IL-1受体拮抗剂anakinra可抑制BM的形成,而单独治疗性给予是无效的。然而,使用STAT3抑制剂SH4-54治疗,无论是单独治疗还是与anakinra联合治疗,都能显著抑制BM中的肿瘤生长。此外,肿瘤分泌的LCN2可以结合肿瘤细胞上的SLC22A17,激活JAK2/STAT3信号通路,促进VEGF-A的表达和释放,从而促进肿瘤新生血管的形成。用SH4-54、贝伐单抗或它们的联合抑制该轴有效地减轻了荷瘤小鼠的肿瘤负荷。这些发现强调了LCN2在驱动脑转移中的核心作用,并强调了针对脑转移性肺癌的潜在治疗策略。
{"title":"Lipocalin-2 drives brain metastatic progression through reciprocal tumor-microenvironment interactions in lung cancer.","authors":"Yixiang Zhu,Jian Zhang,Danming He,Hongqing Cai,Yan He,Li Yuan,Sini Li,Yucheng Dong,Wei Zhuang,Zhijie Wang,Jianchun Duan,Xue Zhang,Zixiao Ma,Hua Bai,Jie Wang","doi":"10.1038/s41392-025-02514-2","DOIUrl":"https://doi.org/10.1038/s41392-025-02514-2","url":null,"abstract":"Brain metastasis is a major contributor to mortality in patients with lung cancer. The unique microenvironment of the brain plays a critical role in the initiation and progression of brain metastases (BM), yet the molecular mechanisms underlying tumor-microenvironment interactions remain poorly understood. Here, we demonstrate that upregulation of lipocalin-2 (LCN2) in tumor cells promotes brain metastatic progression by orchestrating crosstalk among metastatic tumor cells, astrocytes, and macrophages. Brain metastatic tumor cells secrete LCN2, which binds to SLC22A17 on astrocytes, activating JAK2/STAT3 signaling and inducing astrocyte activation and chemokine secretion, thereby facilitating macrophage recruitment. In turn, macrophages secrete IL-1β, which further upregulates LCN2 expression in tumor cells. Prophylactic administration of the IL-1 receptor antagonist anakinra inhibits BM formation, whereas therapeutic administration alone is ineffective. However, treatment with the STAT3 inhibitor SH4-54, either alone or in combination with anakinra, significantly suppressed tumor growth in the BM. Furthermore, tumor-secreted LCN2 can bind to SLC22A17 on tumor cells, activating JAK2/STAT3 signaling and promoting VEGF-A expression and release, which enhances tumor neovascularization. Inhibition of this axis with SH4-54, bevacizumab, or their combination effectively reduces the tumor burden in BM-bearing mice. These findings underscore the central role of LCN2 in driving brain metastasis and highlight a potential therapeutic strategy for targeting brain metastatic lung cancer.","PeriodicalId":21766,"journal":{"name":"Signal Transduction and Targeted Therapy","volume":"23 1","pages":"417"},"PeriodicalIF":39.3,"publicationDate":"2025-12-24","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145813411","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Scarless circular mRNA-based CAR-T cell therapy elicits superior antitumor efficacy. 基于无疤痕环状mrna的CAR-T细胞疗法可获得卓越的抗肿瘤疗效。
IF 52.7 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-23 DOI: 10.1038/s41392-025-02512-4
Qinchao Hu, Hui Zhao, Kaicheng Zhou, Xuefei Tian, Qian Wang, Xianxin Hua, Xuyao Zhang

Messenger RNA (mRNA)-based transient expression of chimeric antigen receptors (CARs) results in optimal safety profiles and provides promising opportunities to address existing challenges associated with viral vector-based CAR-T-cell therapies and to meet emerging medical needs for noncancerous indications. Conventional linear mRNAs, however, are intrinsically unstable and typically support short-lived protein expression, which can constrain therapeutic activity. Here, we engineered a high-efficiency permuted intron exon (PIE) platform to synthesize scarless circular mRNAs (cmRNAs) that drive robust CAR expression with extended durability. The scarless design avoids extraneous junction sequences, streamlining manufacturability and potentially reducing innate immune sensing. Compared with linear mRNAs, cmRNAs significantly increased both the magnitude and duration of anti-CD19 CAR and anti-GPRC5D CAR expression in primary human T cells. Functionally, cmRNA-based CAR-T cells elicited superior antitumor efficacy over their linear mRNA counterparts, as demonstrated by parallel lines of evidence, including in vitro antigen-specific cytotoxicity, cytokine release, and transcriptomics patterns consistent with sustained activation and absence of exhaustion signatures, as well as in vivo models demonstrating tumor elimination and prolonged survival benefits. Collectively, these findings position cmRNA as a next-generation mRNA modality for potent and controllable CAR expression, thereby providing a robust platform to unleash the full potential of mRNA technologies in cellular immunotherapy and precision medicine.

