Journal of Nanobiotechnology最新文献

筛选
英文 中文
An ultra-sensitive and point-of-care time-resolved fluorescence immunochromatographic platform for trace-level brucellosis antibody rapid on-site detection. 一种用于痕量布鲁氏菌病抗体快速现场检测的超灵敏、即时分辨荧光免疫层析平台。
IF 10.6 1区 生物学
Journal of Nanobiotechnology Pub Date : 2025-06-05 DOI: 10.1186/s12951-025-03482-z
Bin Wang, Shusen Ji, Boya Li, Yue Jing, Ying Wang, Xiaomao Luo, Wei Hou, Huachuan Huang, Shouyu Wang, Zhangqi Shen, Haidong Wang
{"title":"An ultra-sensitive and point-of-care time-resolved fluorescence immunochromatographic platform for trace-level brucellosis antibody rapid on-site detection.","authors":"Bin Wang, Shusen Ji, Boya Li, Yue Jing, Ying Wang, Xiaomao Luo, Wei Hou, Huachuan Huang, Shouyu Wang, Zhangqi Shen, Haidong Wang","doi":"10.1186/s12951-025-03482-z","DOIUrl":"10.1186/s12951-025-03482-z","url":null,"abstract":"","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"416"},"PeriodicalIF":10.6,"publicationDate":"2025-06-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12139110/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144234378","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Advancing immunomodulation in organ transplantation: the therapeutic potential of self-assembled rapamycin nanoparticles in allograft rejection. 推进器官移植中的免疫调节:自组装雷帕霉素纳米颗粒治疗同种异体移植排斥反应的潜力。
IF 10.6 1区 生物学
Journal of Nanobiotechnology Pub Date : 2025-06-05 DOI: 10.1186/s12951-025-03498-5
Ruiqi Sun, Zhi Liang, Ning Wang, Xiaona Chen, Jialing Zhao, Hong Tang, Wentao Zhao, Hangxiang Wang, Shusen Zheng, Penghong Song, Haiyang Xie
{"title":"Advancing immunomodulation in organ transplantation: the therapeutic potential of self-assembled rapamycin nanoparticles in allograft rejection.","authors":"Ruiqi Sun, Zhi Liang, Ning Wang, Xiaona Chen, Jialing Zhao, Hong Tang, Wentao Zhao, Hangxiang Wang, Shusen Zheng, Penghong Song, Haiyang Xie","doi":"10.1186/s12951-025-03498-5","DOIUrl":"10.1186/s12951-025-03498-5","url":null,"abstract":"<p><strong>Background: </strong>Transplant rejection remains a significant challenge, necessitating effective post-transplant interventions. Although rapamycin (RAPA) is a recognized immunosuppressant, its utility is limited by poor solubility and delivery efficiency. This study investigates a self-assembly strategy to enhance the solubility and efficacy of RAPA against graft rejection.</p><p><strong>Methods: </strong>We synthesized soluble supramolecular rapamycin nanoparticles (sRNP) using reprecipitation, making RAPA injectable and stable in aqueous solutions.</p><p><strong>Results: </strong>sRNP maintained sustained therapeutic concentrations, exhibited minimal toxicity, and notably enhanced graft survival compared to traditional oral RAPA administration. In murine allograft models, sRNP treatment effectively suppressed T cell proliferation in peripheral immune organs and the circulatory system. Detailed analyses revealed that sRNP significantly increased the population of naive T cells while decreasing effector T cells. Mechanistic investigations indicated that these effects were mediated by the enhanced recruitment of myeloid-derived suppressor cells (MDSC) and the promotion of regulatory T cells homing to lymph nodes. This led to reduced differentiation of Th1 and Th17 cells, along with a decrease in inflammatory cytokines, resulting in significantly prolonged graft survival compared to oral RAPA. Additionally, in a rat orthotopic liver transplantation model, intermittent low-dose sRNP treatment (1 mg/kg every other day intravenously) effectively inhibited T cell proliferation, reduced inflammatory cell infiltration, markedly extended graft survival, and significantly improved liver function.</p><p><strong>Conclusions: </strong>This study highlights sRNP's superiority over oral RAPA in managing allograft rejection by enhancing immune regulation, reducing T cell differentiation, and decreasing inflammation. These effects extend graft survival, underscoring sRNP's potential as an effective anti-rejection therapy.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"418"},"PeriodicalIF":10.6,"publicationDate":"2025-06-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12139188/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144234377","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Orchestrated Cu2+-coordinated tetracycline-porphyrin self-assembly remodels tumor microenvironment for photo-enhanced immuno-chemodynamic therapy. Cu2+协调四环素-卟啉自组装重塑肿瘤微环境,用于光增强免疫化学动力学治疗。
