51
|
Gao T, Tang Y, Zeng T, Wang J, Zhang X, Liu Q, Guan X, Tang X, Lu G, Li J, Liu M, Zhang D, Lv S, Gu J. Neuraminidase 1 Exacerbated Glycolytic Dysregulation and Cardiotoxicity by Destabilizing SIRT1 through Interactions with NRF2 and HIF1α. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2414504. [PMID: 40411250 DOI: 10.1002/advs.202414504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 03/20/2025] [Indexed: 05/26/2025]
Abstract
Despite significant therapeutic advances, cumulative DOX-induced cardiotoxicity (DIC) events remain unacceptably high. Recent evidence has underscored the critical role of impaired glycolytic metabolism in cardiovascular damage. Neuraminidase 1 (NEU1), a member of the neuraminidase family, catalyzes the hydrolysis of terminal sialic acids from glycoconjugates. Here, it is aimed to characterize the role of NEU1 on defective glycolysis during DIC. Mouse models with cardiac-specific genetic modifications of Neu1, Nrf2, and Sirt1 underwent functional analyses, and RNA sequencing to clarify NEU1's role in glycolytic metabolism during DIC. It is discovered that NEU1 is highly expressed after DOX exposure and positively correlated with defective glycolysis phenotypes. Cardiomyocyte-specific deficiency of Neu1 ameliorated impaired glycolytic metabolism and DIC, whereas overexpression of Neu1 in cardiomyocytes exacerbated these pathological phenotypes. Mechanistically, the upregulation of Neu1 is attributed to HIF1α's transcriptional repression, which necessitated the collaboration of NRF2. Additionally, the C-terminal region of NEU1 physically interacted with SIRT1, facilitating its lysosomal-mediated degradation and contributing to the aberrant glycolytic phenotype. The pharmacological or genetic manipulation of NRF2 and HIF1α remarkably abolished DOX-induced NEU1 upregulation, compromised glucose metabolism, and DIC progression. Collectively, NEU1 as a key regulator of cardiac glycolysis is established, offering new therapeutic avenues for DIC through maintaining metabolic flexibility.
Collapse
Affiliation(s)
- Ting Gao
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Yufeng Tang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, 250014, China
| | - Tao Zeng
- School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Jie Wang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Xiaohui Zhang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Qingbo Liu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Xun Guan
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Xinyu Tang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Guangping Lu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Jiahao Li
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Mingrui Liu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Dongmei Zhang
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Sixuan Lv
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Junlian Gu
- School of Nursing and Rehabilitation, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| |
Collapse
|
52
|
Jiang Y, Huang S, Chen HF. ActiMut-XGB: Predicting thermodynamic stability of point mutations for CALB with protein language model. Int J Biol Macromol 2025:144609. [PMID: 40414395 DOI: 10.1016/j.ijbiomac.2025.144609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 05/13/2025] [Accepted: 05/22/2025] [Indexed: 05/27/2025]
Abstract
Predicting the functional impact of single-point mutations on protein residual activity, especially after high-temperature incubation, is critical in protein engineering. We present an innovative machine learning model based on eXtreme Gradient Boosting that leverages protein sequence data to predict thermostability, circumventing the need for three-dimensional structural information. Our model integrates features from the ESM2 language model, physicochemical properties, evolutionary features, and positional features. A key advancement is the use of transfer learning with thermal stability data from various proteins, which enhances prediction accuracy and generalizability. To fine-tune and validate the model, we used experimental data from Candida antarctica lipase B single-point mutants, a widely studied enzyme in biocatalysis and industrial applications. Despite potential limitations of Gibbs free energy values in capturing all factors influencing thermostability, our model represents a significant improvement over traditional approaches, providing valuable insights for protein engineering, enzyme optimization, and therapeutic protein development.
Collapse
Affiliation(s)
- Yuxin Jiang
- State Key Laboratory of Microbial metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, Department of Bioinformatics and Biostatistics, National Experimental Teaching Center for Life Sciences and Biotechnology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shuai Huang
- State Key Laboratory of Microbial metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, Department of Bioinformatics and Biostatistics, National Experimental Teaching Center for Life Sciences and Biotechnology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Hai-Feng Chen
- State Key Laboratory of Microbial metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, Department of Bioinformatics and Biostatistics, National Experimental Teaching Center for Life Sciences and Biotechnology, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
53
|
Li X, Zhang X, Qi Y, Jin W, Wen Z, Zhao Y, Li X, Yao X, Shen Z, Zhang F, Lu P, Huang N, Wang X, Liu Y. Conductive bioadhesive hydrogel with controlled astragaloside IV release for ferroptosis-mediated cardiac repair. J Control Release 2025:113874. [PMID: 40414503 DOI: 10.1016/j.jconrel.2025.113874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2025] [Revised: 05/11/2025] [Accepted: 05/19/2025] [Indexed: 05/27/2025]
Abstract
Myocardial infarction (MI) poses significant therapeutic challenges due to the heart's limited regenerative capacity, often resulting in fibrosis, impaired electrical conduction, and adverse ventricular remodeling. To overcome these challenges, we engineered a conductive and bioadhesive hydrogel capable of controlled, sustained release of Astragaloside IV (AST), targeting ferroptosis-associated pathways to promote cardiac repair. The hydrogel was synthesized through dynamic Schiff-base crosslinking between dopamine-functionalized gelatin (GelDA) and aldehyde-modified Pluronic F127 (F127-CHO), with polydopamine-polypyrrole (PDA-PPy) nanoparticles incorporated to enhance electrical conductivity, mechanical stability, and tissue adhesion via synergistic catechol chemistry. Thermosensitive F127 micelles within the hydrogel efficiently encapsulated and delivered AST to the infarction myocardium, overcoming the bioavailability limitations of hydrophobic therapeutics. In vitro studies confirmed that AST-loaded hydrogels effectively suppressed hypoxia-induced ferroptosis in cardiomyocytes by activating the Nrf2/HO-1 pathway, substantially reducing lipid peroxidation, attenuating inflammation, and enhancing cell viability. In vivo evaluations in a rat MI model demonstrated robust myocardial adhesion, restoration of synchronized electrical conduction, reduced fibrosis, and preservation of mitochondrial integrity. Histological assessments further revealed enhanced cardiomyocyte organization, improved intercellular connectivity, and increased neovascularization, collectively contributing to functional ventricular recovery. These results underscore the therapeutic efficacy and mechanistic innovation of this multifunctional hydrogel, presenting substantial translational potential for controlled, ferroptosis-targeted cardiac repair.
Collapse
Affiliation(s)
- Xiaopei Li
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Xiao Zhang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Yuanpu Qi
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Wanjun Jin
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Ziang Wen
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Yixin Zhao
- Breast Disease Center, The Affiliated Hospital of Qingdao University, Qingdao 266071, China
| | - Xiangyu Li
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Xin Yao
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Zihao Shen
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Feng Zhang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Peng Lu
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China
| | - Ningping Huang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, China.
| | - Xiaowei Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China.
| | - Yun Liu
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210000, China.
| |
Collapse
|
54
|
Zheng Y, Wang T, Zhang J, Wei S, Wu Z, Li J, Shi B, Sun Z, Xu W, Zhu J. Plant-Derived Nanovesicles: A Promising Frontier in Tissue Repair and Antiaging. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025. [PMID: 40402864 DOI: 10.1021/acs.jafc.5c01547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2025]
Abstract
In recent years, mammal-derived extracellular vesicles (EVs) have been widely used in studies on tissue repair and antiaging. Their therapeutic potential lies in mediating intercellular communication through the transfer of various bioactive molecules. As research on nanovesicles progresses, plant-derived nanovesicles (PDNVs) have attracted growing attention as a promising alternative. As an emerging cross-species regulatory "natural force", PDNVs have attracted considerable interest due to their excellent biocompatibility, low immunogenicity, and remarkable therapeutic effects in tissue injury and aging-related diseases. In this review, we examine the bioactive components, drug delivery potential, and functional mechanisms of PDNVs, and we summarize recent advances in their applications for tissue repair and antiaging. In addition, we systematically discuss the major challenges and limitations hindering the clinical translation and industrialization of PDNVs, and we propose five strategic approaches along with future research directions. This review aims to promote further investigation of PDNVs in regenerative medicine and enhance their potential for clinical application.
