Pub Date : 2026-01-02DOI: 10.1016/j.biomaterials.2025.123966
Ruideng Wang , Xi He , Jinwu Bai , Shilong Su , Haifeng Liu , Fang Zhou
Osteoporotic bone defects repair remians significant clinical challenges, characterized by impaired osteogenic differentiation and dysregulated angiogenesis within a microenvironment of oxidative stress and chronic inflammation. Modulating this pathological microenvironment to create favorable conditions is a pivotal strategy for treating osteoporotic bone defects. In this study, we developed a multifunctional composite hydrogel (SMm) through photo-crosslinking, incorporating Metformin (Met) and magnesium oxide nanoparticles (MgO), to promote osteoporotic bone regeneration. The SMm hydrogel demonstrated optimal physicochemical characteristics and excellent biocompatibility in vitro. Through controlled release of Mg2+ ions and Met, the SMm hydrogel exhibited dual bioactivity: (1) robust osteogenic and angiogenic capacity, as evidenced by upregulation of key markers like Col 1, Runx-2 and VEGF, and (2) potent antioxidant and anti-inflammatory effects, effectively scavenging reactive oxygen species (ROS) and suppressing pro-inflammatory cytokines. Notably, under simulated osteoporotic conditions, SMm hydrogel significantly enhanced the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs), overcoming the inhibitory microenvironment. In vivo validation using an osteoporotic bone defect model further confirmed the therapeutic efficacy of SMm hydrogel, with micro-CT scans revealing increased new bone formation in the defect area. In summary, this study establishes SMm hydrogel as a promising biomaterial for osteoporotic bone regeneration, leveraging synergistic ion therapy and metabolic modulation to address pathological barriers.
{"title":"Microenvironment-engineered hydrogels drive osteo-angiogenic coupling to accelerate osteoporotic bone regeneration","authors":"Ruideng Wang , Xi He , Jinwu Bai , Shilong Su , Haifeng Liu , Fang Zhou","doi":"10.1016/j.biomaterials.2025.123966","DOIUrl":"10.1016/j.biomaterials.2025.123966","url":null,"abstract":"<div><div>Osteoporotic bone defects repair remians significant clinical challenges, characterized by impaired osteogenic differentiation and dysregulated angiogenesis within a microenvironment of oxidative stress and chronic inflammation. Modulating this pathological microenvironment to create favorable conditions is a pivotal strategy for treating osteoporotic bone defects. In this study, we developed a multifunctional composite hydrogel (SMm) through photo-crosslinking, incorporating Metformin (Met) and magnesium oxide nanoparticles (MgO), to promote osteoporotic bone regeneration. The SMm hydrogel demonstrated optimal physicochemical characteristics and excellent biocompatibility in vitro. Through controlled release of Mg<sup>2+</sup> ions and Met, the SMm hydrogel exhibited dual bioactivity: (1) robust osteogenic and angiogenic capacity, as evidenced by upregulation of key markers like Col 1, Runx-2 and VEGF, and (2) potent antioxidant and anti-inflammatory effects, effectively scavenging reactive oxygen species (ROS) and suppressing pro-inflammatory cytokines. Notably, under simulated osteoporotic conditions, SMm hydrogel significantly enhanced the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs), overcoming the inhibitory microenvironment. In vivo validation using an osteoporotic bone defect model further confirmed the therapeutic efficacy of SMm hydrogel, with micro-CT scans revealing increased new bone formation in the defect area. In summary, this study establishes SMm hydrogel as a promising biomaterial for osteoporotic bone regeneration, leveraging synergistic ion therapy and metabolic modulation to address pathological barriers.</div></div>","PeriodicalId":254,"journal":{"name":"Biomaterials","volume":"329 ","pages":"Article 123966"},"PeriodicalIF":12.9,"publicationDate":"2026-01-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145922317","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}
Pub Date : 2026-01-01DOI: 10.1016/j.biomaterials.2025.123973
Yan Chen , Jie Wang , Quan Dai , Peng Chen , Haiqing Wang , Ruolan Li , Yanru Gu , Han Liu , Hongtao Xiao , Shugang Qin , Yuying Li , Hongwei Zhang , Qiuju Wang
Overcoming therapeutic resistance mediated by apoptosis evasion represents a critical challenge in cancer treatment. Pyroptosis, an inflammatory form of regulated cell death, provides a promising alternative approach capable of stimulating robust anti-tumor immunity and converting immunologically inert ("cold") tumors into immunologically active ("hot") microenvironments. Importantly, these cell death pathways are not isolated but are interconnected through dynamic molecular switches that determine cellular fate. This review systematically examines the intricate molecular crosstalk between cell death modalities, with particular emphasis on the regulatory roles of the caspase-3/GSDME and caspase-8/GSDMD axes. Current evidence demonstrates that caspase family members, primarily associated with apoptosis, can selectively cleave gasdermin proteins, facilitating a transition from non-inflammatory apoptotic signaling to inflammatory pyroptotic events. We further analyze how inflammasomes, post-translational modifications, and the STING–NF–κB pathway precisely regulate this transition. Through integrated bioinformatic analysis, we identified novel hub genes (e.g., PRKAR1A, PPP2CA, FOSL2) and microRNA networks at the apoptosis-pyroptosis interface, providing novel insights and potential therapeutic targets. Exploiting this 'death switch' offers a novel therapeutic framework through three principal strategies: (1) inducing pyroptosis to eliminate apoptosis-resistant cells, (2) utilizing pyroptosis-induced inflammation to enhance immune checkpoint inhibitor efficacy, and (3) developing targeted therapeutics that directly modulate these switch molecules. Although controlling pyroptosis-associated inflammation remains challenging, understanding and manipulating the apoptosis-to-pyroptosis transition provides an innovative approach to overcome drug resistance and develop more effective cancer treatments.