基于信使RNA (mRNA)的嵌合抗原受体(CARs)的瞬时表达可获得最佳的安全性,并为解决基于病毒载体的car - t细胞疗法相关的现有挑战以及满足非癌性适应症的新兴医疗需求提供了有希望的机会。然而,传统的线性mrna本质上是不稳定的,通常支持短暂的蛋白质表达,这可能会限制治疗活性。在这里,我们设计了一个高效的排列内含子外显子(PIE)平台来合成无疤痕的环状mrna (cmrna),这种环状mrna可以驱动具有较长持久性的强健CAR表达。无疤痕的设计避免了外来的连接序列,简化了可制造性,并潜在地减少了先天免疫感知。与线性mrna相比,cmrna显著增加了人原代T细胞中抗cd19 CAR和抗gprc5d CAR的表达量和持续时间。在功能上,基于cmrna的CAR-T细胞比线性mRNA的细胞具有更强的抗肿瘤功效,这一点得到了平行证据的证明,包括体外抗原特异性细胞毒性、细胞因子释放、与持续激活和缺乏衰竭特征一致的转录组学模式,以及体内模型显示的肿瘤消除和延长生存期的益处。总的来说,这些发现将cmRNA定位为有效和可控的CAR表达的下一代mRNA模式,从而为释放mRNA技术在细胞免疫治疗和精准医学中的全部潜力提供了一个强大的平台。
{"title":"Scarless circular mRNA-based CAR-T cell therapy elicits superior antitumor efficacy.","authors":"Qinchao Hu, Hui Zhao, Kaicheng Zhou, Xuefei Tian, Qian Wang, Xianxin Hua, Xuyao Zhang","doi":"10.1038/s41392-025-02512-4","DOIUrl":"10.1038/s41392-025-02512-4","url":null,"abstract":"<p><p>Messenger RNA (mRNA)-based transient expression of chimeric antigen receptors (CARs) results in optimal safety profiles and provides promising opportunities to address existing challenges associated with viral vector-based CAR-T-cell therapies and to meet emerging medical needs for noncancerous indications. Conventional linear mRNAs, however, are intrinsically unstable and typically support short-lived protein expression, which can constrain therapeutic activity. Here, we engineered a high-efficiency permuted intron exon (PIE) platform to synthesize scarless circular mRNAs (cmRNAs) that drive robust CAR expression with extended durability. The scarless design avoids extraneous junction sequences, streamlining manufacturability and potentially reducing innate immune sensing. Compared with linear mRNAs, cmRNAs significantly increased both the magnitude and duration of anti-CD19 CAR and anti-GPRC5D CAR expression in primary human T cells. Functionally, cmRNA-based CAR-T cells elicited superior antitumor efficacy over their linear mRNA counterparts, as demonstrated by parallel lines of evidence, including in vitro antigen-specific cytotoxicity, cytokine release, and transcriptomics patterns consistent with sustained activation and absence of exhaustion signatures, as well as in vivo models demonstrating tumor elimination and prolonged survival benefits. Collectively, these findings position cmRNA as a next-generation mRNA modality for potent and controllable CAR expression, thereby providing a robust platform to unleash the full potential of mRNA technologies in cellular immunotherapy and precision medicine.</p>","PeriodicalId":21766,"journal":{"name":"Signal Transduction and Targeted Therapy","volume":"10 1","pages":"411"},"PeriodicalIF":52.7,"publicationDate":"2025-12-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12722250/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145811395","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeting ubiquitination in disease and therapy. 靶向泛素化在疾病和治疗中的应用。
IF 39.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-23 DOI: 10.1038/s41392-025-02392-8
Xiaojuan Yang,Tian Lan,Buzhe Zhang,Xue Tao,Weili Qi,Kunlin Xie,Yunshi Cai,Chang Liu,Junhong Han,Hong Wu