Collapse
Affiliation(s)
- Yuzhou Zheng
- Vascular Surgery Department, Affiliated Kunshan Hospital of Jiangsu University, Kunshan 215300, China
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Tangrong Wang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Jiaxin Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Sen Wei
- Vascular Surgery Department, Affiliated Kunshan Hospital of Jiangsu University, Kunshan 215300, China
| | - Zhijing Wu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Jiali Li
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Beihao Shi
- Vascular Surgery Department, Affiliated Kunshan Hospital of Jiangsu University, Kunshan 215300, China
| | - Zixuan Sun
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Wenrong Xu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Jian Zhu
- Vascular Surgery Department, Affiliated Kunshan Hospital of Jiangsu University, Kunshan 215300, China
| |
Collapse
|
55
|
Chen X, Liu J, Tang Z, Liu S, Peng J, Liang H, Niu X. Machine Learning-Enabled Time-Resolved Nanozyme-Encoded Recognition of Endogenous Mercaptans for Disease Diagnosis. Anal Chem 2025; 97:10463-10473. [PMID: 40340311 DOI: 10.1021/acs.analchem.5c01539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
With their important role in regulating intracellular redox balance and maintaining cell homeostasis, endogenous mercaptans are recognized as biomarkers of many diseases in clinical practice, and thus establishing efficient yet simple methods to distinguish and quantify endogenous mercaptans is of great significance for health management. Here, we propose a machine learning-enabled time-resolved nanozyme-encoded strategy to identify endogenous mercaptans in the presence of potential interferents for disease diagnosis. Diethylenetriaminepenta(methylenephosphonic) acid was first employed to coordinate with Mn3+ to prepare a new amorphous nanozyme, which exhibited excellent oxidase-like activity in catalyzing the oxidation of colorless 3,3',5,5'-tetramethylbenzidine to its blue oxide. The addition of endogenous mercaptans (cysteine, homocysteine, and glutathione) could competitively suppress the chromogenic process to different extents due to their discrepant antioxidant abilities, providing specific fingerprints over time for each species. With this mechanism, a time-resolved sensor array with the nanozyme as a sole sensing unit was constructed to accurately identify different types and levels of mercaptans and their various mixtures with the help of pattern recognition. Furthermore, machine learning was combined with the sensor array to construct a stepwise prediction model consisting of concentration-independent classification and concentration-associated regression, which could not only differentiate cancer cells from normal ones based on intracellular glutathione but also evaluate the severity of cardiovascular diseases according to serum homocysteine, showing great application potential in disease diagnosis.
Collapse
Affiliation(s)
- Xinyu Chen
- School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, P. R. China
| | - Jinjin Liu
- School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, P. R. China
| | - Zheng Tang
- School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, P. R. China
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, P. R. China
| | - Shuangquan Liu
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, P. R. China
| | - Jiayi Peng
- School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, P. R. China
| | - Hao Liang
- School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, P. R. China
| | - Xiangheng Niu
- School of Public Health, Hengyang Medical School, University of South China, Hengyang 421001, P. R. China
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang 421001, P. R. China
| |
Collapse
|
56
|
Shahin-Shamsabadi A, Cappuccitti J. Muscle-specific acellular ECM fibers made with anchored cell sheet engineering support regeneration in rat models of volumetric muscle loss. Acta Biomater 2025:S1742-7061(25)00348-4. [PMID: 40399155 DOI: 10.1016/j.actbio.2025.05.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Revised: 04/28/2025] [Accepted: 05/07/2025] [Indexed: 05/23/2025]
Abstract
Volumetric muscle loss (VML), a condition affecting millions due to trauma, represents a critical unmet need in regenerative medicine, with no established standard of care. This study introduces a de novo therapeutic strategy using tissue-specific skeletal muscle acellular extracellular matrix (aECM) fibers fabricated using scaffold-free Anchored Cell Sheet Engineering technique. These engineered fibers replicate the native ECM composition and microarchitecture of skeletal muscle, incorporating essential structural and basement membrane proteins. In a rat VML model, aECM demonstrated promising regenerative capacity compared to commercial porcine-derived small intestine submucosa (SIS) ECM. Over an 8-week period, compared to contralateral muscle, aECM preserved muscle volume and weight, regulated inflammatory and fibrotic responses, and promoted vascularization. In contrast, SIS was rapidly degraded by week 4 and associated with fibrosis. Force recovery in aECM was lower at the 8-week time point (77 % compared to 91 % in control), but histological and immunohistochemical analyses revealed newly formed, dispersed muscle fibers exclusively in aECM treatment. Importantly, muscle weight was preserved only when aECM was used, resulting in similar normalized force-to-weight across all groups (87 % in aECM vs. 88 % in SIS). The histological analyses further demonstrated ongoing tissue remodeling, indicative of sustained regeneration, in contrast to the premature fibrotic healing observed in the other groups. An innovative quantitative image analysis workflow enabled assessment of spatial tissue heterogeneity through histology and immunohistochemistry images, setting a new standard for regeneration analysis. These findings establish engineered tissue-specific aECM as a transformative approach for VML treatment, laying the groundwork for translation to clinical applications. STATEMENT OF SIGNIFICANCE: The current study introduces a transformative approach to treating volumetric muscle loss (VML) through the development of tissue-specific acellular extracellular matrix (aECM) fibers engineered using a scaffold-free biofabrication platform uniquely suited for recreation of such aECM components. The engineered fibers represent a significant advancement over current commercial options by recreating native ECM composition and microarchitecture while eliminating complications associated with xenogenic materials. Through comprehensive in vivo evaluation in a rat model, it is demonstrated that these engineered fibers maintain muscle mass and promote controlled tissue regeneration, addressing key limitations of existing treatments. The scaffold-free biofabrication of tissue-specific aECM provides a new paradigm for biomaterial design in regenerative medicine.
Collapse
Affiliation(s)
| | - John Cappuccitti
- Evolved.Bio, 280 Joseph Street, Kitchener, Ontario, N2G4Z5, Canada
| |
Collapse
|
57
|
Venturini J, Chakraborty A, Baysal MA, Tsimberidou AM. Developments in nanotechnology approaches for the treatment of solid tumors. Exp Hematol Oncol 2025; 14:76. [PMID: 40390104 PMCID: PMC12090476 DOI: 10.1186/s40164-025-00656-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2025] [Accepted: 04/16/2025] [Indexed: 05/21/2025] Open
Abstract
Nanotechnology has revolutionized cancer therapy by introducing advanced drug delivery systems that enhance therapeutic efficacy while reducing adverse effects. By leveraging various nanoparticle platforms-including liposomes, polymeric nanoparticles, and inorganic nanoparticles-researchers have improved drug solubility, stability, and bioavailability. Additionally, new nanodevices are being engineered to respond to specific physiological conditions like temperature and pH variations, enabling controlled drug release and optimizing therapeutic outcomes. Beyond drug delivery, nanotechnology plays a crucial role in the theranostic field due to the functionalization of specific materials that combine tumor detection and targeted treatment features. This review analyzes the clinical impact of nanotechnology, spanning from early-phase trials to pivotal phase 3 studies that have obtained regulatory approval, while also offering a critical perspective on the preclinical domain and its translational potential for future human applications. Despite significant progress, greater attention must be placed on key challenges, such as biocompatibility barriers and the lack of regulatory standardization, to ensure the successful translation of nanomedicine into routine clinical practice.
Collapse
Affiliation(s)
- Jacopo Venturini
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Unit 455, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
- Current Affiliation: Department of Medical Oncology, Careggi University Hospital, Florence, Italy
| | - Abhijit Chakraborty
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Unit 455, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Mehmet A Baysal
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Unit 455, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Apostolia M Tsimberidou
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Unit 455, 1515 Holcombe Boulevard, Houston, TX, 77030, USA.
| |
Collapse
|
58
|
Zhong Y, Fang C, Yao T, Wang H, Jiang M. Association between the serum alpha-1-acid glycoprotein concentrations and depression in US adult women: a cross-sectional study. BMC Psychiatry 2025; 25:489. [PMID: 40375217 DOI: 10.1186/s12888-025-06934-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 05/02/2025] [Indexed: 05/18/2025] Open
Abstract
BACKGROUND Emerging evidence has demonstrated a positive association of inflammation with depression. As an acute-phase reactant predominantly synthesized in hepatocytes, alpha-1-acid glycoprotein (AGP) serves as a sensitive biomarker of inflammation. However, there is a limited study to explore the relationship between AGP and depression. Currently, the association of AGP with depression is controversial. METHODS This study utilized data from the National Health and Nutrition Examination Survey (NHANES) collected between 2021 and 2023. The Patient Health Questionnaire-9 (PHQ-9) was employed to assess depressive symptoms, with a score ≥ 10 indicating clinically relevant depression. We utilized weighted multivariate logistic regression for depression outcomes, weighted linear regression for continuous PHQ-9 scores, and restricted cubic splines (RCS) to examine potential nonlinear relationships between AGP and depression. To evaluate the robustness of these associations, we conducted comprehensive subgroup analyses with interaction tests and multiple sensitivity analyses. RESULTS Serum AGP concentrations exhibited a significant positive association with depression among U.S. adult women, demonstrating a linear dose-response relationship. In the fully adjusted model, each ln-unit increase in AGP concentrations was associated with a 1.13-fold higher odds ratio of depression (OR: 2.13, 95% CI: 1.26-3.64) and a 1.47-point elevation in PHQ-9 values (β: 1.47, 95% CI: 0.37-2.56). Moreover, participants in the highest AGP quartile had a 0.72-fold increased odds ratio of depression (OR: 1.72, 95% CI: 1.03-2.87) and a 1.32-point higher PHQ-9 score (β:1.32, 95% CI: 0.31-2.34) compared to those in the lowest quartile. This positive association remained consistent across multiple subgroup analyses. CONCLUSIONS Serum AGP concentrations demonstrated a significant positive linear association with depressive symptoms among nationally representative samples of U.S. adult women.