{"title":"Harnessing the death switch: Empowering cancer therapy by modulating the apoptosis-pyroptosis transition","authors":"Yan Chen , Jie Wang , Quan Dai , Peng Chen , Haiqing Wang , Ruolan Li , Yanru Gu , Han Liu , Hongtao Xiao , Shugang Qin , Yuying Li , Hongwei Zhang , Qiuju Wang","doi":"10.1016/j.biomaterials.2025.123973","DOIUrl":"10.1016/j.biomaterials.2025.123973","url":null,"abstract":"<div><div>Overcoming therapeutic resistance mediated by apoptosis evasion represents a critical challenge in cancer treatment. Pyroptosis, an inflammatory form of regulated cell death, provides a promising alternative approach capable of stimulating robust anti-tumor immunity and converting immunologically inert (\"cold\") tumors into immunologically active (\"hot\") microenvironments. Importantly, these cell death pathways are not isolated but are interconnected through dynamic molecular switches that determine cellular fate. This review systematically examines the intricate molecular crosstalk between cell death modalities, with particular emphasis on the regulatory roles of the caspase-3/GSDME and caspase-8/GSDMD axes. Current evidence demonstrates that caspase family members, primarily associated with apoptosis, can selectively cleave gasdermin proteins, facilitating a transition from non-inflammatory apoptotic signaling to inflammatory pyroptotic events. We further analyze how inflammasomes, post-translational modifications, and the STING–NF–κB pathway precisely regulate this transition. Through integrated bioinformatic analysis, we identified novel hub genes (e.g., PRKAR1A, PPP2CA, FOSL2) and microRNA networks at the apoptosis-pyroptosis interface, providing novel insights and potential therapeutic targets. Exploiting this 'death switch' offers a novel therapeutic framework through three principal strategies: (1) inducing pyroptosis to eliminate apoptosis-resistant cells, (2) utilizing pyroptosis-induced inflammation to enhance immune checkpoint inhibitor efficacy, and (3) developing targeted therapeutics that directly modulate these switch molecules. Although controlling pyroptosis-associated inflammation remains challenging, understanding and manipulating the apoptosis-to-pyroptosis transition provides an innovative approach to overcome drug resistance and develop more effective cancer treatments.</div></div>","PeriodicalId":254,"journal":{"name":"Biomaterials","volume":"329 ","pages":"Article 123973"},"PeriodicalIF":12.9,"publicationDate":"2026-01-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145905440","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}
Pub Date : 2025-12-31DOI: 10.1016/j.biomaterials.2025.123970
Rui Xue , Fengyu Wang , Beibei Zhang , Jin Wu , Ningnannan Zhang , Chunyang Sun
Targeting phosphorylated tau (p-tau) across the blood–brain barrier (BBB) represents a critical prerequisite for attenuating tau pathology and disease progression in Alzheimer's disease (AD) by alleviating oxidative stress and neuroinflammation. To address this challenge, we developed a novel carrier-free selenium-based nanoassembly stabilized by hydroxyl-rich fingolimod (FTY720), a sphingosine analogue. Following camouflaging with melanoma cell membranes and further functionalizing with T807, the resulting nanocomposite (FSMT) demonstrated robust capacity for BBB crossing and target p-tau both in vitro and in vivo. Additionally, FTY720 and nano-selenium exert remarkable antioxidant and anti-inflammatory effects by modulating the GSK-3β and NF-κB signaling pathways, respectively, thereby attenuating tau hyperphosphorylation and preventing neuronal cell death. In an okadaic acid-induced AD mouse model, the FSMT treatment not only significantly ameliorated oxidative stress and neuroinflammation, but also improved spatial learning and memory impairments. The reduction in abnormal tau aggregation following treatment was confirmed by PET-CT imaging. Overall, this p-tau-targeted biomimetic nanocomposite demonstrated excellent biocompatibility and therapeutic efficacy, presenting a translatable strategy for treating AD and other neurological disorders through analogous mechanisms.