Ubiquitination, a critical posttranslational modification (PTM), involves the enzymatic covalent attachment of ubiquitin to target proteins. This process is fundamental for maintaining cellular homeostasis and regulating key biological functions. The ubiquitination pathway, orchestrated by ubiquitin and its associated enzymes, offers remarkable versatility, acting as a cellular sentinel to ensure precise spatiotemporal control of essential molecular processes. Importantly, the components and mechanisms of ubiquitination can be finely tuned in various ways. Dysregulation of this system can disrupt normal biological processes and contribute to the development of various serious human diseases. These findings underscore the importance of investigating ubiquitination to understand disease mechanisms and develop effective treatment strategies. In this review, we summarize the historical developments and key milestones in ubiquitination research, with a focus on its roles in both health and disease. We explore the components and mechanisms involved, the relevant signaling pathways and their crosstalk, and the multilayered regulatory functions of ubiquitination under physiological and pathological conditions. The pathological contexts discussed include cancer, neurodegenerative disorders, cardiovascular diseases, inflammatory conditions, autoinflammatory disorders and developmental disorders. Enhancing our understanding of ubiquitination could provide novel insights into disease pathogenesis and identify new therapeutic targets. We also highlight emerging strategies for cancer treatment, such as proteolysis-targeting chimeras (PROTACs) and molecular glues. Furthermore, we review therapeutic targets and recent progress in clinical research, including ongoing clinical trials and FDA-approved drugs, aimed at leveraging the ubiquitination pathway for disease treatment.
泛素化是一种关键的翻译后修饰(PTM),涉及泛素与靶蛋白的酶促共价附着。这个过程是维持细胞稳态和调节关键生物功能的基础。泛素化途径由泛素及其相关酶协调,具有显著的多功能性,作为细胞哨兵,确保对基本分子过程的精确时空控制。重要的是,泛素化的成分和机制可以通过各种方式进行微调。该系统的失调可以破坏正常的生物过程,并导致各种严重的人类疾病的发展。这些发现强调了研究泛素化对于了解疾病机制和制定有效治疗策略的重要性。在这篇综述中,我们总结了泛素化研究的历史发展和关键里程碑,重点介绍了泛素化在健康和疾病中的作用。我们探讨了泛素化的组成和机制,相关信号通路及其串扰,以及生理和病理条件下泛素化的多层调控功能。讨论的病理背景包括癌症、神经退行性疾病、心血管疾病、炎症、自身炎症性疾病和发育障碍。增强我们对泛素化的理解可以为疾病的发病机制和寻找新的治疗靶点提供新的见解。我们还强调了癌症治疗的新兴策略,如靶向蛋白水解嵌合体(PROTACs)和分子胶。此外,我们回顾了治疗靶点和临床研究的最新进展,包括正在进行的临床试验和fda批准的药物,旨在利用泛素化途径治疗疾病。
{"title":"Targeting ubiquitination in disease and therapy.","authors":"Xiaojuan Yang,Tian Lan,Buzhe Zhang,Xue Tao,Weili Qi,Kunlin Xie,Yunshi Cai,Chang Liu,Junhong Han,Hong Wu","doi":"10.1038/s41392-025-02392-8","DOIUrl":"https://doi.org/10.1038/s41392-025-02392-8","url":null,"abstract":"Ubiquitination, a critical posttranslational modification (PTM), involves the enzymatic covalent attachment of ubiquitin to target proteins. This process is fundamental for maintaining cellular homeostasis and regulating key biological functions. The ubiquitination pathway, orchestrated by ubiquitin and its associated enzymes, offers remarkable versatility, acting as a cellular sentinel to ensure precise spatiotemporal control of essential molecular processes. Importantly, the components and mechanisms of ubiquitination can be finely tuned in various ways. Dysregulation of this system can disrupt normal biological processes and contribute to the development of various serious human diseases. These findings underscore the importance of investigating ubiquitination to understand disease mechanisms and develop effective treatment strategies. In this review, we