IF 10.6 1区 生物学
Journal of Nanobiotechnology Pub Date : 2025-06-05 DOI: 10.1186/s12951-025-03486-9
Xianbin Sun, Xinyi Wang, Xudong Li, Ya Wang, Ruofei Xu, Xinyi Shen, Ding Tan, Jingjing Liao, Haijun Chen, Yu Gao, Jianping Zheng
{"title":"Orchestrated Cu<sup>2+</sup>-coordinated tetracycline-porphyrin self-assembly remodels tumor microenvironment for photo-enhanced immuno-chemodynamic therapy.","authors":"Xianbin Sun, Xinyi Wang, Xudong Li, Ya Wang, Ruofei Xu, Xinyi Shen, Ding Tan, Jingjing Liao, Haijun Chen, Yu Gao, Jianping Zheng","doi":"10.1186/s12951-025-03486-9","DOIUrl":"10.1186/s12951-025-03486-9","url":null,"abstract":"","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"419"},"PeriodicalIF":10.6,"publicationDate":"2025-06-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12139202/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144234379","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Cell-laden biomimetic microneedles reconstruct skin rete ridge and stem cell niche. 满载细胞的仿生微针重建皮肤网状嵴和干细胞生态位。
IF 10.6 1区 生物学
Journal of Nanobiotechnology Pub Date : 2025-06-04 DOI: 10.1186/s12951-025-03430-x
Xiaohong Zhao, Zongze Wu, Yicheng Guo, Lei Pu, Zixuan Pei, Yuanyuan Liu, Biao Hou, Songlin Xie, Gaoxing Luo, Rixing Zhan
{"title":"Cell-laden biomimetic microneedles reconstruct skin rete ridge and stem cell niche.","authors":"Xiaohong Zhao, Zongze Wu, Yicheng Guo, Lei Pu, Zixuan Pei, Yuanyuan Liu, Biao Hou, Songlin Xie, Gaoxing Luo, Rixing Zhan","doi":"10.1186/s12951-025-03430-x","DOIUrl":"10.1186/s12951-025-03430-x","url":null,"abstract":"<p><p>The traditional skin graft wound repair strategy for burns is unable to reconstruct the normal anatomy of the skin, resulting in quality problems such as scarring, which have always been the bottleneck of burn medicine. The skin rete ridge (RRs) is an important basis for maintaining skin homeostasis, but the complete reconstruction of the RRs in wound repair is still difficult with traditional split-thickness skin grafting and wound cell therapy. In this study, based on our previous experience of wound epidermal stem cell (EpiSCs) therapy, we further designed and optimized the bionic RRs microneedles loaded with human EpiSCs (C-Ms). Transplanting C-Ms into full-thickness wounds in nude mice promotes the formation of RRs similar to natural human epidermis and provides stem cell niches. Pathway enrichment analysis showed that C-Ms transplantation into wounds reshapes the extracellular matrix (ECM) and reduces wound healing fibrosis through FAK, ECM receptor interaction signaling pathways. Moreover, C-Ms improve wound healing by accelerating early vascularization, and they also regulate the proliferation of EpiSCs to further promote the formation of RRs structure. These results suggest that C-Ms can compensate for the lack of skin anatomical structure, which may be suitable for use in clinical patients with large-scale burns.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"415"},"PeriodicalIF":10.6,"publicationDate":"2025-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12135421/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144225698","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Sirtuin-3 activation by honokiol attenuated anesthesia/surgery-induced cognitive impairment and neuronal ferroptosis via inhibiting mitochondrial GPX4 acetylation. 通过抑制线粒体GPX4乙酰化,厚木酚激活Sirtuin-3可减轻麻醉/手术诱导的认知障碍和神经元凋亡。
IF 10.6 1区 生物学
Journal of Nanobiotechnology Pub Date : 2025-06-04 DOI: 10.1186/s12951-025-03502-y
