Zhi-Qi Dai, Sha Lu, Zhen-Tong Shen, Qing-Nan Li, Gui-Mei Han, Jin-Ming Liu, Yan Huang, Hao Zheng, Yi Zhang, Guo Chen, Quan Chen, Yun-Xi Cui, Li-Na Zhu, De-Ming Kong
{"title":"Intracellular In Situ Assembled DNA Networks Targeting Mitochondria Enable Selective Elimination of Senescent Cells and Improve Cell Viability.","authors":"Zhi-Qi Dai, Sha Lu, Zhen-Tong Shen, Qing-Nan Li, Gui-Mei Han, Jin-Ming Liu, Yan Huang, Hao Zheng, Yi Zhang, Guo Chen, Quan Chen, Yun-Xi Cui, Li-Na Zhu, De-Ming Kong","doi":"10.1002/adhm.202501030","DOIUrl":"https://doi.org/10.1002/adhm.202501030","url":null,"abstract":"<p><p>Mitochondria play crucial roles in energy production, metabolism regulation, and cell death. Mitochondrial dysfunction is associated with many diseases, including cancers, aging, and neurodegenerative disorders. Consequently, developing methods for mitochondrial regulation and treating related diseases has garnered significant interest in biological and medical research. Here, a smart framework nucleic acid (FNA) strategy is presented for mitochondrial interference and targeted cell elimination. Our approach involves the design of tetrahedral DNA nanostructures (TDNs) modified with triphenylphosphine and single-stranded DNA sequences responding to specific nucleic acid biomarkers (e.g., microRNAs) presented in target cells. The interlinked DNA networks, formed in situ responding to specific biomarkers, enable targeting and enveloping of the mitochondria, leading to mitochondrial fragmentation and dysfunction. It is demonstrated that TDN-based FNAs targeted the cancer-associated microRNA (miR-21) may enhance the efficacy of cancer therapy by disrupting mitochondrial function, while also serving as carriers of anti-cancer drugs to reduce the side effects. Additionally, FNAs targeting the senescence-associated microRNA (miR-34a) specifically eliminate senescent cells in both cell and Caenorhabditis elegans models, thereby improving overall cell viability within mixed cell populations. This programmable and functionalized TDN-based platform opens new avenues for advancing anti-aging research and treating various diseases by achieving targeted cell elimination through mitochondrial interference.</p>","PeriodicalId":113,"journal":{"name":"Advanced Healthcare Materials","volume":" ","pages":"e2501030"},"PeriodicalIF":10.0,"publicationDate":"2025-05-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144109055","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Hanyue Kang, Lei Qiu, Yecheng Li, Xiaocheng Xu, Renjun Pei, Tongqing Yang, Lizhi Yang, Xiaobin Xu, Na Sun
{"title":"Si Microanemones Integrated Microfluidic Chip for Highly Efficient Isolation of Extracellular Vesicles.","authors":"Hanyue Kang, Lei Qiu, Yecheng Li, Xiaocheng Xu, Renjun Pei, Tongqing Yang, Lizhi Yang, Xiaobin Xu, Na Sun","doi":"10.1002/adhm.202500439","DOIUrl":"https://doi.org/10.1002/adhm.202500439","url":null,"abstract":"<p><p>Liquid biopsy has emerged as a transformative approach for early cancer detection and treatment monitoring, offering significant potential to improve patient outcomes. However, isolating tumor-derived extracellular vesicles (EVs) from body fluids is often impeded by background noise, making subsequent analysis challenging. Herein, a bio-inspired 3D silicon microanemone (SMA) microfluidic chip is reported. This innovative structure is prepared by a two-step lithographic method combined with nanosphere lithography, achieving an impressive isolation efficiency of 89.4%. Simulation results reveal that the hierarchical structure not only provides more antibody binding sites but also synergizes with an integrated chaotic mixer to amplify fluid perturbations, while inducing a flow around circular cylinder phenomenon to enhance EV-antibody interactions. Finally, the SMA chip's performance is assessed with clinical samples and combined with RT-qPCR-based β-actin (ACTB) mRNA quantification in purified EVs. The results demonstrate its high sensitivity and specificity in isolating cancer-related EV subgroups, enabling non-invasive and precise detection of cancer biomarkers in blood samples.</p>","PeriodicalId":113,"journal":{"name":"Advanced Healthcare Materials","volume":" ","pages":"e2500439"},"PeriodicalIF":10.0,"publicationDate":"2025-05-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144109088","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Minji Ahn, Yuhyun Na, Hani Choi, Sanghee Lee, Jangsu Lee, Soon A Park, Sin-Soo Jeun, Kun Na