{"title":"Carrier-free nanoassembly with dual antioxidant and anti-inflammatory activities camouflaged by melanoma cell membrane for tau-targeted therapy of Alzheimer's disease","authors":"Rui Xue , Fengyu Wang , Beibei Zhang , Jin Wu , Ningnannan Zhang , Chunyang Sun","doi":"10.1016/j.biomaterials.2025.123970","DOIUrl":"10.1016/j.biomaterials.2025.123970","url":null,"abstract":"<div><div>Targeting phosphorylated tau (p-tau) across the blood–brain barrier (BBB) represents a critical prerequisite for attenuating tau pathology and disease progression in Alzheimer's disease (AD) by alleviating oxidative stress and neuroinflammation. To address this challenge, we developed a novel carrier-free selenium-based nanoassembly stabilized by hydroxyl-rich fingolimod (FTY720), a sphingosine analogue. Following camouflaging with melanoma cell membranes and further functionalizing with T807, the resulting nanocomposite (FSMT) demonstrated robust capacity for BBB crossing and target p-tau both <em>in vitro</em> and <em>in vivo</em>. Additionally, FTY720 and nano-selenium exert remarkable antioxidant and anti-inflammatory effects by modulating the GSK-3β and NF-κB signaling pathways, respectively, thereby attenuating tau hyperphosphorylation and preventing neuronal cell death. In an okadaic acid-induced AD mouse model, the FSMT treatment not only significantly ameliorated oxidative stress and neuroinflammation, but also improved spatial learning and memory impairments. The reduction in abnormal tau aggregation following treatment was confirmed by PET-CT imaging. Overall, this p-tau-targeted biomimetic nanocomposite demonstrated excellent biocompatibility and therapeutic efficacy, presenting a translatable strategy for treating AD and other neurological disorders through analogous mechanisms.</div></div>","PeriodicalId":254,"journal":{"name":"Biomaterials","volume":"329 ","pages":"Article 123970"},"PeriodicalIF":12.9,"publicationDate":"2025-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145922316","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}
Pub Date : 2025-12-31DOI: 10.1016/j.biomaterials.2025.123972
Yinqi Chen , Jiafeng Wang , Yechun Wang , Zimeng Liu , Jiajia Ying , Xuefei Zhou , Tianhua Zhou , Haiping Jiang , Xiangrui Liu , Quan Zhou
Postoperative adhesions are frequent and challenging complication lacking effective prevention. Evidence indicates that innate immune responses triggered by surgical trauma critically influence whether normal tissue repair progresses to pathological adhesions, highlighting early inflammatory responders as key intervention targets. Here, we successfully transformed natural hyaluronic acid (HA) into a potent innate inflammation modulator through a one-step sulfoxide-conjugation strategy. The resultant sulfoxide-conjugated HA (SOHA) demonstrated near-complete prevention of adhesion formation across multiple clinically relevant animal models, including those for secondary injury, long-term abdominal adhesions, and pericardial–pleural adhesions. Mechanistic investigations reveal that, unlike traditional physical barriers that primarily inhibit the late-stage adhesion of activated fibroblasts, SOHA addresses postoperative adhesions at their inception by reprogramming early innate inflammatory responses. It shifts the neutrophil cell death mode from acute, pro-inflammatory NETosis to more regulated, immunologically silent apoptotic process. Furthermore, it selectively restrains the early accumulation of large peritoneal macrophages (LPMs) in damaged tissues, redirecting them towards a reparative M2-like phenotype through the efferocytic clearance of apoptotic neutrophils, thereby promoting their timely involvement during the resolution phase of inflammation. This dual regulation of innate immunity effectively interrupts the postoperative inflammatory cascade and subsequent fibrotic progression, thus effectively shifting the pathological tissue repair into physiological healing.