summarize the historical developments and key milestones in ubiquitination research, with a focus on its roles in both health and disease. We explore the components and mechanisms involved, the relevant signaling pathways and their crosstalk, and the multilayered regulatory functions of ubiquitination under physiological and pathological conditions. The pathological contexts discussed include cancer, neurodegenerative disorders, cardiovascular diseases, inflammatory conditions, autoinflammatory disorders and developmental disorders. Enhancing our understanding of ubiquitination could provide novel insights into disease pathogenesis and identify new therapeutic targets. We also highlight emerging strategies for cancer treatment, such as proteolysis-targeting chimeras (PROTACs) and molecular glues. Furthermore, we review therapeutic targets and recent progress in clinical research, including ongoing clinical trials and FDA-approved drugs, aimed at leveraging the ubiquitination pathway for disease treatment.","PeriodicalId":21766,"journal":{"name":"Signal Transduction and Targeted Therapy","volume":"154 1","pages":"424"},"PeriodicalIF":39.3,"publicationDate":"2025-12-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145813415","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Oncolytic adenovirus delivery of neoantigens sensitizes low-mutation tumors to anti-PD-1 therapy and prevents metastasis. 溶瘤腺病毒递送新抗原使低突变肿瘤对抗pd -1治疗增敏并阻止转移。
IF 39.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-23 DOI: 10.1038/s41392-025-02511-5
Ke-Yu Shen,Shi-Zhe Yu,Ying-Han Su,Sun-Zhe Xie,Chen Zhang,Hao Xu,SamI Yang,Tian-Tian Zou,Yan Fu,Hao Wang,Lin Fang,Yan Zheng,Chang-Qing Su,Lun-Xiu Qin
Neoantigen vaccines and oncolytic viruses are emerging immunotherapies that can reshape the tumor microenvironment (TME). However, tumors with low mutation burdens often respond poorly to immunotherapies because of their limited immunogenicity. Developing effective immunotherapy strategies for these types of tumors remains a significant challenge. In this study, we engineered oncolytic adenoviruses to accurately amplify neoantigen expression within tumor cells, which demonstrated superior efficacy compared to synthetic long peptide vaccines and showed enhanced effectiveness in a low mutation burden intrahepatic cholangiocarcinoma model. Building on this, we further developed NeoViron, which coexpresses neoantigens and Flt3L, a dendritic cell growth factor, to promote antigen presentation and T-cell infiltration simultaneously. NeoViron significantly inhibited tumor growth and prevented metastasis in intrahepatic cholangiocarcinoma animal models. Mechanistically, NeoViron enhanced the cytotoxicity of CD8+ T cells and promoted the expansion of CD69+ CD8+ tissue-resident memory T cells and TCF1+ CD8+ stem-like T cells to promote anti-tumor immunity and immune memory. When combined with anti-PD-1, it further enhances the cytotoxicity of tissue-resident memory T cells to eradicate solid tumors. These findings demonstrate that NeoViron can effectively sensitize low-mutation tumors to immunotherapy by increasing neoantigen expression and antigen-presentation efficacy, offering a promising strategy for cancer treatment, particularly for tumors with scarce neoantigens.