{"title":"Photoimmuno-Lure Nanoplatform for Enhancing T Cell Expansion in Glioblastoma via Synergistic Treatment of Photodynamic Therapy and Immune Checkpoint Inhibition.","authors":"Minji Ahn, Yuhyun Na, Hani Choi, Sanghee Lee, Jangsu Lee, Soon A Park, Sin-Soo Jeun, Kun Na","doi":"10.1002/adhm.202500880","DOIUrl":"https://doi.org/10.1002/adhm.202500880","url":null,"abstract":"<p><p>The immunosuppressive tumor microenvironment (TME) of glioblastoma (GBM) limits the efficacy of immune checkpoint inhibitors (ICI), primarily due to the absence of cytotoxic T (T<sub>c</sub>) cells. In this study, a photoimmuno-lure nanoplatform is presented that combines amphiphilic photosensitizers (PSs) with Atezolizumab leading to the modulation of the TME of GBM and improvement of the therapeutic efficacy through synergistic photodynamic therapy (PDT). The amphiphilic PSs exhibited four-fold higher GBM specificity, superior photostability, and enhanced singlet oxygen generation efficiency (<sup>1</sup>O<sub>2</sub>Φ<sub>Δ</sub>: 0.92) compared to conventional PSs. In in vitro GBM cell lines, amphiphilic PSs increased immune activation cytokines and improved ICI responsiveness compared to single ICI treatment. In addition, similar results are acquired in a GBM 3D spheroid model, showing significantly elevated T<sub>c</sub> cell activation. In orthotopic in vivo GBM model, the nanoplatform achieved a 100% survival rate for up to 60 days. Immunological analysis revealed each 2.36-fold, 4.19-fold increase in activated dendritic cells and T<sub>c</sub> cells respectively, and significant reductions in MDSCs (0.48-fold) and regulatory T cells (0.5-fold). As a result, this study demonstrates the potential of the synergistic photoimmuno-lure nanoplatform as a clinical solution to overcome the immunosuppressive TME of GBM and activate innate and adaptive immunity for effective treatment.</p>","PeriodicalId":113,"journal":{"name":"Advanced Healthcare Materials","volume":" ","pages":"e2500880"},"PeriodicalIF":10.0,"publicationDate":"2025-05-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144109066","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yuan Cao, Zixuan Zhao, Junyue Fang, Yanan Lu, Zhuoshan Huang, Guo Wu, Qiyuan Gao, Rong Li, Lei Xu, Xiaoding Xu
{"title":"Dual-Responsive Immunomodulatory RNAi Nanoplatform for Effective Immune Checkpoint Blockade and Enhanced Cancer Immunotherapy.","authors":"Yuan Cao, Zixuan Zhao, Junyue Fang, Yanan Lu, Zhuoshan Huang, Guo Wu, Qiyuan Gao, Rong Li, Lei Xu, Xiaoding Xu","doi":"10.1002/adhm.202500646","DOIUrl":"https://doi.org/10.1002/adhm.202500646","url":null,"abstract":"<p><p>Immune checkpoint blockade (ICB) therapy has become the first-line treatment for cancer patients. However, the low response rate remains a clinical pain-point. Anti-hyperglycemic drug metformin has shown remarkable anticancer effect with the unique characteristic of modulating tumor immune microenvironment (TIME). Therefore, combining ICB with metformin could be a promising strategy for enhanced cancer immunotherapy, which however remains challenged due to the low bioavailability and severe adverse effects of metformin. This work herein designs an amphiphilic reduction-responsive metformin prodrug, which could complex small interfering RNA (siRNA) and then co-assemble with an endosomal pH-responsive PEGylated polymer to form a dual-responsive immunomodulatory RNAi nanoplatform. Using the orthotopic and metastatic breast cancer (BCa) tumor models, this work demonstrates that this RNAi nanoplatform could silence PD-L1 expression on BCa cells and suppress their proliferation via activating AMP-activated protein kinase (AMPK). Moreover, this AMPK activation could suppress the secretion of tumor-derived transforming growth factor β (TGF-β) and interleukin 6 (IL-6), which could enhance the maturation of dendritic cells (DCs) and activation of CD8<sup>+</sup> T cells and impair the tumor infiltration of regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs), and tumor-associated macrophages (TAMs), ultimately achieving the goal of enhanced cancer immunotherapy and significant inhibition of BCa tumor growth.</p>","PeriodicalId":113,"journal":{"name":"Advanced Healthcare Materials","volume":" ","pages":"e2500646"},"PeriodicalIF":10.0,"publicationDate":"2025-05-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144109052","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yue Xu, Rui Huang, Wodong Shi, Rong Zhou, Xinling Xie, Miao Wang, Yang Wang, Ping Gu, Ni Ni, Xiaoping Bi