{"title":"An innate immunity-reprogramming hydrogel nips postoperative adhesions in the bud via transforming pathological healing into physiological recovery","authors":"Yinqi Chen , Jiafeng Wang , Yechun Wang , Zimeng Liu , Jiajia Ying , Xuefei Zhou , Tianhua Zhou , Haiping Jiang , Xiangrui Liu , Quan Zhou","doi":"10.1016/j.biomaterials.2025.123972","DOIUrl":"10.1016/j.biomaterials.2025.123972","url":null,"abstract":"<div><div>Postoperative adhesions are frequent and challenging complication lacking effective prevention. Evidence indicates that innate immune responses triggered by surgical trauma critically influence whether normal tissue repair progresses to pathological adhesions, highlighting early inflammatory responders as key intervention targets. Here, we successfully transformed natural hyaluronic acid (HA) into a potent innate inflammation modulator through a one-step sulfoxide-conjugation strategy. The resultant sulfoxide-conjugated HA (SOHA) demonstrated near-complete prevention of adhesion formation across multiple clinically relevant animal models, including those for secondary injury, long-term abdominal adhesions, and pericardial–pleural adhesions. Mechanistic investigations reveal that, unlike traditional physical barriers that primarily inhibit the late-stage adhesion of activated fibroblasts, SOHA addresses postoperative adhesions at their inception by reprogramming early innate inflammatory responses. It shifts the neutrophil cell death mode from acute, pro-inflammatory NETosis to more regulated, immunologically silent apoptotic process. Furthermore, it selectively restrains the early accumulation of large peritoneal macrophages (LPMs) in damaged tissues, redirecting them towards a reparative M2-like phenotype through the efferocytic clearance of apoptotic neutrophils, thereby promoting their timely involvement during the resolution phase of inflammation. This dual regulation of innate immunity effectively interrupts the postoperative inflammatory cascade and subsequent fibrotic progression, thus effectively shifting the pathological tissue repair into physiological healing.</div></div>","PeriodicalId":254,"journal":{"name":"Biomaterials","volume":"329 ","pages":"Article 123972"},"PeriodicalIF":12.9,"publicationDate":"2025-12-31","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145881568","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}
Pub Date : 2025-12-30DOI: 10.1016/j.biomaterials.2025.123951
Shengjie Ma , Zimu Yuan , Chenlong Yu , Bin Sun , Ruibo Tang , Ding Zhou , Shoujun Zhu , Quan Wang
Intraoperative ureter visualization is essential in colorectal cancer (CRC) surgery, but current modalities are limited by invasiveness, poor photostability, and insufficient spatial resolution. Second near-infrared (NIR-II) fluorescence imaging provides a non-invasive approach with improved tissue penetration and spatial resolution. By modulating the aggregation behavior of NIR-II dyes, intravenous administration followed by renal metabolism enables real-time, high-resolution ureteral visualization. In this study, we prepared a series of donor–acceptor–donor-based NIR-II probes through a one-pot process where dye synthesis/PEGylation and free PEG assembly occur concurrently. The IR-FEP1000 cluster absorbs/emits maximally at 784/1025 nm in PBS and exhibits high photobleaching resistance due to its unique structural rigidity. Importantly, the IR-FEP1000 cluster exhibited renal excretion and enabled superior ureteral imaging performance with excellent biocompatibility, high resolution, and a prolonged imaging window. Furthermore, the IR-FEP1000 cluster enabled non-invasive, effective imaging diagnosis in scenarios of accidental ureteral transection, ligation, and clamping. Moreover, the IR-FEP1000 cluster combined with CO-1080 and Er-NPs enables multiplexed NIR-II imaging-guided surgery. High-contrast three-channel intraoperative imaging of the CRC peritoneal metastases, the ureters, and the intestines was provided. Multiplex NIR-II imaging enables precise tumor resection while protecting vital organs from intraoperative injury. These findings highlight that the IR-FEP1000 cluster exhibits advantages in non-invasive, long-duration, and high-resolution ureteral imaging with broad prospects in complex surgical scenarios.
{"title":"Renal-clearable organic NIR-II dye cluster for non-invasive ureteral imaging","authors":"Shengjie Ma , Zimu Yuan , Chenlong Yu , Bin Sun , Ruibo Tang , Ding Zhou , Shoujun Zhu , Quan Wang","doi":"10.1016/j.biomaterials.2025.123951","DOIUrl":"10.1016/j.biomaterials.2025.123951","url":null,"abstract":"<div><div>Intraoperative ureter visualization is essential in colorectal cancer (CRC) surgery, but current modalities are limited by invasiveness, poor photostability, and insufficient spatial resolution. Second near-infrared (NIR-II) fluorescence imaging provides a non-invasive approach with improved tissue penetration and spatial resolution. By modulating the aggregation behavior of NIR-II dyes, intravenous administration followed by renal metabolism enables real-time, high-resolution ureteral visualization. In this study, we prepared a series of donor–acceptor–donor-based NIR-II probes through a one-pot process where dye synthesis/PEGylation and free PEG assembly occur concurrently. The IR-FEP<sub>1000</sub> cluster absorbs/emits maximally at 784/1025 nm in PBS and exhibits high photobleaching resistance due to its unique structural rigidity. Importantly, the IR-FEP<sub>1000</sub> cluster exhibited renal excretion and enabled superior ureteral imaging performance with excellent biocompatibility, high resolution, and a prolonged imaging window. Furthermore, the IR-FEP<sub>1000</sub> cluster enabled non-invasive, effective imaging diagnosis in scenarios of accidental ureteral transection, ligation, and clamping. Moreover, the IR-FEP<sub>1000</sub> cluster combined with CO-1080 and Er-NPs enables multiplexed NIR-II imaging-guided surgery. High-contrast three-channel intraoperative imaging of the CRC peritoneal metastases, the ureters, and the intestines was provided. Multiplex NIR-II imaging enables precise tumor resection while protecting vital organs from intraoperative injury. These findings highlight that the IR-FEP<sub>1000</sub> cluster exhibits advantages in non-invasive, long-duration, and high-resolution ureteral imaging with broad prospects in complex surgical scenarios.</div></div>","PeriodicalId":254,"journal":{"name":"Biomaterials","volume":"329 ","pages":"Article 123951"},"PeriodicalIF":12.9,"publicationDate":"2025-12-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145922314","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}