Collapse
Affiliation(s)
- Yuanyuan Zhong
- Department of Pharmacy, The Third People's Hospital of Yunnan Province, Kunming, Yunnan, 650011, China
| | - Chunyue Fang
- Department of Pharmacy, The Third People's Hospital of Yunnan Province, Kunming, Yunnan, 650011, China
- College of Pharmacy, Dali University, Dali, Yunnan, China
| | - Tianliang Yao
- Department of Control Science and Engineering, College of Electronic and Information Engineering, Tongji University, No. 1239, Siping Road, Shanghai, 200092, China
| | - Hongjin Wang
- Department of Cardio-Thoracic Surgery, Longyan First Affiliated Hospital of Fujian Medical University, Longyan, China.
| | - Min Jiang
- The First Affiliated Hospital , College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China.
| |
Collapse
|
59
|
Yuan X, Wang S, Yuan Z, Wan Z, Zhang L, Song R, Ge L, Zhao Y. Boosting the angiogenesis potential of self-assembled mesenchymal stem cell spheroids by size mediated physiological hypoxia for vascularized pulp regeneration. Acta Biomater 2025; 198:102-114. [PMID: 40216320 DOI: 10.1016/j.actbio.2025.04.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 03/22/2025] [Accepted: 04/08/2025] [Indexed: 04/18/2025]
Abstract
Hypoxia is a pivotal factor in enhancing the vascularization potential of both two-dimensional (2D) cultured cells and three-dimensional (3D) cellular spheroids. Nevertheless, spheroids that closely mimic the in vivo microenvironment often experience excessive hypoxia, leading to the necrotic core and the release of toxic byproducts, ultimately impeding the regenerative process. To balance cell vitality and pro-angiogenic properties of cellular spheroids, this study investigates size-dependent hypoxia in stem cell spheroids utilizing an oxygen transfer finite element model. Subsequently, we develop 3D cultured stem cells from human exfoliated deciduous teeth (SHED) spheroids with regulated size-dependent hypoxia. Comprehensive assessments indicate that SHED spheroids, inoculated at a density of 50,000 cells, display moderate physiological hypoxia, which optimizes their pro-angiogenic potential, fusion capacity, and reattachment ability. Compared with SHED sheets, SHED spheroids enhance vascularized pulp regeneration more effectively with a tightly connected odontoblastic-like layer. Moreover, high-throughput transcriptome sequencing and RT-qPCR analysis further confirm the spheroids' ability to promote angiogenesis and odontogenic differentiation. This study not only introduces a practical and effective approach for regulating size-dependent hypoxia in cellular spheroids, and simultaneously enhancing cell vitality and angiogenic potential, but also paves the way for the clinical application of SHED spheroids in regenerative dental pulp therapies. STATEMENT OF SIGNIFICANCE: The core of three-dimensionally cultured cellular spheroids often experiences hypoxia, and maintaining a balance between the activity and functionality of long-term cultured spheroids in the inevitably hypoxic microenvironment remains a significant challenge. This study introduces a method to optimize the hypoxic conditions of SHED spheroids by employing a reaction-diffusion model, which modulates internal hypoxia to balance cellular viability and angiogenic potential. Compared to two-dimensional cell sheets, the optimized SHED spheroids with high cell vitality, angiogenesis potential, tissue integration and reattatchment ability show superior efficacy in promoting the formation of vascularized pulp-like tissue. This work offers valuable insights into the role of hypoxia in stem cell spheroids functionality and provides a foundation for further research into the optimization of stem cell-based therapies for multiple clinical applications.
Collapse
Affiliation(s)
- Xiaojing Yuan
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, 100081, PR China
| | - Shuyi Wang
- Department of Mechanics and Engineering Science, and Beijing Innovation Center for Engineering Science and Advanced Technology, College of Engineering, Peking University, Beijing 100871, PR China
| | - Zuoying Yuan
- Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, PR China; Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, PR China.
| | - Zhuo Wan
- Department of Mechanics and Engineering Science, and Beijing Innovation Center for Engineering Science and Advanced Technology, College of Engineering, Peking University, Beijing 100871, PR China
| | - Linxue Zhang
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, 100081, PR China
| | - Rui Song
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, 100081, PR China
| | - Lihong Ge
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, 100081, PR China.
| | - Yuming Zhao
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology, National Center for Stomatology, National Clinical Research Center for Oral Diseases, National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing, 100081, PR China.
| |
Collapse
|
60
|
Wu Y, Wang X, Song L, Zhao Z, Xia Y, Tang K, Wang H, Liu J, Wang Z. Tuning macrophage phenotype for enhancing patency rate and tissue regeneration of vascular grafts. Acta Biomater 2025; 198:245-256. [PMID: 40158766 DOI: 10.1016/j.actbio.2025.03.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 03/14/2025] [Accepted: 03/27/2025] [Indexed: 04/02/2025]
Abstract
Macrophages are primary immune cells that play a crucial role in tissue regeneration during the early stages of biomaterial implantation. They create a microenvironment that facilitates cell infiltration, angiogenesis, and tissue remodeling. In the field of vascular tissue engineering, numerous studies have been conducted to modulate the macrophage phenotype by designing various biomaterials, which in turn enhances the regenerative capacity and long-term patency of vascular grafts. However, the mechanism underlying the different phenotypes of macrophages involved in the tissue regeneration of vascular grafts remains unclear. In this study, vascular grafts loaded with various macrophage phenotypes were developed, and their effects were evaluated both in vivo and in vitro. The RAW 264.7 macrophages (M0) were initially treated with LPS or IL-4/IL-10 and polarized into M1 and M2 phenotypes. Subsequently, M0, M1, and M2 macrophages were seeded onto electrospun PCL scaffolds to obtain macrophage-loaded vascular grafts (PCL-M0, PCL-M1, and PCL-M2). As prepared vascular grafts were implanted into the mouse carotid artery for up to one month. The results indicate that the loading of M2 macrophages effectively enhances the patency rate and neotissue formation of vascular grafts. This is achieved through the development of a well-defined endothelium and smooth muscle layer. RNA sequencing was used to investigate the mechanisms of action of different macrophages on tissue regeneration. The study found that M1 macrophages inhibited tissue regeneration by mediating angiogenesis and chronic inflammation through upregulation of VEGFa, IL-1β, and IL-6 expression. In contrast, M2 macrophages regulate the immune microenvironment by upregulating the expression of IL-4 and TGF-β, thereby promoting tissue regeneration. In conclusion, our study demonstrates how different macrophage phenotypes contribute to the initial inflammatory microenvironment surrounding vascular grafts, thereby modulating the biological process of vascular remodeling. STATEMENT OF SIGNIFICANCE: Regulating the biophysical and biochemical characteristics of biomaterials can induce macrophage polarization and enhance vascular remodeling. In previous work, we fabricated a vascular graft with a macroporous structure that promoted macrophage infiltration and polarization into a pro-regenerative phenotype. To illustrate the mechanism, we established a new mouse model and evaluated the effects of different macrophages on vascular regeneration. The study revealed that tuning macrophage phenotype can impact the initial inflammatory microenvironment by secreting cytokines, which can increase the patency rate and regenerative capacity of vascular grafts. These findings provide essential theoretical support for the development of immunoregulatory scaffolds for vascular and other tissue regeneration.
Collapse
Affiliation(s)
- Yifan Wu
- College of Life Sciences, Tiangong University, Tianjin 300387, China; State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Xixi Wang
- College of Life Sciences, Tiangong University, Tianjin 300387, China; Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Lili Song
- College of Life Sciences, Tiangong University, Tianjin 300387, China; Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Zhe Zhao
- College of Life Sciences, Tiangong University, Tianjin 300387, China
| | - Ying Xia
- College of Life Sciences, Tiangong University, Tianjin 300387, China
| | - Kai Tang
- Department of Cardiovascular Surgery, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Fuwai Hospital, Beijing 100037, China
| | - Huiquan Wang
- College of Life Sciences, Tiangong University, Tianjin 300387, China
| | - Jing Liu
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Zhihong Wang
- Institute of Transplant Medicine, School of Medicine, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin 300071, China.
| |
Collapse
|
61
|
Gong J, Qiu Y, Yu C, Cao C, Li X, Lu J, Zhao W, Zhao Z, Zhang H, Yao F, Sun H, Zhang H, Li J. Injectable Hydrogel for Cardiac Repair via Dual Inhibition of Ferroptosis and Oxidative Stress. ACS APPLIED MATERIALS & INTERFACES 2025; 17:27906-27922. [PMID: 40326674 DOI: 10.1021/acsami.5c02666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
Ferroptosis plays a significant role in ischemic heart disease by exacerbating myocardial injury through oxidative stress, iron metabolism disorder, and inflammation. Herein, we develop an injectable hydrogel (HSD/DFO@GMs) with antiferroptosis and antioxidant properties for cardiac repair. The hydrogel is composed of dopamine-grafted oxidized hyaluronic acid, adipic acid dihydrazide grafted hyaluronic acid, and deferoxamine loaded gelatin microsphere, connected via a dynamic Schiff base bond. This hydrogel exhibits a favorable injectability and stable mechanical properties. It effectively chelates Fe2+ and scavenges the reactive oxygen species (ROS), creating a conducive microenvironment for cardiac repair. The dynamic Schiff base bond and gelatin matrix respond to the weakly acidic and MMP-2-rich microenvironment postinjury, enabling on-demand release of DFO in the injured myocardium. In vitro experiments indicate that the hydrogel significantly inhibits the ferroptosis and oxidative stress damage in H9C2 cardiomyocytes under a hypoxia/reoxygenation microenvironment. In an in vivo ischemia-reperfusion model, the HSD/DFO@GMs hydrogel reduces oxidative stress, modulates intracellular labile iron pool levels, and promotes revascularization, ultimately improving cardiac function. Overall, the HSD/DFO@GMs hydrogel provides a new strategy to improve cardiac repair by inhibiting ferroptosis and mitigating oxidative stress damage.
