{"title":"Hemoglobin-loaded hollow mesoporous carbon-gold nanocomposites enhance microwave ablation through hypoxia relief.","authors":"Yitian Zhang, Bitao Li, Jiawen He, Ya Meng, Meixiao Zhan, Cuixia Lu, Yong Li, Feiyu Niu, Liewei Wen","doi":"10.1186/s12951-025-03387-x","DOIUrl":"https://doi.org/10.1186/s12951-025-03387-x","url":null,"abstract":"<p><p>Microwave ablation, as a critical minimally invasive technique for tumor treatment, remains challenging in achieving an optimal balance between incomplete and excessive ablation. In addition to selectively elevating the temperature of tumor lesions through the microwave thermal effect, microwave-responsive nanoparticles can also improve the efficacy of single-session ablation by generating reactive oxygen species (ROS) via the microwave dynamic effect, thereby mitigating the thermal damage to normal tissues caused by high temperature. In this study, ultra-small gold nanoparticles anchored hollow mesoporous carbon nanoparticles (HMCNs) are loaded with hemoglobin (Hb) to serve as microwave ablation nano-sensitizers (HMCN/Au@Hb), which will amplify the microwave dynamic effect by alleviating the hypoxic microenvironment of tumors. Upon microwave irradiation, HMCN/Au@Hb not only improves the microwave-thermal conversion efficiency of tumor lesion but also promotes the ROS generation by increasing oxygen content in the hypoxic tumor microenvironment. More importantly, we found that the hypoxia relief will improve the antitumor response and further enhance the clearance of residual tumor after ablation. Nearly complete ablation was achieved in certain tumor-bearing mice, with no recurrence of the primary tumor observed up to 33 days post-ablation. In comparison to traditional microwave ablation, the survival time of the tumor-bearing mice was significantly extended. Therefore, this work presents an innovative ablation sensitization strategy based on the hypoxia relief and provides a nanosensitizer for microwave ablation integrating great microwave-thermal and dynamic effects along with immune modulation capabilities.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"326"},"PeriodicalIF":10.6,"publicationDate":"2025-04-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12042322/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144022394","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}
Tangbing Xu, Qiming Ma, Chi Zhang, Xiaoyan He, Qian Wang, Yunfeng Wu, Kunpeng Qin, Faxue Liao, Ping Zhou, Pengfei Xu, Jialai Yang, Junjun Yang, Jun Chang, Lei Qiao, Yong Hu
{"title":"A novel nanomedicine for osteosarcoma treatment: triggering ferroptosis through GSH depletion and inhibition for enhanced synergistic PDT/PTT therapy.","authors":"Tangbing Xu, Qiming Ma, Chi Zhang, Xiaoyan He, Qian Wang, Yunfeng Wu, Kunpeng Qin, Faxue Liao, Ping Zhou, Pengfei Xu, Jialai Yang, Junjun Yang, Jun Chang, Lei Qiao, Yong Hu","doi":"10.1186/s12951-025-03380-4","DOIUrl":"https://doi.org/10.1186/s12951-025-03380-4","url":null,"abstract":"<p><p>Osteosarcoma treatment remains challenging due to the limitations of single-modality therapies. To address this, we designed a carrier-free nanomedicine SRF@CuSO4.5H2O@IR780 (CSIR) for synergistic ferroptosis, photodynamic therapy (PDT), and photothermal therapy (PTT) in osteosarcoma. Interestingly, CSIR could harness the enhanced permeability and retention (EPR) effect to effectively enter tumors. Copper ions (Cu<sup>2+</sup>) within CSIR could react with the reductive intracellular environment, depleting glutathione (GSH) levels. Near-infrared (NIR) irradiation of CSIR further depleted GSH through reactive oxygen species (ROS) generation. Additionally, CSIR released sorafenib (SRF), which inhibited cystine-glutamate antiporter system xCT (xCT), thereby blocking GSH biosynthesis. RNA sequencing data confirmed ferroptosis induction by CSIR. This synergistic strategy of GSH depletion-induced ferroptosis, enhanced PDT, and photothermal cascade holds promise for improved osteosarcoma treatment and future nanomedicine design.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"323"},"PeriodicalIF":10.6,"publicationDate":"2025-04-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12039277/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143970398","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}