新抗原疫苗和溶瘤病毒是新兴的免疫疗法,可以重塑肿瘤微环境(TME)。然而,低突变负担的肿瘤由于其有限的免疫原性,往往对免疫疗法反应不佳。为这些类型的肿瘤开发有效的免疫治疗策略仍然是一个重大的挑战。在这项研究中,我们设计了溶瘤腺病毒来准确地扩增肿瘤细胞内的新抗原表达,与合成的长肽疫苗相比,这种方法的效果更好,并且在低突变负荷的肝内胆管癌模型中显示出更高的效果。在此基础上,我们进一步开发了NeoViron,它共表达新抗原和树突状细胞生长因子Flt3L,同时促进抗原呈递和t细胞浸润。在肝内胆管癌动物模型中,NeoViron显著抑制肿瘤生长和阻止转移。机制上,NeoViron增强CD8+ T细胞的细胞毒性,促进CD69+ CD8+组织驻留记忆T细胞和TCF1+ CD8+干细胞样T细胞的扩增,促进抗肿瘤免疫和免疫记忆。当与抗pd -1联合使用时,进一步增强组织驻留记忆T细胞的细胞毒性,以根除实体肿瘤。这些发现表明,NeoViron可以通过增加新抗原表达和抗原呈递效率,有效地使低突变肿瘤对免疫治疗敏感,为癌症治疗提供了一个有希望的策略,特别是对于缺乏新抗原的肿瘤。
{"title":"Oncolytic adenovirus delivery of neoantigens sensitizes low-mutation tumors to anti-PD-1 therapy and prevents metastasis.","authors":"Ke-Yu Shen,Shi-Zhe Yu,Ying-Han Su,Sun-Zhe Xie,Chen Zhang,Hao Xu,SamI Yang,Tian-Tian Zou,Yan Fu,Hao Wang,Lin Fang,Yan Zheng,Chang-Qing Su,Lun-Xiu Qin","doi":"10.1038/s41392-025-02511-5","DOIUrl":"https://doi.org/10.1038/s41392-025-02511-5","url":null,"abstract":"Neoantigen vaccines and oncolytic viruses are emerging immunotherapies that can reshape the tumor microenvironment (TME). However, tumors with low mutation burdens often respond poorly to immunotherapies because of their limited immunogenicity. Developing effective immunotherapy strategies for these types of tumors remains a significant challenge. In this study, we engineered oncolytic adenoviruses to accurately amplify neoantigen expression within tumor cells, which demonstrated superior efficacy compared to synthetic long peptide vaccines and showed enhanced effectiveness in a low mutation burden intrahepatic cholangiocarcinoma model. Building on this, we further developed NeoViron, which coexpresses neoantigens and Flt3L, a dendritic cell growth factor, to promote antigen presentation and T-cell infiltration simultaneously. NeoViron significantly inhibited tumor growth and prevented metastasis in intrahepatic cholangiocarcinoma animal models. Mechanistically, NeoViron enhanced the cytotoxicity of CD8+ T cells and promoted the expansion of CD69+ CD8+ tissue-resident memory T cells and TCF1+ CD8+ stem-like T cells to promote anti-tumor immunity and immune memory. When combined with anti-PD-1, it further enhances the cytotoxicity of tissue-resident memory T cells to eradicate solid tumors. These findings demonstrate that NeoViron can effectively sensitize low-mutation tumors to immunotherapy by increasing neoantigen expression and antigen-presentation efficacy, offering a promising strategy for cancer treatment, particularly for tumors with scarce neoantigens.","PeriodicalId":21766,"journal":{"name":"Signal Transduction and Targeted Therapy","volume":"45 1","pages":"410"},"PeriodicalIF":39.3,"publicationDate":"2025-12-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145807547","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Anti-MSLN chimeric antigen receptor-like NK cell therapy with tumor-penetrating capacity (uCAR-like NK) for solid tumors. 具有肿瘤穿透能力(uCAR-like NK)的抗msln嵌合抗原受体样NK细胞治疗实体瘤
IF 39.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-23 DOI: 10.1038/s41392-025-02524-0
