{"title":"HAMA-SBMA hydrogel with anti-inflammatory properties delivers cartilage organoids, boosting cartilage regeneration.","authors":"Yuyang Gao, Qingshan Li, Zhangzhen Du, Qianru Yao, Gehan Jiang, Wenxing Huang, Xiang Gao, Juntan Li, Tianxu Dou, Fangping Chen, Xu Li, Aiyuan Wang, Jiang Peng","doi":"10.1186/s12951-025-03475-y","DOIUrl":"10.1186/s12951-025-03475-y","url":null,"abstract":"<p><p>Cartilage tissue lacks blood supply, which limits its ability to self-repair. Cartilage organoid (CO) technology, which replicates the structure and function of cartilage, holds significant promise. However, it is essential to maintain cellular function and ensure secure fixation at the site of injury. Therefore, we loaded allogeneic bone marrow mesenchymal stem cells (BMSCs) onto decellularized extracellular matrix microparticles of porcine articular cartilage (CEP) to construct CO-CCO, which demonstrated characteristics of articular cartilage. Additionally, betaine sulfonate methacrylate (SBMA) was incorporated into hyaluronic acid methacrylate (HAMA) to synthesize a novel hydrogel, HAMA-SBMA (HS), characterized by its adhesive properties, promotion of chondrogenesis, and inhibition of inflammation. In Vivo studies revealed that the combination of HS and CCO (HS + CCO) exhibited excellent repair efficacy in both rat and sheep models of cartilage defects. Mechanistically, we found that HS + CCO promoted cartilage repair by activating the Frizzled-related protein (Frzb), which inhibited inflammatory factors and enhanced the expression of the adhesion factor integrin ɑ5β1. This strategy, which combines hydrogels and organoids, enhances cartilage repair, offering substantial potential for clinical applications in cartilage regeneration.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"401"},"PeriodicalIF":10.6,"publicationDate":"2025-05-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12124039/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144187169","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Advanced oral drug delivery systems for gastrointestinal targeted delivery: the design principles and foundations.","authors":"Yafei Zhang, Yiran Wang, Yao Lu, Heng Quan, Yuqi Wang, Sijia Song, Huiyuan Guo","doi":"10.1186/s12951-025-03479-8","DOIUrl":"10.1186/s12951-025-03479-8","url":null,"abstract":"<p><p>Oral administration has long been considered the most convenient method of drug delivery, requiring minimal expertise and invasiveness. Unlike injections, it avoids discomfort, wound infections, and complications, leading to higher patient compliance. However, the effectiveness of oral delivery is often hindered by the harsh biological barriers of the gastrointestinal tract, which limit the bioaccessibility and bioavailability of drugs. The development of oral drug delivery systems (ODDSs) represents a critical area for the advancement of pharmacotherapy. This review highlights the characteristics and precise targeting mechanisms of ODDSs. It first examines the unique properties of each gastrointestinal compartment, including the stomach, small intestine, intestinal mucus, intestinal epithelial barrier, and colon. Based on these features, it outlines the targeting strategies and design principles for ODDSs aimed at overcoming gastrointestinal barriers to enhance disease treatment. Lastly, the review discusses the challenges and potential future directions for ODDS development, emphasizing their importance for advancing drug delivery technologies and accelerating their future growth.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"400"},"PeriodicalIF":10.6,"publicationDate":"2025-05-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12124048/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144187164","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Jiebang Jiang, Zhiyuan Pan, Yiren Su, Lu Dai, Nana Xu, Han Wu, Xin Chen
