Qinghe Meng, David O. Popoola, Chunyan Wang, Yuqin Men, Yuqi Song, Zhi Cao, Adam Novak, Yamin Li and Robert N. Cooney
{"title":"Lung-targeting lipid nanoparticle-mediated sparstolonin B delivery improves acute lung injury","authors":"Qinghe Meng, David O. Popoola, Chunyan Wang, Yuqin Men, Yuqi Song, Zhi Cao, Adam Novak, Yamin Li and Robert N. Cooney","doi":"10.1039/D5BM00590F","DOIUrl":"10.1039/D5BM00590F","url":null,"abstract":"<p >Acute respiratory distress syndrome (ARDS), a severe manifestation of acute lung injury (ALI), is characterized by high morbidity and mortality, with limited therapeutic options. Sparstolonin B (SsnB), a selective antagonist of Toll-like receptors (TLR)-2 and TLR-4 with significant anti-inflammatory activity, has been studied in various diseases. However, its potential for targeted delivery to lung tissues in the treatment of ARDS/ALI remains an underexplored area warranting further investigation. Here, we report the development of a lung-targeting sulfonium lipid nanoparticle (sLNP)-mediated SsnB delivery system in a murine model of lipopolysaccharide (LPS)-induced ALI. Comprehensive analyses of serum, bronchoalveolar lavage fluid (BALF), and lung tissues post-injury revealed that SsnB-loaded sLNP (SsnB/sLNP) significantly mitigated lung injury. This was evidenced by improved lung histology, reduced macrophage counts and neutrophil infiltration in BALF, and decreased levels of pro-inflammatory cytokines, including TNF-α, IL-1β and IL-6, in both BALF and serum. Mechanistic studies further demonstrated that the therapeutic effects of SsnB were mediated through the inhibition of the NF-κB signaling pathway. These findings highlight the potential of lung-targeting sLNP-mediated SsnB delivery as a promising therapeutic strategy for ALI and ARDS.</p>","PeriodicalId":65,"journal":{"name":"Biomaterials Science","volume":" 19","pages":" 5429-5441"},"PeriodicalIF":5.7,"publicationDate":"2025-08-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144935998","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Qian He, Jiawen He, Panyue Wen, Haochen Guo, Masaru Tanaka, Shizhong Luo, Junjie Li, Ye Liu, Liewei Wen and Chenzhi Yao
{"title":"NIR-activatable aza-BODIPY nanoparticles for photoacoustic imaging and synergistic NO–photothermal cancer therapy","authors":"Qian He, Jiawen He, Panyue Wen, Haochen Guo, Masaru Tanaka, Shizhong Luo, Junjie Li, Ye Liu, Liewei Wen and Chenzhi Yao","doi":"10.1039/D5BM00937E","DOIUrl":"10.1039/D5BM00937E","url":null,"abstract":"<p >Near-infrared (NIR) photothermal therapy (PTT) has emerged as a promising modality for cancer treatment due to its minimal invasiveness, precise spatiotemporal control, and potent therapeutic outcomes. However, the clinical application of photothermal agents (PTAs) remains limited by issues such as poor biodegradability, long-term toxicity, and insufficient photothermal conversion efficiency. Herein, we report the development of a novel amphiphilic aza-boron-dipyrromethene (aza-BODIPY)-based photothermal agent, C<small><sub>8</sub></small>-NBDP-OEG<small><sub>4</sub></small>, which self-assembles into monodisperse nanoparticles in aqueous solution. These nanoparticles exhibit excellent chemical and photostability, along with a high photothermal conversion efficiency of 39.8% under 808 nm laser irradiation. To endow the system with multifunctionality, the nitric oxide (NO) donor <em>S</em>-nitroso-<em>N</em>-acetylpenicillamine (SNAP) was co-encapsulated within the nanoparticles, enabling NIR-triggered NO release. This design achieves a dual-mode therapeutic strategy, combining localized hyperthermia and NO-mediated modulation of the tumor microenvironment, thereby significantly enhancing anticancer efficacy. Importantly, the released NO was found to amplify the photoacoustic (PA) signal intensity, facilitating photoacoustic imaging-guided therapy. Both <em>in vitro</em> and <em>in vivo</em> studies demonstrated pronounced tumor growth inhibition with minimal systemic toxicity. Collectively, our study introduces C<small><sub>8</sub></small>-NBDP-OEG<small><sub>4</sub></small>@NO nanoparticles as a multifunctional theranostic nanoplatform, offering NIR-activated, PA imaging-guided synergistic NO–photothermal therapy and showcasing strong potential for precise and effective cancer treatment.</p>","PeriodicalId":65,"journal":{"name":"Biomaterials Science","volume":" 19","pages":" 5369-5381"},"PeriodicalIF":5.7,"publicationDate":"2025-08-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144936275","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Oscar R. Benavides, Sabrina N. VandenHeuvel, Sanjana Roy, Brinlee Goggans, Shubha Holla, Varsha Rajavel, Joseph Duran, Lucia L. Nash, Daniel L. Alge, Alex J. Walsh and Shreya A. Raghavan