Mengchao An,Ying Wang,Jie Shao,Siwen Wu,Jiayao Yan,Yuxiang Li,Liqing Zhong,Jingyi Guo,Tianran Chen,Manman Tian,Qin Liu,Rutian Li,Baorui Liu
Although natural killer (NK) cells are endowed with intrinsic cytotoxicity, their therapeutic application often faces limitations because of their lack of tumor-specific targeting ability and limited ability to infiltrate solid tumors. To overcome these limitations, we developed anti-mesothelin (MSLN) uCAR-like NK cells, which are designed to enhance both the targeting specificity and tumor infiltration capacity, thereby improving the antitumor efficacy of NK cell-based therapies. We constructed, purified, and validated a tetravalent bispecific cell engager (MSLN×CD16A) via the SpyTag/SpyCatcher system. Cytokine-induced memory-like NK cells, induced by IL-12, IL-15, and IL-18, were precomplexed with MSLN×CD16A to generate anti-MSLN CAR-like NK cells. To further enhance tumor penetration, the tumor-penetrating peptide uCendR was integrated into the system to construct anti-MSLN uCAR-like NK cells. In vitro, anti-MSLN CAR-like NK cells demonstrated selective cytotoxicity against MSLN-positive tumor cells through stable binding with MSLN×CD16A while sparing MSLN-negative cells. In xenograft models bearing MSLN-positive tumors, anti-MSLN CAR-like NK cells exhibited significant antitumor activity, with favorable tolerability and no significant body weight loss or toxicity. Notably, anti-MSLN uCAR-like NK cells, which integrate a tumor-penetrating peptide, displayed enhanced intratumor penetration and superior therapeutic efficacy. Overall, this study establishes a modular, nongenetically engineered uCAR-like NK platform that couples targeted recognition with enhanced tissue access. These findings highlight the potential of anti-MSLN CAR-like NK cells, particularly uCAR-like NK cells with enhanced tumor penetration, as promising therapeutic strategies for MSLN-positive solid tumors and lay the foundation for future clinical applications.
虽然自然杀伤细胞(natural killer, NK)具有固有的细胞毒性,但由于其缺乏肿瘤特异性靶向能力和浸润实体瘤的能力有限,其治疗应用往往受到限制。为了克服这些局限性,我们开发了抗间皮素(MSLN) ucar样NK细胞,旨在提高靶向特异性和肿瘤浸润能力,从而提高NK细胞为基础的治疗的抗肿瘤效果。我们通过SpyTag/SpyCatcher系统构建、纯化并验证了四价双特异性细胞接合器(MSLN×CD16A)。细胞因子诱导的记忆样NK细胞,由IL-12、IL-15和IL-18诱导,与MSLN×CD16A预复合生成抗msln car样NK细胞。为了进一步增强肿瘤穿透能力,我们将肿瘤穿透肽uCendR整合到体系中,构建抗msln ucar样NK细胞。在体外,抗msln car -样NK细胞通过与MSLN×CD16A的稳定结合,对msln阳性肿瘤细胞表现出选择性的细胞毒性,同时保留msln阴性细胞。在携带msln阳性肿瘤的异种移植模型中,抗msln car -样NK细胞表现出显著的抗肿瘤活性,具有良好的耐受性,没有明显的体重减轻或毒性。值得注意的是,整合肿瘤穿透肽的抗msln ucar样NK细胞表现出增强的肿瘤内穿透能力和优越的治疗效果。总的来说,本研究建立了一个模块化的、非基因工程的ucar样NK平台,将靶向识别与增强的组织通路结合起来。这些发现突出了抗msln car样NK细胞的潜力,特别是具有增强肿瘤穿透性的ucar样NK细胞,作为msln阳性实体瘤的有希望的治疗策略,并为未来的临床应用奠定了基础。
{"title":"Anti-MSLN chimeric antigen receptor-like NK cell therapy with tumor-penetrating capacity (uCAR-like NK) for solid tumors.","authors":"Mengchao An,Ying Wang,Jie Shao,Siwen Wu,Jiayao Yan,Yuxiang Li,Liqing Zhong,Jingyi Guo,Tianran Chen,Manman Tian,Qin Liu,Rutian Li,Baorui Liu","doi":"10.1038/s41392-025-02524-0","DOIUrl":"https://doi.org/10.1038/s41392-025-02524-0","url":null,"abstract":"Although natural killer (NK) cells are endowed with intrinsic