Lian Zeng, Pengchao Hu, Xuan Wang, Xudong Ding, Qingsong Wang, Li Luo, Yu Zhang, Mingyue Li, Yilin Zhao, Shiyong Li, Ailin Luo
{"title":"Sirtuin-3 activation by honokiol attenuated anesthesia/surgery-induced cognitive impairment and neuronal ferroptosis via inhibiting mitochondrial GPX4 acetylation.","authors":"Lian Zeng, Pengchao Hu, Xuan Wang, Xudong Ding, Qingsong Wang, Li Luo, Yu Zhang, Mingyue Li, Yilin Zhao, Shiyong Li, Ailin Luo","doi":"10.1186/s12951-025-03502-y","DOIUrl":"10.1186/s12951-025-03502-y","url":null,"abstract":"<p><strong>Background: </strong>Perioperative neurocognitive disorder (PND), a prevalent neurological complication in elderly patients following anesthesia and surgery, has recently been linked to ferroptosis as a central pathogenic mechanism. Sirtuin-3 (SIRT3), a NAD<sup>+</sup>-dependent deacetylase, exhibits neuroprotective properties in neurodegenerative disorders, including PND. However, its role in neuronal ferroptosis during PND remains unclear. This study investigates the impact of SIRT3 on ferroptosis modulation in PND and its underlying mechanism.</p><p><strong>Methods: </strong>A murine model of PND was established using tibial fracture surgery under isoflurane anesthesia to assess SIRT3 expression and cognitive function. Mice were treated with Honokiol (HKL) or erastin to evaluate hippocampal ferroptosis. RNA sequencing (RNA-seq) was performed to identify the underlying neuroprotective mechanisms. In vitro, PC12 and HT22 cells were treated with erastin or HKL to analyze ferroptosis markers. GPX4 silencing in HT22 cells was used to validate the effect of HKL in modulating ferroptosis. Adeno-associated virus (AAV)-mediated overexpression of SIRT3 and Co-immunoprecipitation (Co-IP), were employed to further elucidate its mechanism in suppressing ferroptosis.</p><p><strong>Results: </strong>SIRT3 expression was found to be reduced in the hippocampal CA1 and CA3 regions post-surgery. HKL alleviated cognitive decline by inhibiting ferroptosis, evidenced by suppression of iron accumulation, oxidative stress, and mitochondrial dysfunction. In erastin-treated PC12 and HT22 cells, HKL effectively counteracted ferroptosis, which was abolished by GPX4 silencing. SIRT3 overexpression in the mouse hippocampus suppressed anesthesia/surgery-induced neuronal ferroptosis. Mechanistically, HKL-activated SIRT3 upregulated mitochondrial GPX4 expression and reduced its acetylation, thereby inhibiting neuronal ferroptosis.</p><p><strong>Conclusions: </strong>SIRT3 activation by HKL alleviates hippocampal neuronal ferroptosis in PND by suppressing mitochondrial GPX4 acetylation, providing a novel therapeutic strategy for the management of PND.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"414"},"PeriodicalIF":10.6,"publicationDate":"2025-06-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12135415/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144216113","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
In situ and dynamic screening of extracellular vesicles as predictive biomarkers in immune-checkpoint inhibitor therapies. 细胞外囊泡作为免疫检查点抑制剂治疗的预测性生物标志物的原位和动态筛选。
IF 10.6 1区 生物学
Journal of Nanobiotechnology Pub Date : 2025-06-03 DOI: 10.1186/s12951-025-03467-y
Yihe Wang, Yue Sun, Mengqi Liu, Chao Wang, Miao Huang, Jiaoyan Qiu, Ningkai Yang, Yu Zhang, Hong Liu, Lin Han
{"title":"In situ and dynamic screening of extracellular vesicles as predictive biomarkers in immune-checkpoint inhibitor therapies.","authors":"Yihe Wang, Yue Sun, Mengqi Liu, Chao Wang, Miao Huang, Jiaoyan Qiu, Ningkai Yang, Yu Zhang, Hong Liu, Lin Han","doi":"10.1186/s12951-025-03467-y","DOIUrl":"10.1186/s12951-025-03467-y","url":null,"abstract":"<p><p>Extracellular vesicles (EVs) is promising in predicting the efficacy of immune checkpoint inhibitor (ICI) therapies. But it is challenging to determine the level of circulating EVs due to their variations in spatial and temporal distribution. To address this, we developed an in situ EV