Collapse
Affiliation(s)
- Jiazhuo Gong
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Yuwei Qiu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Chaojie Yu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Cheng Cao
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Xiuqiang Li
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Jiajun Lu
- School of Basic Medical Sciences, North China University of Science and Technology, Tangshan 063210, China
| | - Weiqing Zhao
- School of Basic Medical Sciences, North China University of Science and Technology, Tangshan 063210, China
| | - Zhongming Zhao
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Haitao Zhang
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Fanglian Yao
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
| | - Hong Sun
- School of Basic Medical Sciences, North China University of Science and Technology, Tangshan 063210, China
- Hebei Key Laboratory for Rehabilitation Engineering and Regenerative Medicine, Tangshan 063210, China
| | - Hong Zhang
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
- State Key Laboratory of Synthetic Biology (Tianjin University), Tianjin 300250, China
| | - Junjie Li
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300350, China
- State Key Laboratory of Synthetic Biology (Tianjin University), Tianjin 300250, China
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300350, China
- Hebei Key Laboratory for Rehabilitation Engineering and Regenerative Medicine, Tangshan 063210, China
| |
Collapse
|
62
|
Ma M, Chu Z, Quan H, Li H, Zhou Y, Han Y, Li K, Pan W, Wang DY, Yan Y, Shu Z, Qiao Y. Natural products for anti-fibrotic therapy in idiopathic pulmonary fibrosis: marine and terrestrial insights. Front Pharmacol 2025; 16:1524654. [PMID: 40438605 PMCID: PMC12116445 DOI: 10.3389/fphar.2025.1524654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 04/29/2025] [Indexed: 06/01/2025] Open
Abstract
Idiopathic Pulmonary Fibrosis (IPF) is a chronic fibrotic interstitial lung disease (ILD) of unknown etiology, characterized by increasing incidence and intricate pathogenesis. Current FDA-approved drugs suffer from significant side effects and limited efficacy, highlighting the urgent need for innovative therapeutic agents for IPF. Natural products (NPs), with their multi-target and multifaceted properties, present promising candidates for new drug development. This review delineates the anti-fibrotic pathways and targets of various natural products based on the established pathological mechanisms of IPF. It encompasses over 20 compounds, including flavonoids, saponins, polyphenols, terpenoids, natural polysaccharides, cyclic peptides, deep-sea fungal alkaloids, and algal proteins, sourced from both terrestrial and marine environments. The review explores their potential roles in mitigating pulmonary fibrosis, such as inhibiting inflammatory responses, protecting against lipid peroxidation damage, suppressing mesenchymal cell activation and proliferation, inhibiting fibroblast migration, influencing the synthesis and secretion of pro-fibrotic factors, and regulating extracellular matrix (ECM) synthesis and degradation. Additionally, it covers various in vivo and in vitro disease models, methodologies for analyzing marker expression and signaling pathways, and identifies potential new therapeutic targets informed by the latest research on IPF pathogenesis, as well as challenges in bioavailability and clinical translation. This review aims to provide essential theoretical and technical insights for the advancement of novel anti-pulmonary fibrosis drugs.
Collapse
Affiliation(s)
- Meiting Ma
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, China
| | - Zhengqi Chu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, China
| | - Hongyu Quan
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, China
| | - Hanxu Li
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, China
| | - Yuran Zhou
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, China
| | - Yanhong Han
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, China
| | - Kefeng Li
- Faculty of Applied Sciences, Macao Polytechnic University, Macau, Macao SAR, China
| | - Wenjun Pan
- Department of Oncology, The Third Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - De-Yun Wang
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, Singapore, Singapore
| | - Yan Yan
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, China
| | - Zunpeng Shu
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, China
| | - Yongkang Qiao
- Key Laboratory of Cell Proliferation and Regulation Biology, Ministry of Education, Faculty of Arts and Sciences, Beijing Normal University, Zhuhai, China
| |
Collapse
|
63
|
Gao Y, Yuan X, Gu R, Wang N, Ren H, Song R, Wan Z, Huang J, Yi K, Xiong C, Yuan Z, Zhao Y. Affinity Modifications of Porous Microscaffolds Impact Bone Regeneration by Modulating the Delivery Kinetics of Small Extracellular Vesicles. ACS NANO 2025; 19:17813-17823. [PMID: 40305788 DOI: 10.1021/acsnano.5c03297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
Biomaterials functionalized with small extracellular vesicles (sEVs) hold great regenerative potential, and their therapeutic efficacy hinges on the delivery kinetics of the sEVs. Achieving rapid and stable loading, along with precisely controlled release of sEVs, necessitates affinity modifications of biomaterials. Here, we provide a quantitative description of the interaction between sEVs and various affinity molecules (i.e., polydopamine (PDA), tannic acid (TA), heparin, polyethylenimine (PEI), and calcium phosphate (CaP)) through molecular dynamics simulation. The interaction strengths followed the order of PDA < heparin < TA < CaP < PEI. To tailor the delivery kinetics of stem cells from human exfoliated deciduous teeth (SHED)-derived sEVs with concentration-dependent bioactivities, we employed two representative affinity molecules, namely PDA and CaP, to modify PLGA porous microscaffolds (PLGA MS), resulting in PDA-modified PLGA MS (PDA@MS) and biomineralized PDA-modified PLGA MS (B/PDA@MS). The B/PDA@MS exhibited the highest loading efficiency (>20 μg/mg microscaffolds) and optimized the release profile of sEVs over 21 days. Upon injection into a 5 mm defect in the rat cranial bone, sEV-loaded B/PDA@MS demonstrated the highest level of bone regeneration, with the new bone volume fraction (BV/TV) and bone mineral density (BMD) reaching 64.0% and 604.5 mg/cm3 within 8 weeks, respectively. This work not only presents a biomineralized microscaffold with sustained sEVs release and high osteogenic potential but also offers guidance on the further design and translation of sEV-functionalized biomaterials with broader applications.
Collapse
Affiliation(s)
- Yike Gao
- Department of Pediatric Dentistry National Engineering Laboratory for Digital and Material Technology of Stomatology, and Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Xiaojing Yuan
- Department of Pediatric Dentistry National Engineering Laboratory for Digital and Material Technology of Stomatology, and Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Ruoheng Gu
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China
| | - Nan Wang
- Department of Stomatology, Peking University Third Hospital, Beijing 100191, China
| | - Huihui Ren
- Department of Pediatric Dentistry National Engineering Laboratory for Digital and Material Technology of Stomatology, and Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Rui Song
- Department of Pediatric Dentistry National Engineering Laboratory for Digital and Material Technology of Stomatology, and Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing 100081, China
| | - Zhuo Wan
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China
| | - Jianyong Huang
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China
| | - Kaikai Yi
- Department of Neuro-Oncology and Neurosurgery, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin 300060, China
| | - Chunyang Xiong
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China
- Wenzhou Institute, University of Chinese Academy of Sciences; Oujiang Laboratory, Wenzhou, Zhejiang 325000, China
| | - Zuoying Yuan
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing, 100191, China
| | - Yuming Zhao
- Department of Pediatric Dentistry National Engineering Laboratory for Digital and Material Technology of Stomatology, and Beijing Key Laboratory of Digital Stomatology, Peking University School and Hospital of Stomatology, Beijing 100081, China
| |
Collapse
|
64
|
Yu X, Zou Z, Li Y, Li J, Chen Y, Shi W, Liu X, Guo R, Cai X. Fiber-reinforced gelatin-based hydrogel biocomposite tubular scaffolds with programmable mechanical properties. Biomed Mater 2025; 20:035031. [PMID: 40306305 DOI: 10.1088/1748-605x/add2bc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 04/30/2025] [Indexed: 05/02/2025]
Abstract
Tissue-engineered tubular scaffolds (TETS) provide an effective repair solution for human tubular tissue loss and damage caused by congenital defects, disease, or mechanical trauma. However, there are still major challenges to developing TETS with excellent mechanical properties and biocompatibility for human tubular tissue repair. Gelatin-based hydrogels are suitable candidates for tissue-engineered scaffolds because they are hydrolyzed collagen products and have excellent biocompatibility and degradability. However, the mechanical properties of gelatin-based hydrogels are relatively poor and do not align well with the mechanical properties of human tubular tissues. Inspired by the extracellular matrix architecture of human tubular tissues, this study utilizes high-precision 3D printing to fabricate ultrafine fiber network tubular scaffolds (UFNTS) that mimic the arrangement of collagen fibers, which are then embedded in a cell-compatible gelatin-based hydrogel, resulting in the preparation of a fiber/hydrogel biocomposite tubular scaffold (BCTS) with tunable mechanical properties and a J-shaped stress-strain response. Finite element analysis was employed to predict the mechanical behavior of the UFNTS and BCTS. Experimental results indicate that by modifying the structural parameters of the UFNTS, the mechanical properties of the BCTS can be effectively tuned, achieving a programmable range of tensile modulus (0.2-4.35 MPa) and burst pressure (1580-7850 mmHg), which broadly covers the mechanical properties of most human tubular tissues. The design and fabrication of BCTS offer a new approach for the development of TETS while also providing a personalized strategy for such scaffolds in tissue engineering.