{"title":"Seeing deep to map cell–biomaterial interactions via whole mount light sheet imaging and automated analytics in decellularized extracellular matrix models","authors":"Oscar R. Benavides, Sabrina N. VandenHeuvel, Sanjana Roy, Brinlee Goggans, Shubha Holla, Varsha Rajavel, Joseph Duran, Lucia L. Nash, Daniel L. Alge, Alex J. Walsh and Shreya A. Raghavan","doi":"10.1039/D5BM00630A","DOIUrl":"10.1039/D5BM00630A","url":null,"abstract":"<p > <em>In vitro</em> models aim to improve biomimicry of <em>in vivo</em> tissues and disease processes. Decellularized extracellular matrix (dECM) scaffolds mimic cellular interactions with 3D tissue architecture. These complex 3D models require parallel advancements in analytical methods to quantify functional outputs with respect to scaffold architecture and recellularization while retaining spatial integrity. Current imaging methods optimized in other engineered model systems are limited in their application to dECM due to the inherent thickness, high heterogeneity, opacity and autofluorescence of dECM material. Sacrificial sample preparation methods like digestion and tissue sectioning are tedious and reduce the amount of spatially relevant information that can be extracted. Further, imaging depth and resolution are limited due to light scattering within large opaque scaffolds. We aimed to optimize optical imaging and analysis protocols to overcome imaging challenges and enable quantitative assessments of dECM models, demonstrating a use case in engineered <em>in vitro</em> cancer microenvironments. We first combined a series of established sample preparation methods including tissue clearing agents and cell labeling dyes, to optimize dECM scaffold compatibility with volumetric light sheet fluorescence microscopy (LSFM) imaging. We then developed image analysis algorithms capable of overcoming the segmentation limitations of established methods to accurately quantify scaffold porosity as well as cellular occupation and migration at the single cell level within dECM scaffolds. We automated this analysis to increase usability for large data sets and applied the imaging methods to a porcine liver-derived dECM scaffold model, called a biomatrix. Biomatrices recellularized with colorectal cancer spheroids model liver metastasis. The LSFM imaging and analysis successfully detected increased cell numbers between 3 and 5 days of culture on the dECM biomatrix, and the loss of cells in oxaliplatin-treated biomatrices. With the ability to resolve these key changes in proliferation, invasion and therapeutic response, this optimized set of imaging and computational tools will aid in the mechanistic and therapeutic discovery of colorectal cancer liver metastasis. Broadly, the increased volume and resolution of imaging data from our methods can extract spatially relevant scaffold and cellular information within the context of any dECM model, increasing its adoptability to probe complex biological behaviors across diseases.