{"title":"Carbon dots from purple sweet potato as a promising anti-inflammatory biomaterial for alleviating the LPS-induced inflammation in macrophages.","authors":"Jiebang Jiang, Zhiyuan Pan, Yiren Su, Lu Dai, Nana Xu, Han Wu, Xin Chen","doi":"10.1186/s12951-025-03494-9","DOIUrl":"10.1186/s12951-025-03494-9","url":null,"abstract":"<p><p>This study synthesizes carbon dots derived from crude extracts of purple sweet potato (CPP-CDs) and evaluates its anti-inflammatory effects in a lipopolysaccharide (LPS) -induced acute inflammation model. Characterization revealed that CPP-CDs possess a uniform spherical structure and excellent photoluminescent properties. In vitro, CPP-CDs significantly inhibited the expression of pro-inflammatory cytokines (IL-1β, IL-6, TNF-α), reduced the accumulation of reactive oxygen species (ROS), suppressed pyroptosis, and facilitated the polarization of macrophages from the M1 phenotype to the M2 phenotype. In vivo, CPP-CDs significantly improved the survival rates of LPS-treated mice, mitigated tissue damage, and suppressed the levels of pro-inflammatory cytokines. Mechanistic studies indicated that CPP-CDs exert anti-inflammatory effects through the inhibition of the TLR4/NF-κB signaling pathway and the modulation of the NLRP3 inflammasome. Additionally, CPP-CDs exhibited excellent biocompatibility, with no significant toxicity observed in mice. This study provides strong evidence supporting the application of CPP-CDs as a novel anti-inflammatory material, highlighting their potential for acute inflammation treatment and expanding the possibilities for the development of carbon-dot-based anti-inflammatory therapies.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"397"},"PeriodicalIF":10.6,"publicationDate":"2025-05-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12124011/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144187165","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Lin Wu, Yang Liu, Ziqi Fu, Honglei Guo, Kang Liu, Jiafa Ren, Zhimin Huang, Fang Yang, Huijuan Mao
{"title":"TRPC6-targeted dexamethasone nanobubbles with ultrasound-guided theranostics for adriamycin-induced nephropathy.","authors":"Lin Wu, Yang Liu, Ziqi Fu, Honglei Guo, Kang Liu, Jiafa Ren, Zhimin Huang, Fang Yang, Huijuan Mao","doi":"10.1186/s12951-025-03487-8","DOIUrl":"10.1186/s12951-025-03487-8","url":null,"abstract":"<p><strong>Background: </strong>Glucocorticoid (GC) intolerance and systemic toxicity pose significant challenges in the treatment of primary nephrotic syndrome (PNS), underscoring the urgent need for targeted therapies that maximize efficacy while minimizing adverse effects. To address these challenges, we developed TRPC6-targeted dexamethasone-loaded nanobubbles (Dex@NBs-TRPC6)-an innovative therapeutic platform that enables selective podocyte delivery alongside real-time monitoring capabilities.</p><p><strong>Results: </strong>The Dex@NBs-TRPC6 nanobubble system comprises polyethylene glycol-modified lipid vesicles encapsulating dexamethasone (Dex), conjugated with TRPC6-specific antibody for precise podocyte targeting delivery. Comprehensive in vivo and in vitro evaluations demonstrated the robust kidney and podocyte-targeting capabilities of Dex@NBs-TRPC6. Functional assays in mouse podocyte cells revealed that Dex@NBs-TRPC6 significantly outperformed free Dex and non-targeted nanobubbles (Dex@NBs) in mitigating cell apoptosis and inflammation. In an adriamycin-induced mouse nephropathy model, Dex@NBs-TRPC6, administered at half the dosage of free Dex, markedly alleviated proteinuria, glomerular and tubular damage, renal apoptosis, inflammation and fibrosis. Notably, Dex@NBs-TRPC6 attenuated the overexpression of hepatic gluconeogenic genes PCK1 and GCP6, a common adverse effect associated with Dex. Furthermore, leveraging the acoustic response properties of Dex@NBs-TRPC6, this delivery system integrates ultrasound imaging capabilities, enabling real-time visualization and therapeutic monitoring.</p><p><strong>Conclusions: </strong>By simultaneously enhancing therapeutic efficacy, minimizing systemic toxicity, and enabling personalized imaging-guided treatment, Dex@NBs-TRPC6 introduces a transformative approach to GC-based renal therapy.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"398"},"PeriodicalIF":10.6,"publicationDate":"2025-05-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12124086/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144187172","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Yapeng Guo, Lei Yang, Li Yao, Chengdong Zhou, Yuanyuan Zhu, Chenxi Xu, Wenlong Wang, Jian Song, Mingzhen Zhang, Zhichao Deng