{"title":"Ferrocene-derived magnetic fiber-particles from diesel exhaust: enhanced pulmonary toxicity via Bach1-SAT1-polyamine depletion.","authors":"Xinxian Gong, Siyi Wang, Junhua Yuan, Jing Ji, Rui Zhao, Jing Huang, Boyang Li, Yunuo Zhai, Yuxu Zhong, Yuxin Zheng, Qixiao Jiang","doi":"10.1186/s12951-025-03397-9","DOIUrl":"https://doi.org/10.1186/s12951-025-03397-9","url":null,"abstract":"<p><strong>Background and aim: </strong>Magnetic nanoparticles are key components of air pollution. The combustion of diesel engine fuels, especially with ferrocene doping to reduce emissions, may increase exposure to these particles and related health risks. This study aimed to reveal the generation and characterization of ferrocene-derived magnetic particles (FMP) in ferrocene-doped diesel exhaust, and to investigate its toxicities and associated mechanisms in an avian model.</p><p><strong>Methods: </strong>FMP was observed in ferrocene-doped diesel exhaust particles, and extracted with neodymium magnets. Extracted FMP was characterized, and exposed to hatchling chickens via aerosol inhalation. Pulmonary toxicities were assessed with pathological and molecular methods. Associated mechanisms were investigated with RNA-seq, in vitro cell culture, and in vivo gene silencing.</p><p><strong>Results: </strong>FMP was characterized to be fibrous, magnetic iron-containing carbon particles. Extracted FMP could directly induce pulmonary toxicity. Mechanistic investigations revealed molecular mechanism associated with ferroptosis via Bach1, SAT1 and polyamines depletion, and further confirmed with ferroptosis inhibitor treatment, Bach1 inhibitor treatment, supplementation of polyamines or SAT1 silencing.</p><p><strong>Conclusions: </strong>Ferrocene doping could result in formation of magnetic particles in diesel exhaust. For the first time, magnetic fiber-like particles were extracted from ferrocene-doped DE particles, which is a potential source of magnetic particles in air pollution. To better balance emission control and health effects, further investigations are necessary.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"324"},"PeriodicalIF":10.6,"publicationDate":"2025-04-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12039003/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144017487","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}
{"title":"Single BMSC-derived cartilage organoids for gradient heterogeneous osteochondral regeneration by leveraging native vascular microenvironment.","authors":"Zhenying Chen, Qitao Bo, Chao Wang, Yong Xu, Xiang Fei, Ru Chen","doi":"10.1186/s12951-025-03403-0","DOIUrl":"https://doi.org/10.1186/s12951-025-03403-0","url":null,"abstract":"<p><p>Heterogeneous osteochondral regeneration remains a significant challenge due to the distinct microenvironments across the cartilage, calcified cartilage, and subchondral bone layers. The natural gradient of vascularization from the superficial to deep layers of osteochondral tissue plays a critical role in guiding the differentiation of bone marrow stem cells (BMSCs) into chondrocytes and osteoblasts. In this study, we propose a strategy for gradient heterogeneous osteochondral regeneration using cartilage organoids derived from single BMSCs, leveraging the natural vascularization gradient within osteochondral tissue. We successfully isolated BMSCs from rabbits and generated cartilage organoids via in vitro three-dimensional chondrogenic culture. To mimic the pro-vascular microenvironment, we introduced vascular endothelial growth factor, which promoted the hypertrophic differentiation of the cartilage organoids. We then prepared cartilage organoid/GelMA complexes, with or without the anti-vascular drug Axitinib, and implanted them subcutaneously in nude mice. The vascularized subcutaneous microenvironment induced osteogenic differentiation, while Axitinib treatment created an anti-vascular microenvironment, inhibiting osteogenesis and preserving chondrogenesis within the complexes. Both in vitro and in vivo data demonstrated the crucial role of the vascular microenvironment in regulating osteogenic differentiation of cartilage organoids. Finally, organoid/GelMA cylinders were implanted into a rabbit osteochondral defect, where the gradient vascularization at the defect site guided the organoids to differentiate into both cartilage and bone. This single BMSC-derived cartilage organoid approach enables precise gradient heterogeneous osteochondral regeneration, guided by the natural microenvironment within the osteochondral defect site, representing a significant advancement for clinical applications.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"325"},"PeriodicalIF":10.6,"publicationDate":"2025-04-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12042616/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144021447","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}