Collapse
Affiliation(s)
- Xiong Yu
- School of Mechanical Engineering, Guizhou University, Guiyang 550025, People's Republic of China
| | - Zhongfei Zou
- School of Mechanical Engineering, Guizhou Institute of Technology, Guiyang 550003, People's Republic of China
| | - Yi Li
- School of Mechanical Engineering, Guizhou University, Guiyang 550025, People's Republic of China
| | - Jiachun Li
- School of Mechanical Engineering, Guizhou University, Guiyang 550025, People's Republic of China
| | - Yuewei Chen
- School of Mechanical Engineering, Guizhou University, Guiyang 550025, People's Republic of China
| | - Wenhai Shi
- School of Mechanical Engineering, Guizhou University, Guiyang 550025, People's Republic of China
| | - Xixia Liu
- School of Mechanical Engineering, Guizhou University, Guiyang 550025, People's Republic of China
| | - Rui Guo
- School of Mechanical Engineering, Guizhou University, Guiyang 550025, People's Republic of China
| | - Xianhui Cai
- School of Mechanical Engineering, Guizhou University, Guiyang 550025, People's Republic of China
| |
Collapse
|
65
|
Hu Y, Ye A, Cheng L, Lee SJ, Yang Z. Recent progress in fabrication, characterization and application of functional protein aggregates derived from plant proteins. Crit Rev Food Sci Nutr 2025:1-40. [PMID: 40346958 DOI: 10.1080/10408398.2025.2499941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2025]
Abstract
This review highlights recent advancements in fabrication, characterization, and applications of functional plant protein aggregates, emphasizing their growing importance in the food industry because of their sustainability as well as cost-effectiveness compared to animal proteins. While native plant proteins often exhibit limited technofunctional properties, the formation of protein aggregates offers a promising solution. This review explores various aggregation methods, including physical methods (e.g., heat treatment, ultrasonication), chemical modifications (e.g., glycation, acylation), and biological processes (e.g., enzymatic hydrolysis, fermentation), and structural and functional properties changes after these treatments. Advanced characterization techniques such as spectroscopy, microscopy, and rheological methods, are discussed to assess microstructures and key properties like emulsification, gelation, and foaming. Applications of these aggregates in products like beverages, mayonnaise, and whipped cream are highlighted. The review concludes with future research directions to enhance industrial applications and nutritional benefits, providing insights into the potential of plant protein aggregates for developing innovative and sustainable plant-based food and non-food products.
Collapse
Affiliation(s)
- Yinxuan Hu
- School of Food Technology and Natural Sciences, Massey University, Auckland, New Zealand
- Riddet Institute, Massey University, Palmerston North, New Zealand
| | - Aiqian Ye
- Riddet Institute, Massey University, Palmerston North, New Zealand
| | - Lirong Cheng
- Riddet Institute, Massey University, Palmerston North, New Zealand
| | - Sung Je Lee
- School of Food Technology and Natural Sciences, Massey University, Auckland, New Zealand
| | - Zhi Yang
- Department of Human Nutrition, Food and Animal Sciences, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| |
Collapse
|
66
|
Griffiths JI, Chi F, Farmaki E, Medina EF, Cosgrove PA, Karimi KL, Chen J, Grolmusz VK, Adler FR, Khan QJ, Nath A, Chang JT, Bild AH. Blocking cancer-fibroblast mutualism inhibits proliferation of endocrine therapy resistant breast cancer. Mol Syst Biol 2025:10.1038/s44320-025-00104-6. [PMID: 40341770 DOI: 10.1038/s44320-025-00104-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 03/24/2025] [Accepted: 04/10/2025] [Indexed: 05/11/2025] Open
Abstract
In early-stage estrogen receptor-positive (ER + ) breast cancer, resistance to endocrine therapy (ET) and CDK4/6 inhibitors (CDK4/6i) often involve a shift away from estrogen-driven proliferation. The nature and source of compensatory growth signals driving cancer proliferation remain unknown but represent direct therapeutic targets of resistant cells. By analyzing single-cell RNA-sequencing data from serial biopsies of patient tumors, we elucidated compensatory growth signaling pathways activated in ET + CDK4/6i-resistant cancer cells, along with the intercellular growth signal communications within the tumor microenvironment. In most patient tumors, resistant cancer cells increased ERBB growth pathway activity during treatment, only partially through ERBB receptor upregulation. Concurrently, fibroblasts within the tumor increased ERBB ligand communication with cancer cells, as they differentiated to a proliferative and mesenchymal phenotype in response to TGF β signals from cancer cells. In vitro model systems demonstrated molecularly how therapy induces a mutualistic cycle of crosstalk between cancer cells and fibroblasts, fostering a growth factor-rich tumor microenvironment circumventing estrogen reliance. We show that ERBB inhibition can break this cancer-fibroblasts mutualism, targeting an acquired sensitivity of resistant cancer cells.
Collapse
Affiliation(s)
- Jason I Griffiths
- Department of Medical Oncology & Therapeutics, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA, 91010, USA.
- Department of Mathematics, University of Utah 155 South 1400 East, Salt Lake City, UT, 84112, USA.
| | - Feng Chi
- Department of Medical Oncology & Therapeutics, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | - Elena Farmaki
- Department of Medical Oncology & Therapeutics, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | - Eric F Medina
- Department of Medical Oncology & Therapeutics, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | - Patrick A Cosgrove
- Department of Medical Oncology & Therapeutics, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | - Kimya L Karimi
- Department of Medical Oncology & Therapeutics, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | - Jinfeng Chen
- Department of Medical Oncology & Therapeutics, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | - Vince K Grolmusz
- Department of Medical Oncology & Therapeutics, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | - Frederick R Adler
- Department of Mathematics, University of Utah 155 South 1400 East, Salt Lake City, UT, 84112, USA
- School of Biological Sciences, University of Utah 257 South 1400 East, Salt Lake City, UT, 84112, USA
| | - Qamar J Khan
- Division of Medical Oncology, Department of Internal Medicine, The University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Aritro Nath
- Department of Medical Oncology & Therapeutics, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA, 91010, USA
| | - Jeffrey T Chang
- Department of Integrative Biology and Pharmacology, School of Medicine, School of Biomedical Informatics, UT Health Science Center at Houston, Houston, TX, 77030, USA
| | - Andrea H Bild
- Department of Medical Oncology & Therapeutics, City of Hope National Medical Center, 1500 East Duarte Road, Duarte, CA, 91010, USA.
| |
Collapse
|
67
|
Xuan X, Fan J, Zhang J, Ren M, Feng L. Immune in myocardial ischemia/reperfusion injury: potential mechanisms and therapeutic strategies. Front Immunol 2025; 16:1558484. [PMID: 40406107 PMCID: PMC12094985 DOI: 10.3389/fimmu.2025.1558484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 04/14/2025] [Indexed: 05/26/2025] Open
Abstract
Myocardial infarction (MI), which is characterized by high morbidity and mortality, is a serious threat to human life and health, and timely reperfusion therapy to save ischemic myocardium is currently the most effective intervention. Although reperfusion therapy effectively restores coronary blood flow and maximally limits the infarct size, it triggers additional cell death and tissue damage, which is known as myocardial ischemia/reperfusion injury (MIRI). Multiple immune cells are present in the reperfusion area, executing specific functions and engaging in crosstalk during diverse stages, constituting a complex immune microenvironment involved in tissue repair and regeneration after MIRI. Immunotherapy brings new hope for treating ischemic heart disease by modulating the immune microenvironment. In this paper, we explore the regulatory roles of various immune cells during MIRI and the close relationship between different cell deaths and the immune microenvironment. In addition, we present the current status of research on targeting the immune system to intervene in MIRI, with the expectation of providing a basis for achieving clinical translation.
Collapse
Affiliation(s)
- Xiaoyu Xuan
- College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jilin Fan
- Department of Rehabilitation, The Affiliated Hospital of Binzhou Medical University, Binzhou, Shandong, China
| | - Jingyi Zhang
- Department of Traditional Chinese Medicine, The Second Affiliated Hospital of Shandong First Medical University, Shandong, Taian, China
| | - Ming Ren
- Baokang Hospital Affiliated to Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Limin Feng
- The Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Binhai New Area Traditional Chinese Medicine Hospital, Tianjin, China
| |
Collapse
|
68
|
Cuijpers I, Katsburg J, van Loon LJC, Troost FJ, Sthijns MMJPE. Nutritional strategies targeting age-related skeletal muscle fibrosis: underlying mechanisms. Crit Rev Food Sci Nutr 2025:1-21. [PMID: 40336331 DOI: 10.1080/10408398.2025.2498676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
Aging is associated with a reduced number and function of muscle stem cells (MuSC). This results in a decreased muscle regenerative capacity and increased formation of fibrotic tissue, impairing skeletal muscle function. This review provides an overview of in vitro and in vivo animal studies investigating nutritional interventions with the potential to inhibit pathophysiological mechanisms involved in the development of skeletal muscle fibrosis. Mechanism targets include 1) MuSC function and myogenic differentiation, 2) M1 to M2 macrophage polarization, 3) myofibroblast activity or extracellular matrix (ECM) deposition, and 4) reactive oxygen species (ROS) mediated pathways, such as NOX2/4 activity. Most promising nutrients described in this review are phytonutrients, vitamins and amino acids. Quercetin targets multiple pathways (showing decreased inflammation, ECM expression and NOX2/4 activity) in various cell types and tissues (kidney, aorta, liver and (heart) muscle) of rodents and rabbits, which could contribute to fibrosis development. Additionally, sulforaphane is a promising candidate as it inhibits inflammation, ECM expression, and ROS production in mouse skeletal muscle. After validation of the effects in human skeletal muscle, supplementation with these nutrients could be implemented in a multifaceted intervention (including exercise and adequate protein intake) targeting age-related skeletal muscle fibrosis.