{"title":"Carbon dots derived from Ligusticum Chuanxiong mitigate cardiac injury by disrupting the harmful oxidative stress-apoptosis cycle.","authors":"Yapeng Guo, Lei Yang, Li Yao, Chengdong Zhou, Yuanyuan Zhu, Chenxi Xu, Wenlong Wang, Jian Song, Mingzhen Zhang, Zhichao Deng","doi":"10.1186/s12951-025-03477-w","DOIUrl":"10.1186/s12951-025-03477-w","url":null,"abstract":"<p><strong>Background: </strong>Myocardial ischemia-reperfusion injury (MIRI) represents a significant complication following myocardial infarction surgery, for which preventive strategies remain limited. The primary pathological characteristics of MIRI include oxidative stress and apoptosis.</p><p><strong>Results: </strong>This study presents the synthesis of carbon dots derived from Ligusticum Chuanxiong (LC-CDs) through the application of the hydrothermal method. The LC-CDs show strong scavenging abilities for free radicals, effectively reducing oxidative stress and preventing apoptosis, which helps combat MIRI. The findings demonstrate that LC-CDs can effectively neutralize excessive ROS within cells, thereby alleviating oxidative stress, restoring mitochondrial function, and preventing DNA damage. Concurrently, LC-CDs suppress the polarization of M1-type macrophages and reduce the secretion of pro-inflammatory cytokines. Following the in situ administration of LC-CDs into the hearts of MIRI-model rats, a significant reduction in the necrotic area of the myocardium was observed, alongside the restoration of cardiac function, with no adverse reactions reported. Moreover, similar to the pharmacological effects of Ligusticum chuanxiong, LC-CDs can also inhibit apoptosis by protecting mitochondria and suppressing the expression of apoptotic proteins (Caspase3, Caspase9, and Bax).</p><p><strong>Conclusions: </strong>The intervention strategy employing LC-CDs, which targets oxidative stress and apoptosis in MIRI, holds promise as a potential model for the clinical treatment of MIRI.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"391"},"PeriodicalIF":10.6,"publicationDate":"2025-05-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12121267/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144181587","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Zhe Zhang, Guangyu Jia, Qi Wang, Yamei Yu, Xiaolong Tang, Heqing Zheng, Xinyu Yang, Yao Xiao, Yangrui Ou, Jingjing Jiang, Hua Guo, Ye Wang, Shiyong Li
{"title":"Atherosclerosis enhances the efficacy of liposome-encapsulated bromocriptine in reducing the incidence of prolactinemia in pituitary tumors.","authors":"Zhe Zhang, Guangyu Jia, Qi Wang, Yamei Yu, Xiaolong Tang, Heqing Zheng, Xinyu Yang, Yao Xiao, Yangrui Ou, Jingjing Jiang, Hua Guo, Ye Wang, Shiyong Li","doi":"10.1186/s12951-025-03465-0","DOIUrl":"10.1186/s12951-025-03465-0","url":null,"abstract":"<p><p>Intranasal drug delivery via nanocarriers has long been a research focus for enhancing drug concentration in the brain. However, the strategy of exploiting blood-brain barrier (BBB) alterations in atherosclerotic mouse models to enhance nanoparticle-mediated delivery of bromocriptine to the hypothalamus for the treatment of prolactinomas with hyperprolactinemia has not yet been reported. This study reveals that in patients with prolactinomas complicated by arteriosclerosis, bromocriptine therapy more effectively attenuates postoperative elevations in prolactin levels. In a mouse model, liposome-encapsulated bromocriptine efficiently traversed the nasal mucosa and entered the intracranial space. Compared with normal mice, bromocriptine-loaded liposomes delivered higher bromocriptine concentrations to the hypothalamus. Single-cell RNA sequencing revealed a significant upregulation of organic anion-transporting polypeptide 1a4 (Oatp1a4) expression in the brain endothelial cells of atherosclerotic mice. Importantly, bromocriptine-loaded liposomes more effectively reduced prolactin levels in a mouse model of prolactinoma with concurrent atherosclerosis. This study provides a theoretical foundation for the precision treatment of prolactinomas in arteriosclerosis.