detection platform integrating multiplex EV capture with microfluidic-generated immune-tumor spheroids. This platform enables in situ monitoring of EV secretion dynamics under ICI and chemotherapeutic treatments, capturing localized and temporal changes in EV release. Using predictive models, we identified EVs carrying programmed cell death ligand 1 (PD-L1) as the most robust predictors of spheroid viability during treatment. RNA sequencing further revealed that dynamic EV expression changes are driven by gene transcription, providing a temporal understanding of EV regulation. Our platform overcomes the limitations of traditional methods by offering a physiologically relevant system to study EV-mediated immune responses. By addressing the spatial and temporal heterogeneity of EVs, this work advances EV-based biomarker discovery and provides a foundation for optimizing personalized immunotherapies.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"411"},"PeriodicalIF":10.6,"publicationDate":"2025-06-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12131400/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144208734","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","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 CD39 boosts PD-1 blockade antitumor therapeutic efficacy via strengthening CD8 + TILs function and recruiting B cells in cervical cancer. 靶向CD39可通过增强CD8 + TILs功能和募集B细胞增强PD-1阻断子宫颈癌的抗肿瘤治疗效果。
IF 10.6 1区 生物学
Journal of Nanobiotechnology Pub Date : 2025-06-03 DOI: 10.1186/s12951-025-03500-0
Lili Jiang, Tong Wu, Xinyu Qu, Shuqi Li, Qi'an Jiang, Tingting Ren, Jiali Liang, Yan Ding, Keqin Hua, Zhongmin Tang, Junjun Qiu
{"title":"Targeting CD39 boosts PD-1 blockade antitumor therapeutic efficacy via strengthening CD8 + TILs function and recruiting B cells in cervical cancer.","authors":"Lili Jiang, Tong Wu, Xinyu Qu, Shuqi Li, Qi'an Jiang, Tingting Ren, Jiali Liang, Yan Ding, Keqin Hua, Zhongmin Tang, Junjun Qiu","doi":"10.1186/s12951-025-03500-0","DOIUrl":"10.1186/s12951-025-03500-0","url":null,"abstract":"<p><p>Although the programmed cell death protein 1 (PD-1) blockade has been authorized for the treatment of recurrent and metastatic cervical cancer (CC) patients, a significant proportion of CC patients show low objective response rates (ORR) to immune checkpoint blockades (ICBs). Therefore, identifying novel combination treatment strategies to enhance ICBs therapeutic efficacy for CC patients is urgently needed. Here, we discovered that CD39 was highly expressed in exhausted CD8 + T cells from 10 CC patients in our center via single-cell RNA sequencing (scRNA-seq). Furthermore, we validated that CC patients with CD39 highly expressed in CD8 + T cells associated with poor prognosis and immunoevasive subtype of CC both in cohort from our center and the Cancer Genome Atlas (TCGA) database. Moreover, it was also confirmed that CD39-inhibiting not only enhanced the cytotoxicity of CD8 + tumor-infiltrating lymphocytes (TILs) but also promoted the infiltration of B cells through increasing CXCL13 secretion both in vitro experiments and subcutaneous tumor models, thereby amplifying anti-tumor immunity of PD-1 blockade. What was more, we have developed a liposome containing POM-1, which effectively enhanced the anti-tumor effect of POM-1. Our findings provide compelling evidence that targeting CD39 represents a promising \"two birds with one stone\" strategy for cervical cancer treatment.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"413"},"PeriodicalIF":10.6,"publicationDate":"2025-06-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12131488/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144216114","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Biomimetic nanocomplexes loading with evolocumab and curcumin for synergistic anti-atherosclerosis therapy in ApoE-/- mice. 载evolocumab和姜黄素的仿生纳米复合物对ApoE-/-小鼠的协同抗动脉粥样硬化治疗。
IF 10.6 1区 生物学
Journal of Nanobiotechnology Pub Date : 2025-06-03 DOI: 10.1186/s12951-025-03444-5