{"title":"ROS-Responsive Hydrogel Delivering METRNL Enhances Bone Regeneration via Dual Stem Cell Homing and Vasculogenesis Activation.","authors":"Yue Xu, Rui Huang, Wodong Shi, Rong Zhou, Xinling Xie, Miao Wang, Yang Wang, Ping Gu, Ni Ni, Xiaoping Bi","doi":"10.1002/adhm.202500060","DOIUrl":"https://doi.org/10.1002/adhm.202500060","url":null,"abstract":"<p><p>Critical-sized bone defects arising from bone-related diseases pose a clinical challenge, exceeding the body's natural healing capacity. Evidence has shown that a disordered microenvironment characterized by reactive oxygen species (ROS) overproduction, vascular damage, and osteoblast deficiency severely hinders bone repair. Therefore, the reconstruction of microenvironmental homeostasis post-injury is of utmost importance. Herein, a ROS-responsive scavenging GelMA loaded with METRNL (RRG-MRL) is developed, serving as a \"bone microenvironment-modulating system\" for targeted delivery of METRNL, which stimulates bone marrow mesenchymal stem cells (BMSCs) homing and angiogenic sprouting. Upon exposure to elevated levels of ROS within the defect region, ROS-cleavable NHS-TK-NHS linkers are disrupted, triggering responsive degradation and METRNL release. This treatment significantly reduced ROS levels and alleviated inflammation, along with increasing the levels of anti-apoptotic factors. Meanwhile, released METRNL induced endothelial cell angiogenesis by activating the c-Kit/PI3K/Akt pathway and increased secretion of SDF-1α (CXCL12) to promote BMSCs recruitment. Rat models of cranial bone defects treated with RRG-MRL demonstrated reduced ROS signal intensity in situ, increased endogenous BMSCs count, and enhanced neovascularization, resulting in accelerated bone regeneration. The proposed platform offers a multistage therapeutic approach facilitating rapid reconstruction of microenvironment homeostasis to promote bone regeneration, indicating significant clinical potential.</p>","PeriodicalId":113,"journal":{"name":"Advanced Healthcare Materials","volume":" ","pages":"e2500060"},"PeriodicalIF":10.0,"publicationDate":"2025-05-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144109086","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"NIR-II-Responsive Chainmail Nanocatalysts for Spatiotemporally Controlled Enzymatic Tumor Therapy.","authors":"Hongji Liu, Zhenxing Wang, Xinyue Hu, Shanshan Rao, Hui Wang, Hui Xie","doi":"10.1002/adhm.202501111","DOIUrl":"https://doi.org/10.1002/adhm.202501111","url":null,"abstract":"<p><p>The clinical translation of metal-based peroxidase-like nanozymes for antitumor therapy faces two critical challenges: off-target catalytic activation and suboptimal hydroxyl radical (•OH) generation efficiency. To address these limitations, an innovative chainmail nanocatalyst featuring nitrogen-doped carbon-encapsulated nanoceria is developed, which combines spatial confinement effects with photo-trigger catalytic enhancement. The graphitic carbon shell serves as a physical barrier that effectively isolates metallic cerium from the biological environment, reducing nonspecific catalytic activation by 100% compared to bare nanoceria. Remarkably, under 1064 nm laser irradiation, electrons of cerium species can penetrate the carbon confinement through quantum tunneling effects, activating multiple enzymatic pathways. Vacancy engineering further optimizes the Ce<sup>3+</sup>/Ce<sup>4+</sup> redox pair ratio (1.75 vs 0.44 in pristine nanoceria), establishing an electron reservoir that facilitates catalytic amplification of H<sub>2</sub>O<sub>2</sub>-to-•OH conversion and glutathione oxidase-mimicking activity for tumor microenvironment remodeling. This dual mechanism synergistically elevates intracellular oxidative stress while preserving normal tissue viability. In vivo evaluations demonstrate that the photoactivated nanocatalyst exhibits remarkable tumor suppression efficacy, prolonging the survival duration of tumor-bearing mice from 33 days to 70 days. The light-gated chainmail architecture provides a paradigm for spatiotemporally controlled catalytic therapy, resolving the critical dilemma between catalytic potency and biological specificity in nanozyme design.</p>","PeriodicalId":113,"journal":{"name":"Advanced Healthcare Materials","volume":" ","pages":"e2501111"},"PeriodicalIF":10.0,"publicationDate":"2025-05-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144109063","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Perumal Ramesh Kannan, Liuting Chen, Yudie Lv, Ruibo Zhao, Yeting Hu, M Zubair Iqbal, Qianqian Han, Xiangdong Kong, Yao Li