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"392"},"PeriodicalIF":10.6,"publicationDate":"2025-05-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12121278/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144181602","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Myogenic nano-adjuvant for orthopedic-related sarcopenia via mitochondrial homeostasis modulation in macrophage-myosatellite metabolic crosstalk.","authors":"Xudong Zhang, Peng Zhang, Yunliang Zhu, Jiaqing Lou, Peng Wu, Yingjie Wang, Zhengxi Wang, Quan Liu, Baoliang Lu, Qianming Li, Jiawei Mei, Chen Zhu, Wanbo Zhu, Xianzuo Zhang","doi":"10.1186/s12951-025-03480-1","DOIUrl":"10.1186/s12951-025-03480-1","url":null,"abstract":"<p><p>The decline in skeletal muscle mass and muscle strength linked to aging, also known as sarcopenia, is strongly associated with disability, traumatic injury, and metabolic disease in patients. Meanwhile, sarcopenia increases the risk of adverse orthopedic perioperative complications including implant dislocation, infection, loosening, and poor wound healing. Mitochondrial dyshomeostasis in the immune-myosatellite metabolic crosstalk is one of the major pathological factors in sarcopenia. To reduce the incidence of orthopedic perioperative complications in patients, we designed and developed a nano-adjuvant based on two-dimensional layer double hydroxide (LDH) for sustained improvement of systemic and orthopedic-related sarcopenia. Construction of MgAlCo-LDH@UA (MACL@UA) nano-adjuvant was performed by introducing cobalt in magnesium-aluminum LDH and further loading urolithin A (UA). The release of magnesium ions and UA promoted myocyte proliferation, angiogenesis and improved mitochondrial homeostasis. Al acted as an immunomodulatory adjuvant to enhance the metabolic crosstalk between macrophages and myosatellite cells, and prompted macrophage-derived glutamine nourishment. Animal experiments confirmed that vaccination with MACL@UA in systemic sarcopenia and intensive orthopedic perioperative vaccination with MACL@UA significantly enhanced quadriceps muscle mass in rats. This nano-adjuvant offers a solution for long-term improvement of sarcopenia and short-term significant reduction of orthopedic perioperative complications in patients, with promising prospects for clinical application and commercial translation.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"390"},"PeriodicalIF":10.6,"publicationDate":"2025-05-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12117855/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144173996","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Ziyi Mei, Hanyue Li, Chuling Huang, Shiyong Ma, Yuejia Li, Pingmeng Deng, Sha Zhou, Aizhuo Qian, Bin Yang, Jie Li
{"title":"Extracellular vesicles from adipose-derived stromal/stem cells reprogram dendritic cells to alleviate rat TMJOA by transferring mitochondria.","authors":"Ziyi Mei, Hanyue Li, Chuling Huang, Shiyong Ma, Yuejia Li, Pingmeng Deng, Sha Zhou, Aizhuo Qian, Bin Yang, Jie Li","doi":"10.1186/s12951-025-03478-9","DOIUrl":"10.1186/s12951-025-03478-9","url":null,"abstract":"<p><p>Temporomandibular joint osteoarthritis (TMJOA) urgently needs regenerative therapies due to the limited effects of traditional treatments. Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) are considered a potent alternative for MSC therapy for the treatment of TMJOA. However, the specific mechanisms remain inadequately investigated. In this study, we explored how EVs from adipose-derived stromal/stem cells (ASCs) influence the TMJOA model triggered by Complete Freund's Adjuvant in rats and their impact on the state of dendritic cells (DCs) under pathological conditions. Subsequently, we conducted transcriptomic and metabolomic analyses to elucidate the specific mechanisms by which EVs affect DCs. Mechanistically, we demonstrate that EVs transferred functional mitochondria to DCs, which reverses their metabolic states. The internalized functional mitochondria from EVs activate the MAPK/ERK1/2/FoxO1/autophagy pathway, which causes the metabolic reprogramming of DCs and facilitates the achievement of therapeutic effects. These findings provide a mechanistic rationale for utilizing ASCs-EVs as cell-free alternatives to MSC transplantation in TMJOA therapy.</p>","PeriodicalId":16383,"journal":{"name":"Journal of Nanobiotechnology","volume":"23 1","pages":"389"},"PeriodicalIF":10.6,"publicationDate":"2025-05-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12117725/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144159651","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":1,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}