Collapse
Affiliation(s)
- Iris Cuijpers
- Department of Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands
- Food Innovation and Health, Centre for Healthy Eating and Food Innovation, Maastricht University Campus Venlo, Venlo, The Netherlands
| | - Joey Katsburg
- Food Innovation and Health, Centre for Healthy Eating and Food Innovation, Maastricht University Campus Venlo, Venlo, The Netherlands
| | - Luc J C van Loon
- Department of Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands
| | - Freddy J Troost
- Department of Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands
- Food Innovation and Health, Centre for Healthy Eating and Food Innovation, Maastricht University Campus Venlo, Venlo, The Netherlands
| | - Mireille M J P E Sthijns
- Department of Human Biology, Institute of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University, Maastricht, The Netherlands
- Food Innovation and Health, Centre for Healthy Eating and Food Innovation, Maastricht University Campus Venlo, Venlo, The Netherlands
| |
Collapse
|
69
|
Zhao L, Xia J, Yang C, Tao J, Zhang S, Hu W, Zhang C, Zhang J, Li Q, Xu Y. Poly(L-lactic acid)/gelatin microfiber membrane loaded with mitomycin C promoting bladder defect repair by anti-fibrosis and antibacterial action. J Mater Chem B 2025; 13:5427-5439. [PMID: 40237778 DOI: 10.1039/d4tb02521k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
When the bladder is affected by factors such as tumors, tuberculosis, or trauma, its normal structure and function can be severely damaged. At this point, bladder replacement or enlargement surgery may become necessary to restore its structure and improve the patient's quality of life. Traditional bladder reconstruction surgery, specifically ileum augmentation cystoplasty, involves a complex surgical process, along with relatively large operative wounds and possible intestinal obstruction complications. There is also an urgent need for suitable bladder replacement materials. Over the past decade, synthetic bladder scaffolds have emerged as alternatives; however, most fail to simultaneously address the issues of postoperative fibrosis and bacterial infection. To overcome these challenges, we developed an electrospun poly(L-lactic acid)/gelatin (PLLA/GEL) microfiber membrane loaded with mitomycin C (MMC). By optimizing the PLLA/GEL ratio (7 : 3), the membrane exhibited both good rigidity and excellent elasticity. The MMC-loaded fibers demonstrated sustained drug release, effectively inhibiting E. coli and S. aureus in vitro. In rat partial cystectomy models, the PLLA/GEL/MMC group showed a reduction in collagen III deposition through inhibition of the TGF-β/Smad2 pathway, as well as enhanced urothelium regeneration and promotion of angiogenesis. Notably, the membrane maintained cell viability for both urothelial cells and smooth muscle cells over 7 days, confirming its biocompatibility. These findings highlight the promising potential of the PLLA/GEL/MMC microfiber membrane not only as a material for bladder tissue engineering but also as a tool for therapeutic intervention that addresses multiple facets of bladder healing and regeneration.
Collapse
Affiliation(s)
- Lunjie Zhao
- Department of Urology, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, China.
| | - Jianyou Xia
- Department of Urinary Surgery, General Hospital of Central Theater Command, Wuhan, 430012, China
| | - Congcong Yang
- Department of Urology, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, China.
| | - Jianping Tao
- Department of Urology, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, China.
| | - Shanfu Zhang
- Department of Urology, The First Affiliated Hospital of University of Science and Technology of China, Hefei, 230001, China
| | - Wentao Hu
- Department of Urology, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, China.
| | - Chen Zhang
- Department of Urology, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, China.
| | - Jiuwu Zhang
- Department of Urology, Huangshan City People's Hospital, Huangshan, 245000, China
| | - Qiang Li
- Human Anatomy Experimental Training Center, School of Basic Medical Sciences, Wannan Medical College, Wuhu, Anhui, 241002, China
| | - Yujie Xu
- Department of Urology, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, China.
| |
Collapse
|
70
|
Wang Z, Liu Y. Dual-modulation of nutrient-transporter axis and functionalized carriers: A paradigm shift for precision oral vitamin D delivery. Colloids Surf B Biointerfaces 2025; 253:114769. [PMID: 40344743 DOI: 10.1016/j.colsurfb.2025.114769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/25/2025] [Accepted: 05/05/2025] [Indexed: 05/11/2025]
Abstract
The transintestinal epithelial absorption of vitamin D is intricately regulated by specific transport protein networks. Emerging evidence from molecular nutrition research reveals that certain dietary nutrients can enhance intestinal vitamin D absorption through targeted modulation of lipid transport pathways. Despite significant advancements in vitamin D delivery systems demonstrating excellent intestinal mucoadhesion and in vitro bioaccessibility, their clinical translation remains limited by suboptimal in vivo bioavailability. To address this critical challenge, we propose an innovative synergistic nutrient absorption strategy that establishes precise coordination among three key elements: dietary nutrient composition, transport protein regulation, and intestinal absorption optimization. This comprehensive review systematically examines: (1) The molecular mechanisms governing transintestinal vitamin D transport and physiological modulation of protein-mediated absorption pathways; (2) The regulatory effects of dietary components on vitamin D absorption efficiency through protein pathway modulation, proposing a novel "nutrient-transporter-vitamin D axis" strategy integrating cutting-edge carrier technologies; (3) Future perspectives for developing functionalized vitamin D delivery systems. The proposed paradigm shift, combining nutrient-mediated transport enhancement with advanced carrier engineering, represents a transformative approach to overcome current limitations in oral vitamin D delivery. This dual-modulation strategy synergistically improves intestinal absorption and systemic bioavailability through simultaneous optimization of biological transport mechanisms and pharmaceutical delivery parameters, offering new possibilities for precision nutrition interventions.
Collapse
Affiliation(s)
- Zixiao Wang
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, PR China
| | - Yixiang Liu
- College of Ocean Food and Biological Engineering, Jimei University, Xiamen, Fujian 361021, PR China.
| |
Collapse
|
71
|
Huang W, Zong J, Li M, Li TF, Pan S, Xiao Z. Challenges and Opportunities: Nanomaterials in Epilepsy Diagnosis. ACS NANO 2025; 19:16224-16247. [PMID: 40266286 DOI: 10.1021/acsnano.5c01203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/24/2025]
Abstract
Epilepsy is a common neurological disorder characterized by a significant rate of disability. Accurate early diagnosis and precise localization of the epileptogenic zone are essential for timely intervention, seizure prevention, and personalized treatment. However, over 30% of patients with epilepsy exhibit negative results on electroencephalography and magnetic resonance imaging (MRI), which can lead to misdiagnosis and subsequent delays in treatment. Consequently, enhancing diagnostic methodologies is imperative for effective epilepsy management. The integration of nanomaterials with biomedicine has led to the development of diagnostic tools for epilepsy. Key advancements include nanomaterial-enhanced neural electrodes, contrast agents, and biochemical sensors. Nanomaterials improve the quality of electrophysiological signals and broaden the detection range of electrodes. In imaging, functionalized magnetic nanoparticles enhance MRI sensitivity, facilitating localization of the epileptogenic zone. NIR-II nanoprobes enable tracking of seizure-related biomarkers with deep tissue penetration. Furthermore, nanomaterials enhance the sensitivity of biochemical sensors for detecting epilepsy biomarkers, which is crucial for early detection. These advancements significantly increase diagnostic sensitivity and specificity. However, challenges remain, particularly regarding biosafety, quality control, and the scalability of fabrication processes. Overcoming these obstacles is essential for successful clinical translation. Artificial-intelligence-based big data analytics can facilitate the development of diagnostic tools by screening nanomaterials with specific properties. This approach may help to address current limitations and improve both effectiveness and safety. This review explores the application of nanomaterials in the diagnosis and detection of epilepsy, with the objective of inspiring innovative ideas and strategies to enhance diagnostic effectiveness.
Collapse
Affiliation(s)
- Wanbin Huang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jiabin Zong
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Ming Li
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Tong-Fei Li
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Songqing Pan
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zheman Xiao
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
72
|
Liu H, Li S, Yu X, Xu Q, Tang C, Yin C. Modulating the Protein Corona on Nanoparticles by Finely Tuning Cross-Linkers Improves Macrophage Targeting in Oral Small Interfering RNA Delivery. ACS NANO 2025; 19:16469-16487. [PMID: 40275505 DOI: 10.1021/acsnano.4c18033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
The protein corona (PC) plays an important role in regulating the in vivo fate of nanoparticles (NPs). Modulating the surface chemical properties of NPs to control PC formation provides an alternative impetus for the oral delivery of small interfering RNA (siRNA). Herein, using tripolyphosphate (TPP), hyaluronic acid, and poly-γ-glutamic acid as cross-linkers, three types of mannose-modified trimethyl chitosan-cysteine (MTC)-based NPs with distinct surface chemistries were prepared to encapsulate siRNA via ionic gelation. The MTC-based NPs that were cross-linked exclusively with TPP (MTC/TPP/siRNA NPs) exhibited greater thiol group accessibility on their surfaces, resulting in a stronger affinity for apolipoprotein (APO) B48 during translocation across intestinal epithelia. Moreover, intracellular transport of MTC/TPP/siRNA NPs via the endoplasmic reticulum and Golgi apparatus further increased adsorption of APOB48, a key component of chylomicrons, which follow a similar transport pathway. Benefiting from the elevated APOB48 levels within the PC, the orally delivered MTC/TPP/siRNA NPs showed higher uptake by hepatic macrophages and better therapeutic efficacy for acute liver injury. Our results elucidate the role of NP surface chemical characteristics and translocation mechanisms across intestinal epithelia in forming oral PC, providing valuable insights for designing NPs that achieve effective oral gene delivery by customizing PC formation in vivo.