Ruoyan Wei, Yunzhe Wang, Ziqing Feng, Rui Liu, Chang Liu, Xiaojun Hu, Yujia Liu, Bin Kong, Xingtao Zhou, Meiyan Li
{"title":"Self-healing adhesive oxidized guar gum hydrogel loaded with mesenchymal stem cell exosomes for corneal wound healing.","authors":"Ruoyan Wei, Yunzhe Wang, Ziqing Feng, Rui Liu, Chang Liu, Xiaojun Hu, Yujia Liu, Bin Kong, Xingtao Zhou, Meiyan Li","doi":"10.1186/s12951-025-03366-2","DOIUrl":"https://doi.org/10.1186/s12951-025-03366-2","url":null,"abstract":"<p><p>Hydrogels have shown great potential and value in wound healing. In this study, we construct a mesenchymal stem cell exosomes (MSC-Exos) laden natural biopolymer-based hydrogel with transparent, self-healing, injectable, and tissue adhesive properties to promote corneal regeneration. This hydrogel is synthesized by combining aldehyde-modified oxidized guar gum (OGG) and carboxymethyl chitosan (CMCS) through the dynamic, reversible Schiff base bonds, which endow it with outstanding shear-thinning and self-healing properties, facilitating easy injection through a needle. The physicochemical properties, such as porosity, mechanical strength, and light transmittance, could be precisely tunable by adjusting OGG concentrations. The resultant hydrogel achieves robust tissue adhesion at physiological temperatures due to Schiff base interactions. Besides, the Exos can be uptaken by the corneal epithelial cells and subsequently promote the migration of the cells. We have proven that the MSC-Exos-loaded hydrogel adheres firmly to the defected cornea and significantly improves wound repair by enhancing collagen deposition and reducing inflammation in a rabbit cornea defect model. These results indicated that this multifunctional hydrogel holds immense scientific promise and offers a wide range of clinical applications.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"321"},"PeriodicalIF":10.6,"publicationDate":"2025-04-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12036215/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144015724","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}
Su Young Oh, Dong Yeon Kim, Kah Young Lee, Dae-Lyong Ha, Tae-Lyn Kim, Tae-Geon Kwon, Jin-Wook Kim, Heon-Jin Lee, So-Young Choi, Su-Hyung Hong
{"title":"Streptococcus mutans-derived extracellular vesicles promote skin wound healing via tRNA cargo.","authors":"Su Young Oh, Dong Yeon Kim, Kah Young Lee, Dae-Lyong Ha, Tae-Lyn Kim, Tae-Geon Kwon, Jin-Wook Kim, Heon-Jin Lee, So-Young Choi, Su-Hyung Hong","doi":"10.1186/s12951-025-03410-1","DOIUrl":"https://doi.org/10.1186/s12951-025-03410-1","url":null,"abstract":"<p><strong>Background: </strong>The human oral cavity harbors a diverse microbiota, including Streptococcus species. Oral mucosal wounds heal rapidly, although the exact cause remains unclear. This study investigates the impact of Streptococcus mutans-derived extracellular vesicles (Sm EVs) on wound healing in both oral mucosal organoids and mouse skin. To explore whether microbial EV RNA cargo influences wound healing, RNA sequences from Sm EVs were identified, and the most abundant sequences were synthesized into oligomers and encapsulated in E. coli EVs (Ec EVs) for further in vivo testing. We assessed the role of Toll-like receptor 3 (TLR3) in the wound healing mechanism in TLR3 knockout (KO) mice.</p><p><strong>Results: </strong>Sm EVs significantly enhanced cell proliferation and migration in oral mucosa, with enhanced focal adhesion complex formation. Sm EVs improved wound healing in mouse dorsal skin compared to PBS controls. RNA sequencing revealed that bacterial tRNAs, particularly the tRNA-Met variant (Oligo 1), were the most abundant RNAs in Sm EVs. Ec EVs carrying Oligo 1 produced similar wound healing effects to Sm EVs in mucosal organoids and mouse dorsal skin. However, in TLR3 knockout mice, Oligo 1 did not improve wound healing.</p><p><strong>Conclusions: </strong>This study highlights the role of Sm EVs, particularly their tRNA variants, in promoting skin wound healing through a TLR3-dependent mechanism. These findings suggest that EVs from oral commensal bacteria may offer therapeutic potential for chronic, non-healing skin wounds.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"322"},"PeriodicalIF":10.6,"publicationDate":"2025-04-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12036164/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144001989","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}