Yi Liu, Shengchao Ma, Feng Li, Hanshuang Ding, Qi Zhang, Feifei Yu, Huiping Zhang, Yinju Hao, Bin Liu, Yideng Jiang
{"title":"Biomimetic nanocomplexes loading with evolocumab and curcumin for synergistic anti-atherosclerosis therapy in ApoE<sup>-/-</sup> mice.","authors":"Yi Liu, Shengchao Ma, Feng Li, Hanshuang Ding, Qi Zhang, Feifei Yu, Huiping Zhang, Yinju Hao, Bin Liu, Yideng Jiang","doi":"10.1186/s12951-025-03444-5","DOIUrl":"10.1186/s12951-025-03444-5","url":null,"abstract":"<p><p>Atherosclerosis (AS), a leading contributor to global cardiovascular mortality, is primarily driven by the dual pathological processes of chronic persistent inflammation and dysregulated lipid metabolism. Current clinical interventions are predominantly limited to single-target approaches (e.g., lipid-lowering therapies), which are insufficient for simultaneously modulating the two pathophysiological mechanisms and inhibiting atherosclerotic progression. Recently, combination therapeutic strategies based on multi-target and multi-organ synergistic effects have gained increasing attention in AS treatment. In this study, we developed a dual-functional nanodelivery system co-encapsulating PCSK9 inhibitor of evolocumab and natural anti-inflammatory agent of curcumin, with surface modification using macrophage membranes (Møm) and hyaluronic acid (HA). This novel design not only confers immune evasion capability to the nanocomplex but also facilitates drug accumulation in atherosclerotic lesions and hepatic tissues, thereby enabling synchronous regulation of the inflammatory microenvironment and lipid metabolic homeostasis. In vivo studies demonstrated remarkable therapeutic efficacy of this nanoformulation on atherosclerosis by effectively reducing plaque area, enhancing plaque stability and markedly ameliorating hepatic lipid accumulation. Overall, the proposed strategy, which enables multi-target and multi-organ synergistic regulation of inflammatory responses and lipid metabolism disorder, provides a promising approach for the clinical management of atherosclerosis.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"412"},"PeriodicalIF":10.6,"publicationDate":"2025-06-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12131610/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144216112","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Targeted tumor cell-intrinsic CTRP6 biomimetic codelivery synergistically amplifies ferroptosis and immune activation to boost anti-PD-L1 immunotherapy efficacy in lung cancer. 靶向肿瘤细胞内生性CTRP6仿生共递送协同放大铁凋亡和免疫激活,提高肺癌抗pd - l1免疫治疗效果。
IF 10.6 1区 生物学
Journal of Nanobiotechnology Pub Date : 2025-06-02 DOI: 10.1186/s12951-025-03428-5
Songhua Cai, Jing Huang, Hongjie Fan, Zhilin Sui, Chujian Huang, Youjun Deng, Ran Jia, Lixu Wang, Kai Ma, Xiaotong Guo, Jie He, Baihua Zhang, Zhentao Yu
{"title":"Targeted tumor cell-intrinsic CTRP6 biomimetic codelivery synergistically amplifies ferroptosis and immune activation to boost anti-PD-L1 immunotherapy efficacy in lung cancer.","authors":"Songhua Cai, Jing Huang, Hongjie Fan, Zhilin Sui, Chujian Huang, Youjun Deng, Ran Jia, Lixu Wang, Kai Ma, Xiaotong Guo, Jie He, Baihua Zhang, Zhentao Yu","doi":"10.1186/s12951-025-03428-5","DOIUrl":"10.1186/s12951-025-03428-5","url":null,"abstract":"<p><strong>Background: </strong>Lung adenocarcinoma (LUAD) is a refractory tumor with high incidence, high mortality, and easy development of drug resistance. Represented by PD-L1, the rise of immunotherapy and multidrug combinations offers a reliable approach to treating LUAD. However, there are still some patients with immunoresistance or insensitivity, and new combination therapies are still needed.</p><p><strong>Methods: </strong>In this study, cyclic arginine-glycine-aspartate (cRGD)/erythrocyte membrane (RBCM) double-head nanocarriers were designed. This nanocarrier platform targets CTRP6 in tumors and delivers gemcitabine to block the progression of lung cancer. PD-L1 monoclonal antibodies were used as a codelivery platform to explore the effect of the codelivery platform on immunotherapy.