</p>","PeriodicalId":65,"journal":{"name":"Biomaterials Science","volume":" 19","pages":" 5442-5459"},"PeriodicalIF":5.7,"publicationDate":"2025-08-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12368843/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144936223","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Real-time monitoring of mitochondrial temperature and calcium spikes using fluorescent organic probes","authors":"Mengnan Sun, Bo Chen and Aiguo Wu","doi":"10.1039/D5BM00691K","DOIUrl":"10.1039/D5BM00691K","url":null,"abstract":"<p >Astrocytes, the abundant glial cells, maintain cerebral homeostasis and cognitive functions through calcium signalling – a regulatory pathway that is frequently altered in brain disease. Mitochondria serve as thermal hubs in living systems, generating metabolic heat during respiratory substrate oxidation and ATP synthesis. Crucially, mitochondrial temperature variations directly reflect metabolic status, as impaired ATP production induces thermodynamic shifts. Here, we utilized a fluorescent thermometer probe MTY for <em>in vitro</em> determination and visualization of intracellular mitochondrial temperatures at the single-cell level. Through precisely controlled thermal modulation of fixed, living, and laser-stimulated astrocytes, we established a platform extendable to MCF-7 and Panc02 cell lines. The methodology enabled real-time tracking of near-infrared-induced thermal perturbations and FCCP-mediated uncoupling effects. Spinning-disk confocal microscopy revealed synchronized mitochondrial thermogenesis and calcium transients, with thermal/laser stimulation inducing 2–4-fold greater calcium spiking <em>versus</em> controls. Mechanistic analysis suggested this response was likely mediated through TRPV4 channel-mediated extracellular Ca<small><sup>2+</sup></small> influx and/or intracellular calcium release from mitochondrial and endoplasmic reticulum stores.</p>","PeriodicalId":65,"journal":{"name":"Biomaterials Science","volume":" 19","pages":" 5512-5521"},"PeriodicalIF":5.7,"publicationDate":"2025-08-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://pubs.rsc.org/en/content/articlepdf/2025/bm/d5bm00691k?page=search","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144936242","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Ke Son Phan, Bich Thuy Doan, Yiqian Wang, Thi Thu Huong Le, Thi Thu Trang Mai, Hong Nam Pham, Ha Bao Hung Bui, Le Hang Dang, Ngoc Quyen Tran and Phuong Thu Ha
{"title":"Harnessing for quantitative MRI, bioluminescence, chemo-magnetic hyperthermia, and near-infrared optical imaging of a magnetic nanotheranostic for in vivo passive targeted monitoring in colorectal cancer models","authors":"Ke Son Phan, Bich Thuy Doan, Yiqian Wang, Thi Thu Huong Le, Thi Thu Trang Mai, Hong Nam Pham, Ha Bao Hung Bui, Le Hang Dang, Ngoc Quyen Tran and Phuong Thu Ha","doi":"10.1039/D5BM00659G","DOIUrl":"10.1039/D5BM00659G","url":null,"abstract":"<p >Colorectal cancer is the second foremost cause of cancer-related fatalities, but it is currently missing an effective treatment. For researchers, the ability to monitor therapeutic nanoparticles of interest within a living animal using non-invasive observation tools has been a futuristic idea for a long time. In this work, a multifunctional magnetic nanotheranostic functionalized with Cyanine 5.5 and Doxorubicin was tested to enhance bio-monitoring and therapy efficiency in an <em>in vivo</em> CT26 murine colon model <em>via</em> retro-orbital injection. The <em>in vivo</em> biodistribution was measured by near-infrared optical imaging, which showed that the nanotheranostic largely accumulated in the tumor with maximum uptake observed at the 3 h postinjection time point. This nanotheranostic was then metabolized and eliminated <em>via</em> the kidneys after 6 h and both the liver and spleen after 72 h of injection. Besides, the magnetic resonance imaging (MRI) modality with a specific <em>T</em>2*-weighted sequence demonstrates efficient nanoparticle accumulation within the tumor by the %<em>I</em><small><sub>0.25</sub></small> quantitative method of hyposignal processing. Moreover, bioluminescence imaging demonstrated a significant chemotherapeutic effect after 72 h of injection of a 15 mg kg<small><sup>−1</sup></small> single dose. Hyperthermia treatment by AMF significantly impacts the synergistic efficiency of this second therapy provided by the Fe<small><sub>3</sub></small>O<small><sub>4</sub></small> nanoparticle (NP) content. The resulting nanotheranostic demonstrated enhanced passive accumulation and selectively accumulated in the tumor with negligible distribution to adjacent healthy tissue, effectively suppressing tumor proliferation when combined with alternating magnetic field (AMF) stimulation. This powerful synergistic approach has proven to be a robust and versatile nanotheranostic for the effective treatment of colorectal cancer.