Keng Ye, Kongwen Lin, Chengkun Wu, Zhenhuan Zou, Yang Zhu, Yanfang Xu
{"title":"Ultrasmall platinum single-atom enzyme alleviates oxidative stress and macrophage polarization induced by acute kidney ischemia-reperfusion injury through inhibition of cell death storm.","authors":"Keng Ye, Kongwen Lin, Chengkun Wu, Zhenhuan Zou, Yang Zhu, Yanfang Xu","doi":"10.1186/s12951-025-03392-0","DOIUrl":"https://doi.org/10.1186/s12951-025-03392-0","url":null,"abstract":"<p><p>Acute kidney injury (AKI), characterized by a rapid decline in renal function, is associated with impaired mitochondrial function and excessive reactive oxygen species (ROS). Therefore, the exploration of ROS scavengers provides promising new opportunities for the prevention and treatment of AKI by mitigating oxidative stress. Here, we construct an ultrasmall platinum single-atom enzyme (Pt/SAE) with multiple antioxidant enzyme activities to protect against acute kidney ischemia-reperfusion (I/R) injury. Pt/SAE not only mimics superoxide dismutase and catalase activities to convert superoxide anion into water and oxygen, but also exhibits impressive hydroxyl radical scavenging capacity, thereby reducing pro-inflammatory macrophage levels and preventing inflammation. Furthermore, Pt/SAE reduces the accumulation of Z-form DNA, which excessively accumulates following I/R damage, thus decreasing its interaction with Z-DNA binding protein 1, consequently preventing the progression of PANoptosis following I/R stress. Additionally, the downregulation of ROS levels induced by Pt/SAE suppresses lipid peroxidation, which in return preventing the progression of ferroptosis following I/R. Both in vitro and in vivo experiments confirm that Pt/SAE effectively mitigates inflammatory cell infiltration and promotes a shift in macrophage polarization from the M1-like to M2-like subtype. This study provides promising information for the development of novel SAEs as a viable treatment method for AKI.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"320"},"PeriodicalIF":10.6,"publicationDate":"2025-04-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12034168/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144014619","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}
Yanguo Su, Rui Chen, Bingjie Wang, Teng Wang, Jiaojiao Tao, Qijie Diao, Tianze Jiang, Xia Zhao
{"title":"Erythrocyte membrane camouflaged celastrol and bilirubin self-assembly for rheumatoid arthritis immunotherapy based on STING inhibition and RONS clearance.","authors":"Yanguo Su, Rui Chen, Bingjie Wang, Teng Wang, Jiaojiao Tao, Qijie Diao, Tianze Jiang, Xia Zhao","doi":"10.1186/s12951-025-03389-9","DOIUrl":"https://doi.org/10.1186/s12951-025-03389-9","url":null,"abstract":"<p><p>Activation of cGAS-STING signaling pathway and accumulation of reactive oxygen and nitrogen species (RONS) are important issues facing the treatment of rheumatoid arthritis (RA). Here, we report a biomimetic nano-Chinese medicine (HA-RM-Cel-BR) for RA immunotherapy based on STING inhibition of celastrol (Cel) and RONS clearance of bilirubin (BR). HA-RM-Cel-BR is constructed by the carrier-free self-assembly of active ingredients Cel and BR from traditional Chinese medicine, and then camouflaged by hyaluronic acid (HA)-modified red blood cell membranes (RM). HA-RM-Cel-BR prolongs circulation time through RM camouflage, targets inflamed joints by HA modification, and remodels the joint immune microenvironment by STING inhibition and RONS clearance. More importantly, HA-RM-Cel-BR shows excellent therapeutic effects on RA rat model, and significantly reduces hepatotoxicity associated with Cel. Our work provides a new strategy for RA immunotherapy with traditional Chinese medicine ingredients.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"318"},"PeriodicalIF":10.6,"publicationDate":"2025-04-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12032812/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144012164","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}
Xuan Jiang, Jun Wang, Lin Gan, Zengshuai Wu, Tong Wu, Fengyang Li, Xiaowei Xu, Liancheng Lei, Na Li