Pub Date : 2025-12-29DOI: 10.1016/j.biomaterials.2025.123967
Jianing Yi , Yuanyu Tang , Yilin Chen , Liang Chen , Dongxue Geng , Luyao Liu , Jie Yu , Lianhong Zou , Jie Zeng , Minhuan Lan , Wenjie Gao , Ming Gao
Type 2 diabetes is a systemic disorder characterized by metabolic dysfunction and chronic inflammation, yet strategies that address both aspects remain limited. Here, we present kudzu root–derived carbon dots (KRCDs) as a natural nanomaterial that reprograms the gut microbiota-metabolite-immune axis to restore systemic homeostasis. KRCDs exhibit nanoscale crystallinity, abundant O/N functional groups, and strong antioxidant activity. In high-fat diet/streptozotocin-induced diabetic mice, KRCDs significantly lowered fasting glucose, improved glucose tolerance and insulin sensitivity, corrected lipid profiles, and reduced hepatic steatosis without detectable toxicity. Multi-omics analyses revealed increased microbial diversity, enrichment of beneficial genera such as Anaerostipes, and remodeling of fecal metabolites with a marked rise in indole-3-carboxaldehyde (I3A). This metabolite correlated with enhanced M2-like macrophage polarization across adipose tissue, intestine, kidney, liver, and pancreas, as confirmed by flow cytometry and immunofluorescence. Fecal microbiota transplantation from KRCDs-treated donors reproduced both the metabolic improvements and the organ-wide M2 polarization, confirming a microbiota-dependent mechanism. By establishing a gut microbiota–metabolite–macrophage polarization pathway, KRCDs act as safe, plant-based nanoplatforms that simultaneously correct metabolic and immune imbalance, offering a promising strategy for multi-target intervention in diabetes.
{"title":"Kudzu root-derived carbon dots modulate gut microbiota and metabolites for pan-organ targeted macrophage polarization in synergistic diabetes therapy","authors":"Jianing Yi , Yuanyu Tang , Yilin Chen , Liang Chen , Dongxue Geng , Luyao Liu , Jie Yu , Lianhong Zou , Jie Zeng , Minhuan Lan , Wenjie Gao , Ming Gao","doi":"10.1016/j.biomaterials.2025.123967","DOIUrl":"10.1016/j.biomaterials.2025.123967","url":null,"abstract":"<div><div>Type 2 diabetes is a systemic disorder characterized by metabolic dysfunction and chronic inflammation, yet strategies that address both aspects remain limited. Here, we present kudzu root–derived carbon dots (KRCDs) as a natural nanomaterial that reprograms the gut microbiota-metabolite-immune axis to restore systemic homeostasis. KRCDs exhibit nanoscale crystallinity, abundant O/N functional groups, and strong antioxidant activity. In high-fat diet/streptozotocin-induced diabetic mice, KRCDs significantly lowered fasting glucose, improved glucose tolerance and insulin sensitivity, corrected lipid profiles, and reduced hepatic steatosis without detectable toxicity. Multi-omics analyses revealed increased microbial diversity, enrichment of beneficial genera such as <em>Anaerostipes</em>, and remodeling of fecal metabolites with a marked rise in indole-3-carboxaldehyde (I3A). This metabolite correlated with enhanced M2-like macrophage polarization across adipose tissue, intestine, kidney, liver, and pancreas, as confirmed by flow cytometry and immunofluorescence. Fecal microbiota transplantation from KRCDs-treated donors reproduced both the metabolic improvements and the organ-wide M2 polarization, confirming a microbiota-dependent mechanism. By establishing a gut microbiota–metabolite–macrophage polarization pathway, KRCDs act as safe, plant-based nanoplatforms that simultaneously correct metabolic and immune imbalance, offering a promising strategy for multi-target intervention in diabetes.</div></div>","PeriodicalId":254,"journal":{"name":"Biomaterials","volume":"329 ","pages":"Article 123967"},"PeriodicalIF":12.9,"publicationDate":"2025-12-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145909424","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}
Oxidative stress and fibrosis are core drivers of post-myocardial infarction dysfunction. Conventional therapies predominantly address either acute oxidative damage or chronic fibrosis, lacking strategies that concurrently target the two interconnected processes. Here, a microenvironment-responsive nanomedicine delivering nitric oxide (NO) and curcumin specifically to the infarcted heart is presented (named C@Si-NM). Constructed from glutathione-sensitive cyclic nitrate-trimethylene carbonate-polyethylene glycol block copolymer micelles crosslinked with silica, the system releases both NO and the encapsulated curcumin upon encountering elevated glutathione levels at the infarction site. In cardiomyocytes, NO suppresses the opening of mitochondrial permeability transition pore, inhibiting the release of reactive oxygen species, while curcumin directly acts as an antioxidant. In fibroblasts, NO inhibits the TGF-β/Smad pathway and curcumin downregulates the production of TGF-β, jointly suppressing fibrosis. The combined nanomedicine C@Si-NM significantly improved cardiac function in a rat model of MI after four weeks of treatment, resulting in an ejection fraction of 74 %. This value approached the normal range of 81 % and was superior to the outcomes observed with NO monotherapy at 62 % or curcumin monotherapy at 63 %. This strategy effectively mitigates both oxidative damage and fibrosis, bridging the gap between acute and chronic therapeutic interventions of myocardial infarction.