Collapse
Affiliation(s)
- Hengqing Liu
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200438, China
- MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Shengqi Li
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200438, China
- MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Xin Yu
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200438, China
- MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Qian Xu
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200438, China
- MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Cui Tang
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200438, China
- MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Chunhua Yin
- State Key Laboratory of Genetic Engineering, Department of Pharmaceutical Sciences, School of Life Sciences, Fudan University, Shanghai 200438, China
- MOE Engineering Research Center of Gene Technology, School of Life Sciences, Fudan University, Shanghai 200438, China
| |
Collapse
|
73
|
Yin J, Sun W, Xiong H, Yao W, Liu X, Jiang H, Wang X. Photoactivated in-situ engineered-bacteria as an efficient H 2S generator to enhance photodynamic immunotherapy via remodeling the tumor microenvironment. Biomaterials 2025; 322:123388. [PMID: 40344882 DOI: 10.1016/j.biomaterials.2025.123388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Revised: 04/28/2025] [Accepted: 05/02/2025] [Indexed: 05/11/2025]
Abstract
Based on the unique biological advantages of bacteria and their derivatives, biosynthetic nanomaterials have been widely used in the field of tumor therapy. Although conventional bacterial treatments demonstrate potential in activating tumor immunity, their efficacy in inhibiting tumor growth remains constrained. In this study, a photoactivated hydrogen sulfide (H2S) generator was successfully prepared by in-situ engineering of bacteria, after Pt/MoS2 nanocomposites were in-situ generated by Escherichia coli (E. coli) and loaded with photosensitizer Ce6. This engineered-bacteria has been proved to have good tumor targeting ability and can enhance the effect of photodynamic therapy in the hypoxic tumor microenvironment. While reactive oxygen species (ROS) is effectively released, the fragmentation of bacteria can accelerate the release of abundant H2S, and promote tumor-specific H2S gas therapy, which can effectively remodel the tumor microenvironment and promote the activation of anti-tumor immunotherapy. This engineered bacteria not only improves the tumor specificity and effectiveness of H2S treatment, but also provides a new idea for nanomaterials in bacterial-mediated synergistic cancer treatment.
Collapse
Affiliation(s)
- Jiajia Yin
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Wenyu Sun
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Hongjie Xiong
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Wenyan Yao
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Xiaohui Liu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
| | - Hui Jiang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
| | - Xuemei Wang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China.
| |
Collapse
|
74
|
Li Z, Xu J, Lin H, Yu S, Sun J, Zhang C, Zhang S, Li T, Yang A, Lu W. Interleukin-15Rα-Sushi-Fc Fusion Protein Co-Hitchhikes Interleukin-15 and Pheophorbide A for Cancer Photoimmunotherapy. Pharmaceutics 2025; 17:615. [PMID: 40430906 PMCID: PMC12114846 DOI: 10.3390/pharmaceutics17050615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 04/20/2025] [Accepted: 04/26/2025] [Indexed: 05/29/2025] Open
Abstract
Background: Interleukin-15 (IL-15) stimulates the proliferation of natural killer cells or T cells, which, in combination with photodynamic therapy (PDT), has emerged as an effective strategy for cancer photoimmunotherapy. Instead of direct cytokine receptor activation, IL-15 necessitates first binding to the IL-15 receptor α chain subunit (IL-15Rα), followed by trans-presentation to the IL-15 receptor β/γ chain subunit on the effector cells for pharmacologic activation. Therefore, the delivery of IL-15 remains a major challenge owing to its short half-life, its lack of targeting activity, and the limited availability of IL-15Rα. Methods: A co-hitchhiking delivery approach using recombinant IL-15 (rIL-15) and a photosensitizer, pheophorbide A (PhA), is developed for enhanced combinatorial cancer immunotherapy with PDT. A recombinant IL-15Rα-sushi-Fc fusion protein (rILR-Fc) is designed to load rIL-15 through the IL-15Rα sushi domain, which mimics its trans-presentation. Moreover, the Fc moiety of rILR-Fc can load PhA based on its high binding affinity. Results: Through self-assembly, rILR-Fc/PhA/rIL-15 nanoparticles (NPs) are formulated to co-hitchhike PhA and rIL-15, which improves the tumor accumulation of PhA and rIL-15 through receptor-mediated transcytosis. Moreover, the nanoparticles prolong the blood half-life of rIL-15 but do not alter the elimination rate of PhA from the blood. The rILR-Fc/PhA/rIL-15 NPs effectively elicit potent systemic antitumor immunity and long-lasting immune memory against tumor rechallenge in model mice bearing orthotopic colon tumors. Conclusions: The enhanced antitumor therapeutic effect demonstrates that the co-hitchhiking delivery strategy, optimizing the pharmacokinetics of both the photosensitizer and IL-15, provides a promising strategy for combinatorial photodynamic and IL-15 immunotherapy.
Collapse
Affiliation(s)
- Zhe Li
- School of Pharmacy & Minhang Hospital, Key Laboratory of Smart Drug Delivery Ministry of Education & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Jiaojiao Xu
- School of Pharmacy & Minhang Hospital, Key Laboratory of Smart Drug Delivery Ministry of Education & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Hongzheng Lin
- School of Pharmacy & Minhang Hospital, Key Laboratory of Smart Drug Delivery Ministry of Education & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Sheng Yu
- School of Pharmacy & Minhang Hospital, Key Laboratory of Smart Drug Delivery Ministry of Education & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Jingwen Sun
- School of Pharmacy & Minhang Hospital, Key Laboratory of Smart Drug Delivery Ministry of Education & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Chen Zhang
- School of Pharmacy & Minhang Hospital, Key Laboratory of Smart Drug Delivery Ministry of Education & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Sihang Zhang
- School of Pharmacy & Minhang Hospital, Key Laboratory of Smart Drug Delivery Ministry of Education & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Tingting Li
- School of Pharmacy & Minhang Hospital, Key Laboratory of Smart Drug Delivery Ministry of Education & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Afeng Yang
- School of Pharmacy & Minhang Hospital, Key Laboratory of Smart Drug Delivery Ministry of Education & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
| | - Wei Lu
- School of Pharmacy & Minhang Hospital, Key Laboratory of Smart Drug Delivery Ministry of Education & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, 826 Zhangheng Road, Shanghai 201203, China
- Quzhou Fudan Institute, 108 Minjiang Avenue, Quzhou 324002, China
| |
Collapse
|
75
|
Froom ZSCS, Callaghan NI, Davenport Huyer L. Cellular crosstalk in fibrosis: insights into macrophage and fibroblast dynamics. J Biol Chem 2025:110203. [PMID: 40334985 DOI: 10.1016/j.jbc.2025.110203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/27/2025] [Accepted: 04/29/2025] [Indexed: 05/09/2025] Open
Abstract
Pathological fibrosis, the excessive deposition of extracellular matrix and tissue stiffening that causes progressive organ dysfunction, underlies diverse chronic diseases. The fibrotic microenvironment is driven by the dynamic microenvironmental interaction between various cell types; macrophages and fibroblasts play central roles in fibrotic disease initiation, maintenance, and progression. Macrophage functional plasticity to microenvironmental stimuli modulates fibroblast functionality by releasing pro-inflammatory cytokines, growth factors, and matrix remodeling enzymes that promote fibroblast proliferation, activation, and differentiation into myofibroblasts. Activated fibroblasts and myofibroblasts serve as the fibrotic effector cells, secreting extracellular matrix components and initiating microenvironmental contracture. Fibroblasts also modulate macrophage function through the release of their own pro-inflammatory cytokines and growth factors, creating bidirectional crosstalk that reinforces the chronic fibrotic cycle. The intricate interplay between macrophages and fibroblasts, including their secretomes and signaling interactions, leads to tissue damage and pathological loss of tissue function. In this review, we examine macrophage-fibroblast reciprocal dynamic interactions in pathological fibrotic conditions. We discuss the specific lineages and functionality of macrophages and fibroblasts implicated in fibrotic progression, with focus on their signal transduction pathways and secretory signalling that enables their pro-fibrotic behaviour. We then finish with a set of recommendations for future experimentation with the goal of developing a set of potential targets for anti-fibrotic therapeutic candidates. Understanding the cellular interactions between macrophages and fibroblasts provides valuable insights into potential therapeutic strategies to mitigate fibrotic disease progression.