</p>","PeriodicalId":65,"journal":{"name":"Biomaterials Science","volume":" 19","pages":" 5402-5421"},"PeriodicalIF":5.7,"publicationDate":"2025-08-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144936066","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Andrea Cavero-Arrivasplata, David Hyram Hernández-Medina, Irving Isaí Rendón-Moreno, Diego Alonso Quevedo-Moreno, Dariush Ebrahimibagha, Sanal Kozhiparambil Chandran, Julio Ernesto Valdivia Silva, Claudia Maribel Luna-Aguirre, Mario Moisés Alvarez, Grissel Trujillo-de Santiago
{"title":"Multimaterial chaotic printing of reinforced and prevascularized hydrogel filaments: Fabrication of mechanical robust constructs for long-term muscle tissue culture.","authors":"Andrea Cavero-Arrivasplata, David Hyram Hernández-Medina, Irving Isaí Rendón-Moreno, Diego Alonso Quevedo-Moreno, Dariush Ebrahimibagha, Sanal Kozhiparambil Chandran, Julio Ernesto Valdivia Silva, Claudia Maribel Luna-Aguirre, Mario Moisés Alvarez, Grissel Trujillo-de Santiago","doi":"10.1039/d4bm01674b","DOIUrl":"https://doi.org/10.1039/d4bm01674b","url":null,"abstract":"<p><p>Engineering vascularization in hydrogel constructs remains a significant challenge in tissue engineering. Prevascularized hydrogels, engineered with void channels, enhance cell viability but often lack the mechanical stability needed for long-term culture, which is crucial for proper tissue maturation. In this study, we introduce chaotic bioprinting-a chaos-enabled biofabrication strategy-to produce mechanically robust hydrogel prevascularized filaments (with inner void channels) suited for extended culture. Utilizing a Kenics Static Mixer (KSM) printhead with various inlets (4 or 8), we developed fibers with intercalated layers of a myoblast-laden gelatin methacryloyl (GelMA)-alginate bioink, a sacrificial material for channel formation, and a reinforcing alginate scaffold. By optimizing ink ratios, we maximized cell-laden compartments while reinforcing the fiber structure and embedding microchannels for efficient mass and gas transport. Mechanical testing and degradation analysis reveal that optimized fibers achieve sufficient resistance (elastic modulus = 12.8 kPa) to withstand extended periods of cell culture up to 21 days. Additionally, C2C12 myoblasts cultured within these prevascularized and reinforced hydrogel filaments maintained high cell viability (>90%) for more than 21 days and demonstrated superior cell proliferation, spreading, and alignment throughout the filament volume compared to solid fibers (reinforced but without inner void channels). Sacrificial layers created void microchannels, enhancing mass and gas transport, while the reinforcing layers provided structural integrity. Multimaterial chaotic printing enabled the fabrication of mechanically stable, functional constructs with compartmentalized architectures, facilitating extended culture and tissue maturation. Our results demonstrate the potential of this method for engineering thick tissues, including skeletal muscle, and highlight its versatility for various regenerative medicine applications, advancing biofabrication towards thicker and mature tissues.</p>","PeriodicalId":65,"journal":{"name":"Biomaterials Science","volume":" ","pages":""},"PeriodicalIF":5.7,"publicationDate":"2025-08-11","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144815276","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Silk-based double-sided magnetic microneedles for enhanced targeted retention in the gastrointestinal tract","authors":"Niping Deng, Hao Lyu and Chengchen Guo","doi":"10.1039/D5BM00125K","DOIUrl":"10.1039/D5BM00125K","url":null,"abstract":"<p >Gastrointestinal-retentive drug delivery devices represent a novel oral drug delivery approach capable of significantly enhancing patient compliance and improving local drug delivery efficiency compared to conventional oral formulations. However, developing gastrointestinal-retentive drug delivery devices with high