{"title":"Smart Silk-Based In Situ Sol-Gel Modulates Rectal Microenvironment for Effective Ulcerative Colitis Alleviation.","authors":"Perumal Ramesh Kannan, Liuting Chen, Yudie Lv, Ruibo Zhao, Yeting Hu, M Zubair Iqbal, Qianqian Han, Xiangdong Kong, Yao Li","doi":"10.1002/adhm.202500984","DOIUrl":"https://doi.org/10.1002/adhm.202500984","url":null,"abstract":"<p><p>Ulcerative colitis (UC) is a chronic inflammatory bowel disease, with untreated cases often progressing to colorectal cancer. Current treatments aim to induce inflammatory remission but often neglect the surrounding microenvironment, which significantly impairs mucosal healing and contributes to treatment failures. This study presents a novel silk fibroin-based fucoidan (SFU) in situ rectal gel, with sol-gel transition confirmed through rheological analysis under physiological pH and temperature conditions. The SFU gel exhibits strong antioxidant activity, achieving a DPPH radical scavenging rate of 73.3 ± 1.52%. The gel efficiently reduces reactive oxygen species (ROS) and nitric oxide (NO) production, demonstrating its reliable antioxidant effects. In a DSS-induced UC mouse model, SFU effectively alleviates colitis symptoms, including weight loss and disease activity index (DAI) reduction, with improved stool consistency and reduced rectal bleeding. Moreover, SFU therapy reprograms macrophages from proinflammatory M1 to anti-inflammatory M2 phenotypes, significantly lowering IL-6 and TNF-α levels, suggesting anti-inflammatory properties. Furthermore, SFU increased tight junction proteins Occludin-1 and ZO-1, indicating gut mucosal barrier integrity. SFU treatment restores goblet cells and mucin production while preventing fibrosis, demonstrating its potential as a natural therapy for UC treatment.</p>","PeriodicalId":113,"journal":{"name":"Advanced Healthcare Materials","volume":" ","pages":"e2500984"},"PeriodicalIF":10.0,"publicationDate":"2025-05-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144109102","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yufei Du, Yiyin Mai, Zhiwen Liu, Guanghui Lin, Siweier Luo, Chipeng Guo, Ge Qiao, Le Wang, Shuang Zhu, Yiming Zhou, Yue Pan
{"title":"Synergistic Provoking of Pyroptosis and STING Pathway by Multifunctional Manganese-Polydopamine Nano-Immunomodulator for Enhanced Renal Cell Carcinoma Immunotherapy.","authors":"Yufei Du, Yiyin Mai, Zhiwen Liu, Guanghui Lin, Siweier Luo, Chipeng Guo, Ge Qiao, Le Wang, Shuang Zhu, Yiming Zhou, Yue Pan","doi":"10.1002/adhm.202500141","DOIUrl":"https://doi.org/10.1002/adhm.202500141","url":null,"abstract":"<p><p>Manganese ions are known to enhance anti-tumor immunity by activating the cGAS-STING signaling pathway. However, precise modulation of the tumor microenvironment using manganese ions remains a challenge. Dopamine, with its controlled release properties within the tumor microenvironment, offers significant potential for precision drug delivery systems. Metastatic renal cell carcinoma (RCC), being refractory to conventional treatments, necessitates innovative therapeutic approaches. In this study, a multifunctional manganese-polydopamine nano-immunomodulator coated with hyaluronic acid (PDA-Mn-HA NPs) is developed. These nanoparticles selectively bind to CD44 molecules, which are highly expressed in tumor-associated macrophages and RCC cells, and release manganese ions in a tumor microenvironment-responsive manner. Treatment with PDA-Mn-HA NPs effectively induces macrophage M1 polarization, triggers the production of pro-inflammatory cytokines and chemokines. Transcriptomic analysis reveals that PDA-Mn-HA NPs polarize and activate macrophages through the reactive oxygen species(ROS)-STING-p38/MAPK signaling pathway. Additionally, PDA-Mn-HA NPs induce ROS-caspase-3/GSDME-dependent pyroptosis in RCC cells via a Fenton-like reaction. In RCC mouse models, PDA-Mn-HA NPs remodel the macrophage-mediated immune microenvironment, enhance immune cell infiltration, and significantly suppress tumor growth. In conclusion, multifunctional PDA-Mn-HA NPs demonstrate translational potential by addressing the limitations of precision manganese delivery and achieving synergistic targeting of macrophages and tumor cells, offering a promising therapeutic strategy for RCC.