</p><p><strong>Results: </strong>CTRP6 expression was negatively correlated with the prognosis of patients with lung adenocarcinoma. The codelivery platform @RBCM/cRGD-PhLips effectively targeted tumor cells. Co-carrying gemcitabine and targeting CTRP6 expression, it amplified ferroptosis of tumor cells through the NRF2/STAT3 signaling pathway, activated intratumoral immunity, and promoted M1-like macrophage transformation and CD8<sup>+</sup> T-cell recruitment. This platform amplified the immune effect of PD-L1 monoclonal antibodies to play an anti-lung cancer role.</p><p><strong>Conclusions: </strong>The synergistic delivery of the targeted tumor cell-intrinsic CTRP6 biomimetic nanocarrier provides a new approach to the combined immunotherapy of lung cancer.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"409"},"PeriodicalIF":10.6,"publicationDate":"2025-06-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12131836/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144208736","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Fucoidan-decorated metal-zoledronic acid nanocomplexes suppress tumor metastasis by inducing ferroptotic cell death and enhancing cancer immunotherapy. 岩藻糖烷修饰的金属-唑来膦酸纳米复合物通过诱导铁致细胞死亡和增强肿瘤免疫治疗抑制肿瘤转移。
IF 10.6 1区 生物学
Journal of Nanobiotechnology Pub Date : 2025-06-02 DOI: 10.1186/s12951-025-03473-0
Hsin-Ting Tsai, Chi Lin, Chu-Hung Chung, Wen-Jing Hsu, Ming-Yi Hsieh, Ming-Cheng Chiang, Tzu-Wei Lu, Fwu-Long Mi, Cheng-Wei Lin
{"title":"Fucoidan-decorated metal-zoledronic acid nanocomplexes suppress tumor metastasis by inducing ferroptotic cell death and enhancing cancer immunotherapy.","authors":"Hsin-Ting Tsai, Chi Lin, Chu-Hung Chung, Wen-Jing Hsu, Ming-Yi Hsieh, Ming-Cheng Chiang, Tzu-Wei Lu, Fwu-Long Mi, Cheng-Wei Lin","doi":"10.1186/s12951-025-03473-0","DOIUrl":"10.1186/s12951-025-03473-0","url":null,"abstract":"<p><p>Metastatic tumor cells that escape from immune surveillance are a dilemma in cancer treatment, and thus developing selective targeting agents to treat metastatic tumor and reinstate immune perception is imperative for clinical applications. Herein, a multifunctional nanoplatform of supramolecular assembled nanoparticles (SANs) comprising a core structure of metal ion (Fe<sup>3+</sup>) and organic ligands including tannic acid (TA), and zoledronic acid (Zol) was developed. The FTZ SANs was further decorated with fucoidan (Fu), a P-selectin ligand, which greatly enhanced specific binding affinity of FTZ@Fu SANs towards metastatic tumor cells and suppressed tumor aggressiveness. The Fe-TA-Zol coordination network constructed through competitive ligand substitution facilitated the releases of Zol in response to the acidic tumor microenvironment (TME), which also benefited iron redox cycling of the Fenton reaction and further trigger ferritinophagy. Subsequently, Zol coordinately exerted ferroptotic-inducing activity accompanied by induction of stimulator of interferon genes (STING) pathway to aggravate immunogenic cell death (ICD) and enhance the antitumor immune response. Furthermore, FTZ@Fu effectively attenuated the immunosuppressive TME to suppress tumor growth and distant metastasis, and FTZ@Fu potentiated the therapeutic efficacy in combination with immune checkpoint blockade (ICB) therapy. Importantly, FTZ@Fu SANs suppressed metastatic tumor growth and reshaped the immune microenvironment. Our nanosystem provides a promising avenue for synergetic cancer targeting and chemoimmunotherapy, paving the way for targeted therapeutic strategies.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"405"},"PeriodicalIF":10.6,"publicationDate":"2025-06-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12128240/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144199362","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
确定
请完成安全验证×
相关产品
×
本文献相关产品
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信