retention capability based on compatible material systems presents significant challenges. Here, we introduce silk-based double-sided magnetic microneedles (SDMMs) designed to facilitate targeted oral drug delivery with precise local retention in the gastrointestinal tract. The SDMM platform demonstrates robust mechanical properties, site-specific degradation, and compatibility with various capsule geometries, facilitating seamless integration into existing oral delivery systems. <em>In vitro</em> analyses demonstrate region-dependent drug release from the SDMM platform, supporting its potential for localized delivery of drugs in the gastrointestinal tract. The incorporation of iron oxide nanoparticles (IONPs) confers magnetically responsive targeting, offering a significant advantage over conventional approaches by ensuring precise retention and enhancing the targeted delivery potential under external magnetic fields. Notably, SDMMs retain their positions under fluid flow rates up to 550 mL min<small><sup>−1</sup></small> (stomach) and 400 mL min<small><sup>−1</sup></small> (small intestine), respectively, underscoring their superior gastrointestinal retention. Furthermore, <em>in vivo</em> retention experiments also showed significantly enhanced retention in the gastrointestinal tract, with the retention time of SDMMs reaching almost 15 hours with magnet assistance, representing significantly prolonged retention compared to typical transit times in these regions. These findings establish SDMMs as a promising platform for precise, efficient, and patient-friendly oral drug delivery.</p>","PeriodicalId":65,"journal":{"name":"Biomaterials Science","volume":" 19","pages":" 5460-5474"},"PeriodicalIF":5.7,"publicationDate":"2025-08-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144936280","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Thi Kim Ngan Duong, Thi Thuy Truong, Thi Nhat Linh Phan, Thi Xuan Nguyen, Vu Hoang Minh Doan, Jaeyeop Choi, Thanh Phuoc Nguyen, Songyi Lee, Jaesung Ahn, Junghwan Oh and Sudip Mondal
{"title":"Near-infrared responsive Au–Mn–HAp nanocomposite for fluorescence imaging and combined phototherapy for breast cancer","authors":"Thi Kim Ngan Duong, Thi Thuy Truong, Thi Nhat Linh Phan, Thi Xuan Nguyen, Vu Hoang Minh Doan, Jaeyeop Choi, Thanh Phuoc Nguyen, Songyi Lee, Jaesung Ahn, Junghwan Oh and Sudip Mondal","doi":"10.1039/D5BM00871A","DOIUrl":"10.1039/D5BM00871A","url":null,"abstract":"<p >This study investigated a microwave-assisted synthesis method for the rapid deposition of gold (Au) nanoparticles (NPs) doped with manganese (Mn) onto the surface of hydroxyapatite (HAp). HAp NPs were first synthesized using a wet precipitation method, followed by Au loading and Mn doping to achieve a synergistic imaging-guided photothermal/photodynamic effect in breast cancer treatment. The substitution of Ca<small><sup>2+</sup></small> ions with smaller Mn<small><sup>2+</sup></small> ions enhanced the formation of Mn–HAp, which served as a nano-carrier with a large surface area, enabling conjugation of folic acid (FA) and indocyanine green (ICG) fluorescent dye without any toxicity. This innovative approach considerably enhanced the efficacy of photodynamic therapy (PDT) by generating reactive oxygen species (ROS) to eliminate cancer cells. The Au–Mn–HAp–FA–ICG nanocomposite showed promising contrast efficiency and enhanced PDT, acting as an effective photodynamic probe, as confirmed by DCFH-DA staining. <em>In vitro</em> experiments demonstrated the potent efficacy of Au NPs in combination with fluorescence-imaging-guided PDT treatment, achieving effective photothermal ablation of cancer cells after 5 min of laser irradiation. The biocompatibility of the synthesized NPs was assessed using an MTT assay and fluorescence staining, which revealed that the nanostructured materials could be ideal agents for imaging-guided cancer therapy.</p>","PeriodicalId":65,"journal":{"name":"Biomaterials Science","volume":" 18","pages":" 5145-5163"},"PeriodicalIF":5.7,"publicationDate":"2025-08-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144792985","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yin Chen, Na Zhao, Songling Han, Gaomei Zhao, Yiyi Jiang, Chenwenya Li, Yingjuan He, Yangxue Ou, Jining Gao, Tao Wang, Jinghong Zhao, Jia Cao, Shilei Chen, Junping Wang, Cheng Wang