{"title":"Bridging acute-chronic myocardial infarction treatment: Dual-regulating of ROS/fibrosis via microenvironment-responsive release of NO and curcumin","authors":"Fang Zhang , Bingbing Zhao , Keqiang Lu , Xueping Zeng , Zikun Wang , Wei Chen , Yunyun Zhou , Juanjuan Peng , Lingzhi Zhao","doi":"10.1016/j.biomaterials.2025.123954","DOIUrl":"10.1016/j.biomaterials.2025.123954","url":null,"abstract":"<div><div>Oxidative stress and fibrosis are core drivers of post-myocardial infarction dysfunction. Conventional therapies predominantly address either acute oxidative damage or chronic fibrosis, lacking strategies that concurrently target the two interconnected processes. Here, a microenvironment-responsive nanomedicine delivering nitric oxide (NO) and curcumin specifically to the infarcted heart is presented (named C@Si-NM). Constructed from glutathione-sensitive cyclic nitrate-trimethylene carbonate-polyethylene glycol block copolymer micelles crosslinked with silica, the system releases both NO and the encapsulated curcumin upon encountering elevated glutathione levels at the infarction site. In cardiomyocytes, NO suppresses the opening of mitochondrial permeability transition pore, inhibiting the release of reactive oxygen species, while curcumin directly acts as an antioxidant. In fibroblasts, NO inhibits the TGF-β/Smad pathway and curcumin downregulates the production of TGF-β, jointly suppressing fibrosis. The combined nanomedicine C@Si-NM significantly improved cardiac function in a rat model of MI after four weeks of treatment, resulting in an ejection fraction of 74 %. This value approached the normal range of 81 % and was superior to the outcomes observed with NO monotherapy at 62 % or curcumin monotherapy at 63 %. This strategy effectively mitigates both oxidative damage and fibrosis, bridging the gap between acute and chronic therapeutic interventions of myocardial infarction.</div></div>","PeriodicalId":254,"journal":{"name":"Biomaterials","volume":"329 ","pages":"Article 123954"},"PeriodicalIF":12.9,"publicationDate":"2025-12-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145881626","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}
Pub Date : 2025-12-29DOI: 10.1016/j.biomaterials.2025.123965
Hongjuan Weng , Monize C. Decarli , Wen Chen , Katrien V. Bernaerts , Lorenzo Moroni
Natural hydrogels (e.g., collagen hydrogels) show good potential in understanding cell-matrix interaction and find application in tissue engineering. However, it remains challenging to bioprint cell-laden natural hydrogels with good printability, shape retention and stability. In this study, non-water-soluble short collagen type I microfibers (COL-I μFiber) were blended with water-soluble methacrylated collagen peptide (COPMA) and xanthan gum (XG), forming an interpenetrated network, and bioprinted into stable natural-derived COPMA-μFiber-XG constructs, followed by in situ stem cell proliferation and differentiation. First, to enhance the printability and the mechanical properties of COPMA, a COPMA-μFiber-XG bioink was developed, featuring rapid UV-curing and self-healing properties. The encapsulated human mesenchymal stem cells (hMSCs) spread along the COL-I μFibers in the bioprinted constructs, with increased metabolic activity and production of extracellular matrix and bioactive proteins (COL-I and scleraxis) in 28 days. The internal biophysical and biochemical signals provided by COL-I μFibers and the fibrous COPMA matrix synergistically interacted with exogenous biochemical signals (e.g., transforming growth factor-beta 3) to further promote stem cell differentiation. Overall, bioprinted fibrous COPMA-μFiber-XG constructs are biocompatible and bioactive matrices to support hMSCs proliferation and differentiation.