{"title":"Carbon dot-based treatment for bacterial pneumonia by promoting a PI3K-mediated M1 polarization of macrophages.","authors":"Xuan Jiang, Jun Wang, Lin Gan, Zengshuai Wu, Tong Wu, Fengyang Li, Xiaowei Xu, Liancheng Lei, Na Li","doi":"10.1186/s12951-025-03399-7","DOIUrl":"https://doi.org/10.1186/s12951-025-03399-7","url":null,"abstract":"<p><p>As the incessant emergence of drug-resistant bacterial strains, bacterial pneumonia poses a serious threat to the public health worldwide. There is an urgent need to explore unconventional therapeutic strategies. Carbon dots are usually designed to directly kill bacteria, however, carbon dots that enhance the anti-infection function of immune cells are rarely reported. In the present study, CDots were synthesized with ascorbic acid and polyethyleneimine, exhibiting outstanding biocompatibility. Functionally, the CDots exhibited a well therapeutic impact on bacterial pneumonia induced by gram-positive multidrug-resistant Staphylococcus aureus (MRSA) or gram-negative Klebsiella pneumoniae (K. pneumoniae) in mice. Utilizing in vitro models of macrophages infected with MRSA and K. pneumoniae, we discovered that CDots augmented the M1 polarization of macrophages, subsequently enhancing their survival and activity of phagocytosis and bactericidal. Further investigations through molecular dynamics simulations and in vitro experiments validated that CDots directly bind to the catalytic subunit (PIK3CD) of phosphoinositide 3-kinase (PI3K), resulting in the inhibition of the PI3K/AKT/mTOR signaling pathway. Moreover, the crucial domain for the binding was located in amino acids 752-787 of PIK3CD. In summary, CDots exerted a protective effect on bacterial pneumonia by targeting the PIK3CD and fostering the PI3K-mediated M1 polarization of macrophages. These findings not only reveal a new role of CDots in the treatment of bacterial pneumonia, but also provide potential targets for future treatment strategies.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"315"},"PeriodicalIF":10.6,"publicationDate":"2025-04-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12032645/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144024289","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}
Long Liu, Siying Zhang, Yuqing Ren, Ruizhi Wang, Yuyuan Zhang, Siyuan Weng, Zhaokai Zhou, Peng Luo, Quan Cheng, Hui Xu, Yuhao Ba, Anning Zuo, Shutong Liu, Zaoqu Liu, Xinwei Han
{"title":"Macrophage-derived exosomes in cancer: a double-edged sword with therapeutic potential.","authors":"Long Liu, Siying Zhang, Yuqing Ren, Ruizhi Wang, Yuyuan Zhang, Siyuan Weng, Zhaokai Zhou, Peng Luo, Quan Cheng, Hui Xu, Yuhao Ba, Anning Zuo, Shutong Liu, Zaoqu Liu, Xinwei Han","doi":"10.1186/s12951-025-03321-1","DOIUrl":"https://doi.org/10.1186/s12951-025-03321-1","url":null,"abstract":"<p><p>Solid cancer contains a complicated communication network between cancer cells and components in the tumor microenvironment (TME), significantly influencing the progression of cancer. Exosomes function as key carriers of signaling molecules in these communications, including the intricate signalings of tumor-associated macrophages (TAMs) on cancer cells and the TME. With their natural lipid bilayer structures and biological activity that relates to their original cell, exosomes have emerged as efficient carriers in studies on cancer therapy. Intrigued by the heterogeneity and plasticity of both macrophages and exosomes, we regard macrophage-derived exosomes in cancer as a double-edged sword. For instance, TAM-derived exosomes, educated by the TME, can promote resistance to cancer therapies, while macrophage-derived exosomes generated in vitro have shown favorable potential in cancer therapy. Here, we depict the reasons for the heterogeneity of TAM-derived exosomes, as well as the manifold roles of TAM-derived exosomes in cancer progression, metastasis, and resistance to cancer therapy. In particular, we emphasize the recent advancements of modified macrophage-derived exosomes in diverse cancer therapies, arguing that these modified exosomes are endowed with unique advantages by their macrophage origin. We outline the challenges in translating these scientific discoveries into clinical cancer therapy, aiming to provide patients with safe and effective treatments.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"319"},"PeriodicalIF":10.6,"publicationDate":"2025-04-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12034189/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"143986948","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}