{"title":"Tricobalt tetraselenide nanoparticles improve intestinal barrier function by reshaping the gut microbiota and fortifying epithelial tight junctions.","authors":"Yin Chen, Na Zhao, Songling Han, Gaomei Zhao, Yiyi Jiang, Chenwenya Li, Yingjuan He, Yangxue Ou, Jining Gao, Tao Wang, Jinghong Zhao, Jia Cao, Shilei Chen, Junping Wang, Cheng Wang","doi":"10.1039/d5bm00712g","DOIUrl":"https://doi.org/10.1039/d5bm00712g","url":null,"abstract":"<p><p>The disrupted intestinal mechanical barrier contributes to the pathogenesis of inflammatory bowel disease (IBD) and irradiation-induced intestinal injury (IRIII), both of which lack sufficient medical countermeasures currently. Reshaping the gut microbiota to increase the short-chain fatty acid (SCFA) levels is instrumental for fortifying the mechanical barrier and mitigating these diseases. Inspired by the fact that supplementation of essential trace elements selenium (Se) and cobalt (Co) can enrich SCFA-producing bacteria and promote microbial fermentation, respectively, we prepared tricobalt tetraselenide nanoparticles (Co<sub>3</sub>Se<sub>4</sub> NPs) using a facile biomineralization approach in this study. Oral Co<sub>3</sub>Se<sub>4</sub> NP administration increased intestinal microbial diversity and enriched <i>Alloprevotella</i>, which produced SCFAs, particularly butyric acid (BA), in the gut. Further investigation revealed that BA activated the cell-surface G-protein-coupled receptor 43 (GPR43) in enterocytes and induced protein kinase B (AKT) phosphorylation, resulting in increased expression of the tight junction (TJ) proteins occludin and zonula occludens-1. Accordingly, Co<sub>3</sub>Se<sub>4</sub> NPs ameliorated the gut microbiota dysbiosis and TJ disruption caused by dextran sulfate sodium salt (DSS) and total abdominal γ-ray irradiation, producing a fortified mechanical barrier and alleviating DSS-induced colitis and IRIII in mice. This study demonstrated a biocompatible nanomaterial that can reshape the gut microbiota and fortify epithelial TJs, thereby providing an effective strategy for IBD and IRIII therapies.</p>","PeriodicalId":65,"journal":{"name":"Biomaterials Science","volume":" ","pages":""},"PeriodicalIF":5.7,"publicationDate":"2025-08-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144774351","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Immune modulation strategies to reduce in-stent restenosis","authors":"Mei Yu, Chengyi Dai, Jingjing Shi and Jiayin Fu","doi":"10.1039/D5BM00687B","DOIUrl":"10.1039/D5BM00687B","url":null,"abstract":"<p >In-stent restenosis remains a significant complication following stent implantation, driven by complex interactions between immune responses, vascular injury, and inflammatory cascades. Despite advancements in stent technology, ISR persists, underscoring the need for innovative strategies to modulate immune activity and promote vascular healing. This review presents current knowledge on immune-mediated mechanisms of ISR, highlighting the pivotal roles of immune cell like neutrophils, macrophages in neointimal hyperplasia and chronic inflammation. We explore recent immunomodulatory approaches, including stent surface modifications, bioactive molecule delivery, and emerging technologies. Furthermore, we evaluate the clinical potential of next-generation stents such as endothelial-mimetic designs to mitigate ISR by balancing pro-reparative and anti-inflammatory signals. By integrating insights from preclinical and clinical studies, this review provides a new perspective for developing “immune-friendly” stents, emphasizing interdisciplinary strategies to attenuate ISR following stent implantation.</p>","PeriodicalId":65,"journal":{"name":"Biomaterials Science","volume":" 18","pages":" 4898-4915"},"PeriodicalIF":5.7,"publicationDate":"2025-08-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144774350","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":3,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}