cytotoxicity, their therapeutic application often faces limitations because of their lack of tumor-specific targeting ability and limited ability to infiltrate solid tumors. To overcome these limitations, we developed anti-mesothelin (MSLN) uCAR-like NK cells, which are designed to enhance both the targeting specificity and tumor infiltration capacity, thereby improving the antitumor efficacy of NK cell-based therapies. We constructed, purified, and validated a tetravalent bispecific cell engager (MSLN×CD16A) via the SpyTag/SpyCatcher system. Cytokine-induced memory-like NK cells, induced by IL-12, IL-15, and IL-18, were precomplexed with MSLN×CD16A to generate anti-MSLN CAR-like NK cells. To further enhance tumor penetration, the tumor-penetrating peptide uCendR was integrated into the system to construct anti-MSLN uCAR-like NK cells. In vitro, anti-MSLN CAR-like NK cells demonstrated selective cytotoxicity against MSLN-positive tumor cells through stable binding with MSLN×CD16A while sparing MSLN-negative cells. In xenograft models bearing MSLN-positive tumors, anti-MSLN CAR-like NK cells exhibited significant antitumor activity, with favorable tolerability and no significant body weight loss or toxicity. Notably, anti-MSLN uCAR-like NK cells, which integrate a tumor-penetrating peptide, displayed enhanced intratumor penetration and superior therapeutic efficacy. Overall, this study establishes a modular, nongenetically engineered uCAR-like NK platform that couples targeted recognition with enhanced tissue access. These findings highlight the potential of anti-MSLN CAR-like NK cells, particularly uCAR-like NK cells with enhanced tumor penetration, as promising therapeutic strategies for MSLN-positive solid tumors and lay the foundation for future clinical applications.","PeriodicalId":21766,"journal":{"name":"Signal Transduction and Targeted Therapy","volume":"25 1","pages":"425"},"PeriodicalIF":39.3,"publicationDate":"2025-12-23","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145813414","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Working together: a multi-component intranasal vaccine provides synergistic protection against COVID-19. 共同努力:多组分鼻内疫苗可提供针对COVID-19的协同保护。
IF 39.3 1区 医学 Q1 BIOCHEMISTRY & MOLECULAR BIOLOGY Pub Date : 2025-12-22 DOI: 10.1038/s41392-025-02508-0
Jessica A Breznik,Chris P Verschoor
{"title":"Working together: a multi-component intranasal vaccine provides synergistic protection against COVID-19.","authors":"Jessica A Breznik,Chris P Verschoor","doi":"10.1038/s41392-025-02508-0","DOIUrl":"https://doi.org/10.1038/s41392-025-02508-0","url":null,"abstract":"","PeriodicalId":21766,"journal":{"name":"Signal Transduction and Targeted Therapy","volume":"24 1","pages":"408"},"PeriodicalIF":39.3,"publicationDate":"2025-12-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145801281","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Signal Transduction and Targeted Therapy
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:604180095
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1