{"title":"Engineering bioactive fibrous constructs: Bioprinting stem cell-laden collagen-derived hydrogels with short collagen microfibers","authors":"Hongjuan Weng , Monize C. Decarli , Wen Chen , Katrien V. Bernaerts , Lorenzo Moroni","doi":"10.1016/j.biomaterials.2025.123965","DOIUrl":"10.1016/j.biomaterials.2025.123965","url":null,"abstract":"<div><div>Natural hydrogels (e.g., collagen hydrogels) show good potential in understanding cell-matrix interaction and find application in tissue engineering. However, it remains challenging to bioprint cell-laden natural hydrogels with good printability, shape retention and stability. In this study, non-water-soluble short collagen type I microfibers (COL-I μFiber) were blended with water-soluble methacrylated collagen peptide (COPMA) and xanthan gum (XG), forming an interpenetrated network, and bioprinted into stable natural-derived COPMA-μFiber-XG constructs, followed by <em>in situ</em> stem cell proliferation and differentiation. First, to enhance the printability and the mechanical properties of COPMA, a COPMA-μFiber-XG bioink was developed, featuring rapid UV-curing and self-healing properties. The encapsulated human mesenchymal stem cells (hMSCs) spread along the COL-I μFibers in the bioprinted constructs, with increased metabolic activity and production of extracellular matrix and bioactive proteins (COL-I and scleraxis) in 28 days. The internal biophysical and biochemical signals provided by COL-I μFibers and the fibrous COPMA matrix synergistically interacted with exogenous biochemical signals (e.g., transforming growth factor-beta 3) to further promote stem cell differentiation. Overall, bioprinted fibrous COPMA-μFiber-XG constructs are biocompatible and bioactive matrices to support hMSCs proliferation and differentiation.</div></div>","PeriodicalId":254,"journal":{"name":"Biomaterials","volume":"329 ","pages":"Article 123965"},"PeriodicalIF":12.9,"publicationDate":"2025-12-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145964872","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}
Pub Date : 2025-12-27DOI: 10.1016/j.biomaterials.2025.123953
Bibi S. Subhan , Sydney E. Hanson , Dianny Almanzar , Juan F. Cortes Troncoso , Priya Katyal , Jonathan W. Sun , Hao-Wei Shih , Tamara Mestvirishvili , Michael Meleties , Fernando Arias , Andrew Wang , Kelly Ruggles , Igor Dolgalev , Paolo Mita , Jin Kim Montclare , Piul S. Rabbani
Chronic wounds, especially in diabetic patients, pose a significant clinical challenge due to impaired microvasculature and delayed healing. This study presents Exo-Q, a novel thermoresponsive hydrogel formed by co-gelation of engineered Q protein nanofibers with exosomes, a class of vesicular intercellular communication mediators. Exo-Q transitions from a gel to a viscoelastic solution at physiological temperature, enabling localized, topical delivery of exosomes with an initial burst release followed by sustained release. In a diabetic mouse wound model, Exo-Q effectively delivered human bone marrow multipotent stromal cell-derived exosomes directly to the wound bed, where they accumulated in endothelial cells of granulation tissue without detectable systemic distribution. Exosomes produced under stringent and replicable cell culture conditions consistently carried biomacromolecular cargo enriched for miRNAs with validated targets in angiogenesis-associated genes, indicative of their therapeutic potential. Topical application of Exo-Q resulted in extensive neovascularized granulation tissue, significantly accelerating wound closure to levels comparable to non-diabetic wounds. Importantly, the hydrogel’s modular design maintained the functional integrity of Q protein nanofibers and exosomes, demonstrating compatibility with full-thickness human wounds. This platform allows for tailored customization to address critical stages of diabetic wound healing while ensuring efficacy at low dosages, potentially enabling patient-administered treatment. By leveraging advanced biomaterials, Exo-Q advances the therapeutic efficacy of exosome-based interventions for diabetic wounds, offering a localized, non-invasive solution to chronic, non-healing wounds. This innovative hydrogel platform represents a modular therapeutic strategy with significant potential for clinical applications in regenerative medicine.
{"title":"Duo-nano exosome encapsulating hydrogel boosts wound healing across xenogenic and allogenic models","authors":"Bibi S. Subhan , Sydney E. Hanson , Dianny Almanzar , Juan F. Cortes Troncoso , Priya Katyal , Jonathan W. Sun , Hao-Wei Shih , Tamara Mestvirishvili , Michael Meleties , Fernando Arias , Andrew Wang , Kelly Ruggles , Igor Dolgalev , Paolo Mita , Jin Kim Montclare , Piul S. Rabbani","doi":"10.1016/j.biomaterials.2025.123953","DOIUrl":"10.1016/j.biomaterials.2025.123953","url":null,"abstract":"<div><div>Chronic wounds, especially in diabetic patients, pose a significant clinical challenge due to impaired microvasculature and delayed healing. This study presents Exo-Q, a novel thermoresponsive hydrogel formed by co-gelation of engineered Q protein nanofibers with exosomes, a class of vesicular intercellular communication mediators. Exo-Q transitions from a gel to a viscoelastic solution at physiological temperature, enabling localized, topical delivery of exosomes with an initial burst release followed by sustained release. In a diabetic mouse wound model, Exo-Q effectively delivered human bone marrow multipotent stromal cell-derived exosomes directly to the wound bed, where they accumulated in endothelial cells of granulation tissue without detectable systemic distribution. Exosomes produced under stringent and replicable cell culture conditions consistently carried biomacromolecular cargo enriched