</p>","PeriodicalId":113,"journal":{"name":"Advanced Healthcare Materials","volume":" ","pages":"e2500141"},"PeriodicalIF":10.0,"publicationDate":"2025-05-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144109106","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"A 3D Co-Culture System Inspired by Fracture Healing Cell Interactions for Bone Tissue Engineering.","authors":"Jicenyuan Wu, Yuxuan Wang, Liang Wang, Wenjia Xie, Qianbing Wan, Jian Wang, Junyu Chen, Xibo Pei, Zhou Zhu","doi":"10.1002/adhm.202500534","DOIUrl":"https://doi.org/10.1002/adhm.202500534","url":null,"abstract":"<p><p>Peri-bone fibroblasts play a crucial role in regulating bone regeneration during early fracture healing. Inspired by the synergy between osteoblasts and fibroblasts at fracture sites, a biomimetic three-dimensional (3D) indirect co-culture system is developed, comprising a 3D scaffold and co-cultured cells. To mimic cellular interactions in the fracture healing zone, the scaffold features an inner-outer ring structure with communication channels that support indirect cell co-culture. This setup provides fibroblasts and osteoblasts with a 3D culture environment resembling the in vivo extracellular matrix, enhancing intercellular signaling while minimizing risks of direct contact. Mechanically tunable bioinks are formulated by incorporating hyaluronic acid methacrylate (HAMA) hydrogel into gelatin methacryloyl (GelMA) hydrogel to construct the scaffold. The optimal co-culture ratio is established in vitro, where fibroblasts are found to regulate the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) via zinc ion transport mechanisms. In vivo validations are conducted, including ectopic bone formation in nude mice and bone regeneration in rat cranial defect and tooth extraction socket models. This 3D indirect co-culture system enhances osteogenesis by promoting functional fibroblast-osteoblast interactions, offering a novel platform for co-culture studies and a promising strategy for clinical bone regeneration applications.</p>","PeriodicalId":113,"journal":{"name":"Advanced Healthcare Materials","volume":" ","pages":"e2500534"},"PeriodicalIF":10.0,"publicationDate":"2025-05-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144109188","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"A Polysaccharide-Based Self-Gelling Powder With Antibacterial and Antioxidant Capacities for Acute Hemostasis and Efficient Infected Wound Healing.","authors":"Wei Li, Jiahao Yang, Weishi Kong, Peng Fan, Dingding Guan, Yulu Bao, Guosheng Wu, Shige Wang, Yu Sun","doi":"10.1002/adhm.202501101","DOIUrl":"https://doi.org/10.1002/adhm.202501101","url":null,"abstract":"<p><p>Traumatic wounds with severe bleeding and heavy contamination present significant challenges, including delayed healing and high infection rates. An ideal wound dressing should achieve rapid hemostasis, provide antibacterial activity, and regulate the microenvironment to facilitate tissue regeneration. To address these needs, this work develops a novel selfgelling powder (COT) comprising carboxymethyl chitosan (CMCS), oxidized konjac glucomannan (OKGM), and tannic acid (TA). The COT powder exhibits rapid gelation upon contact with water, forming a COT hydrogel, making it suitable for controlling hemorrhage across diverse wound sites. In vitro and in vivo studies confirm that the COT possesses broad-spectrum antibacterial activity and potent free radical scavenging capacity. When applied to infected wounds, the COT hydrogel significantly reduces oxidative stress and inflammatory responses while promoting angiogenesis and re-epithelialization. Transcriptomic analysis suggests that COT modulates wound inflammation, likely by suppressing the NF-κB signaling pathway. Given its favorable mechanical properties, strong antibacterial/antioxidant effects, ease of application, and rapid hemostatic performance, the COT emerges as a promising translational wound dressing for managing infected wounds.</p>","PeriodicalId":113,"journal":{"name":"Advanced Healthcare Materials","volume":" ","pages":"e2501101"},"PeriodicalIF":10.0,"publicationDate":"2025-05-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144109190","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}