for miRNAs with validated targets in angiogenesis-associated genes, indicative of their therapeutic potential. Topical application of Exo-Q resulted in extensive neovascularized granulation tissue, significantly accelerating wound closure to levels comparable to non-diabetic wounds. Importantly, the hydrogel’s modular design maintained the functional integrity of Q protein nanofibers and exosomes, demonstrating compatibility with full-thickness human wounds. This platform allows for tailored customization to address critical stages of diabetic wound healing while ensuring efficacy at low dosages, potentially enabling patient-administered treatment. By leveraging advanced biomaterials, Exo-Q advances the therapeutic efficacy of exosome-based interventions for diabetic wounds, offering a localized, non-invasive solution to chronic, non-healing wounds. This innovative hydrogel platform represents a modular therapeutic strategy with significant potential for clinical applications in regenerative medicine.</div></div>","PeriodicalId":254,"journal":{"name":"Biomaterials","volume":"329 ","pages":"Article 123953"},"PeriodicalIF":12.9,"publicationDate":"2025-12-27","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145922313","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}
Pub Date : 2025-12-26DOI: 10.1016/j.biomaterials.2025.123952
Bing Chen , Hanye Xing , Xingyu Liu , Jinlong Hu , Liang Dong , Xu Yan , Yonghong Song , Yang Lu
Disintegrating hydrogels offer the advantages of synergistically enhancing therapeutic efficacy in locoregional percutaneous treatment for early hepatocellular carcinoma, enabling on-demand drug delivery with reduced side effects. However, current disintegration processes suffer from limited precise control and weak tissue penetration. Herein, we constructed an injectable ferrimagnetic hydrogen bonding cross-linked hydrogel (named as FPH) as a locoregional percutaneous agent by integrating ferrimagnetic nanoparticles into a polyvinyl alcohol (PVA) crosslinked network. This hydrogel enables remote magnetothermally triggered disintegration under an alternating magnetic field (AMF), with its disintegration temperature precisely tunable by adjusting the hydrogel's solid content. Under magnetic heating effect, FPH achieved remote “gel to disintegration” behavior at a desired temperature range of around 47 °C, effectively suppressing tumor cells and minimizing harm to normal tissues. By contrast, other commonly used hydrogen-bonded network of gelatin hydrogel disintegrated below body temperature or agarose hydrogel disintegrated over 70 °C. Benefiting from magnetothermal-controlled disintegration, drug-loaded FPH exhibited an increased release efficiency from ∼8 % (without AMF) to ∼45 % (with AMF) within 1 h. Following ultrasound-guided percutaneous delivery, FPHDOX exhibited synergistic efficacy with magnetic hyperthermia and disintegration-mediated chemotherapy in rabbit liver tumors. Additionally, FPH is fabricated using clinically approved pharmaceutical excipients, ensuring excellent biocompatibility. This strategy inspires the design of spatiotemporally controllable disintegrating hydrogels with limitless tissue penetration depth, and expands their potential in percutaneous hepatocellular carcinoma treatment.
{"title":"Spatiotemporal controlled disintegration enabling injected magnetic hydrogel for percutaneous hepatocellular carcinoma treatment","authors":"Bing Chen , Hanye Xing , Xingyu Liu , Jinlong Hu , Liang Dong , Xu Yan , Yonghong Song , Yang Lu","doi":"10.1016/j.biomaterials.2025.123952","DOIUrl":"10.1016/j.biomaterials.2025.123952","url":null,"abstract":"<div><div>Disintegrating hydrogels offer the advantages of synergistically enhancing therapeutic efficacy in locoregional percutaneous treatment for early hepatocellular carcinoma, enabling on-demand drug delivery with reduced side effects. However, current disintegration processes suffer from limited precise control and weak tissue penetration. Herein, we constructed an injectable ferrimagnetic hydrogen bonding cross-linked hydrogel (named as FPH) as a locoregional percutaneous agent by integrating ferrimagnetic nanoparticles into a polyvinyl alcohol (PVA) crosslinked network. This hydrogel enables remote magnetothermally triggered disintegration under an alternating magnetic field (AMF), with its disintegration temperature precisely tunable by adjusting the hydrogel's solid content. Under magnetic heating effect, FPH achieved remote “gel to disintegration” behavior at a desired temperature range of around 47 °C, effectively suppressing tumor cells and minimizing harm to normal tissues. By contrast, other commonly used hydrogen-bonded network of gelatin hydrogel disintegrated below body temperature or agarose hydrogel disintegrated over 70 °C. Benefiting from magnetothermal-controlled disintegration, drug-loaded FPH exhibited an increased release efficiency from ∼8 % (without AMF) to ∼45 % (with AMF) within 1 h. Following ultrasound-guided percutaneous delivery, FPH<sub>DOX</sub> exhibited synergistic efficacy with magnetic hyperthermia and disintegration-mediated chemotherapy in rabbit liver tumors. Additionally, FPH is fabricated using clinically approved pharmaceutical excipients, ensuring excellent biocompatibility. This strategy inspires the design of spatiotemporally controllable disintegrating hydrogels with limitless tissue penetration depth, and expands their potential in percutaneous hepatocellular carcinoma treatment.</div></div>","PeriodicalId":254,"journal":{"name":"Biomaterials","volume":"329 ","pages":"Article 123952"},"PeriodicalIF":12.9,"publicationDate":"2025-12-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145922318","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}