Collapse
Affiliation(s)
- Zachary S C S Froom
- School of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Neal I Callaghan
- Department of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Locke Davenport Huyer
- School of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, NS B3H 4R2, Canada; Department of Microbiology & Immunology, Faculty of Medicine, Dalhousie University, Halifax, NS B3H 4R2, Canada; Department of Biomaterials & Applied Oral Sciences, Faculty of Dentistry, Dalhousie University, Halifax, NS B3H 4R2, Canada; Nova Scotia Health, Halifax, NS B3S 0H6, Canada.
| |
Collapse
|
76
|
Shi S, Ou X, Wang Q, Zhang L. Macrophage-Derived Extracellular Vesicles: A Novel Therapeutic Alternative for Diabetic Wound. Int J Nanomedicine 2025; 20:5763-5777. [PMID: 40343196 PMCID: PMC12060905 DOI: 10.2147/ijn.s518655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 04/25/2025] [Indexed: 05/11/2025] Open
Abstract
Diabetic wounds represent a significant clinical and economic challenge owing to their chronicity and susceptibility to complications. Dysregulated macrophage function is a key factor in delayed wound healing. Recent studies have emphasized the therapeutic potential of macrophage-derived extracellular vesicles (MDEVs), which are enriched with bioactive molecules such as proteins, lipids, and nucleic acids that mirror the state of their parent cells. MDEVs influence immune modulation, angiogenesis, extracellular matrix remodeling, and intercellular communication. In this review, we summarize and discuss the biological properties and therapeutic mechanisms of MDEVs in diabetic wound healing, highlighting strategies to enhance their efficacy through bioengineering and advanced delivery systems. We also explore the integration of MDEVs into innovative wound care technologies. Addressing current limitations and advancing clinical translation of MDEVs could advance diabetic wound management, offering a precise, effective, and versatile therapeutic option.
Collapse
Affiliation(s)
- Shaoyan Shi
- Department of Hand Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an Honghui Hospital North District, Xi’an, Shaanxi, 710000, People’s Republic of China
| | - Xuehai Ou
- Department of Hand Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an Honghui Hospital North District, Xi’an, Shaanxi, 710000, People’s Republic of China
| | - Qian Wang
- Department of Hand Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an Honghui Hospital North District, Xi’an, Shaanxi, 710000, People’s Republic of China
| | - Li Zhang
- Department of Hand Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an Honghui Hospital North District, Xi’an, Shaanxi, 710000, People’s Republic of China
| |
Collapse
|
77
|
Zhong R, Xu X, Tutoni G, Liu M, Yang K, Li K, Jin K, Chen Y, Mai JDH, Becker ML, Huang TJ. An acoustofluidic embedding platform for rapid multiphase microparticle injection. Nat Commun 2025; 16:4144. [PMID: 40319024 PMCID: PMC12049528 DOI: 10.1038/s41467-025-59146-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Accepted: 04/09/2025] [Indexed: 05/07/2025] Open
Abstract
Droplet manipulation technologies play a critical role in many aspects of biochemical research, including in complex reaction assays useful for drug delivery, for building artificial cells, and in synthetic biology. While advancements have been made in manipulating liquid droplets, the capability to freely and dynamically manipulate solid objects across aqueous and oil phases remains unexplored. Here, we develop an acoustofluidic frequency-associated microsphere embedding platform, which enables microscale rapid injection of microparticles from a fluorinated oil into aqueous droplets. By observing different embedding mechanisms at low and high acoustic frequencies, we establish a theoretical model and practical principles for cross-phase manipulations. The proposed system not only enables multi-phase manipulation but also provides contactless control of specific microparticles within various distinctive phases. We demonstrate the acoustic-driven embedding and subsequent on-demand disassembly of hydrogel microspheres. This system indicates potential for reagent delivery and molecule capture applications. It enhances existing droplet manipulation technologies by enabling both multi-phase and cross-phase operations, paving the way for solid-liquid interaction studies in artificial cell research. The capability for intricate multi-phase loading, transport, and reactions offers promising implications for various fields, including in-droplet biochemical assays, drug delivery, and synthetic biology.
Collapse
Grants
- R01GM141055 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R01 GM143439 NIGMS NIH HHS
- R01GM145960 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R44GM154514 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R01 GM144417 NIGMS NIH HHS
- R44AG063643 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R44OD024963 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R44GM154515 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R01 GM141055 NIGMS NIH HHS
- R44 AG063643 NIA NIH HHS
- R44 GM154515 NIGMS NIH HHS
- R01GM144417 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R01 GM145960 NIGMS NIH HHS
- R01 AG084098 NIA NIH HHS
- CMMI-2104295 National Science Foundation (NSF)
- R44 GM154514 NIGMS NIH HHS
- R44 OD024963 NIH HHS
- R01 HD103727 NICHD NIH HHS
- R01AG084098 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R01HD103727 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- R01GM143439 Foundation for the National Institutes of Health (Foundation for the National Institutes of Health, Inc.)
- National Science Foundation Graduate Research Fellowship, Grant DGE 2139754
Collapse
Affiliation(s)
- Ruoyu Zhong
- Thomas Lord Department of Mechanical Engineering and Material Science, Duke University, Durham, NC, USA
| | - Xianchen Xu
- Thomas Lord Department of Mechanical Engineering and Material Science, Duke University, Durham, NC, USA
| | - Gianna Tutoni
- Department of Chemistry, Duke University, Durham, NC, USA
| | - Mingyuan Liu
- Thomas Lord Department of Mechanical Engineering and Material Science, Duke University, Durham, NC, USA
- Department of Electrical and Computer Engineering, Duke University, Durham, NC, USA
| | - Kaichun Yang
- Thomas Lord Department of Mechanical Engineering and Material Science, Duke University, Durham, NC, USA
| | - Ke Li
- Thomas Lord Department of Mechanical Engineering and Material Science, Duke University, Durham, NC, USA
| | - Ke Jin
- Thomas Lord Department of Mechanical Engineering and Material Science, Duke University, Durham, NC, USA
| | - Ying Chen
- Thomas Lord Department of Mechanical Engineering and Material Science, Duke University, Durham, NC, USA
| | - John D H Mai
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Matthew L Becker
- Thomas Lord Department of Mechanical Engineering and Material Science, Duke University, Durham, NC, USA
- Department of Chemistry, Duke University, Durham, NC, USA
| | - Tony Jun Huang
- Thomas Lord Department of Mechanical Engineering and Material Science, Duke University, Durham, NC, USA.
| |
Collapse
|
78
|
Wang R, Wei X, He X, Wang L, Zhou M, Tang J, Che X, Zhou G, Liu H. ML228-loaded nanoparticles with platelet membrane coating promote endothelialization of vascular grafts by enhancing HIF-1α expression. Colloids Surf B Biointerfaces 2025; 253:114756. [PMID: 40334473 DOI: 10.1016/j.colsurfb.2025.114756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 04/28/2025] [Accepted: 04/29/2025] [Indexed: 05/09/2025]
Abstract
Small-diameter vascular grafts (SDVGs) often struggle to maintain long-term patency due to thrombus formation, intimal hyperplasia, and inflammation. Endothelialization emerges as a pivotal strategy for addressing these concerns. As a representative activator of the hypoxia-inducible factor (HIF) pathway, ML228 can stimulate the expression of downstream target genes like vascular endothelial growth factor (VEGF) to induce angiogenesis, yet it requires encapsulation by nanoparticles for optimal delivery and efficacy. However, the immune system often recognizes nanoparticles as foreign entities, posing a significant risk of clearance. In this study, we developed ML228-loaded poly (lactic-co-glycolic acid) (PLGA) nanoparticles and coated them with platelet membranes, thereby enhancing their biocompatibility and enabling immune escape. The ML228-loaded PLGA nanoparticles coated with platelet membranes (MPNP) were immobilized onto electrospinning SDVGs made of silk fibroin (SF) and polycaprolactone (PCL) to obtain MPNP-coated grafts (SF/PCL@MPNP) with the ability to promote endothelialization. In vitro biological activity studies demonstrated that SF/PCL@MPNP activated the HIF pathway, upregulating the downstream target gene VEGF, which facilitated endothelial cells migration and angiogenesis. In vivo implantation in a rat abdominal aorta model revealed that SF/PCL@MPNP promoted endothelialization, supported the regeneration of contractile smooth muscle cells, and modulated inflammatory responses. Overall, this study presents a strategy for constructing SDVGs using ML228-loaded nanoparticles with platelet membrane coating, highlighting the promises of using ML228 to activate the HIF pathway and membrane-coated nanoparticles to improve endothelialization in vascular graft applications.
Collapse
Affiliation(s)
- Ruichen Wang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education; Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering); School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Xinbo Wei
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education; Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering); School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Xi He
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education; Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering); School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Li Wang
- Nanjing Stomatological Hospital, Affiliated Hospital of Medical School, Nanjing University, 30 Zhongyang Road, Nanjing, Jiangsu 210008, P. R. China
| | - Moyan Zhou
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education; Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering); School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Jiarui Tang
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education; Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering); School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Xin Che
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education; Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering); School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Gang Zhou
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education; Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering); School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China
| | - Haifeng Liu
- Key Laboratory of Biomechanics and Mechanobiology (Beihang University), Ministry of Education; Key Laboratory of Innovation and Transformation of Advanced Medical Devices, Ministry of Industry and Information Technology; National Medical Innovation Platform for Industry-Education Integration in Advanced Medical Devices (Interdiscipline of Medicine and Engineering); School of Biological Science and Medical Engineering, Beihang University, Beijing, 100191, China.
| |
Collapse
|
79
|
Sanz-Velasco A, Patrian M, Nieddu M, Shen B, Fuenzalida Werner JP, Kostiainen MA, Costa RD, Anaya-Plaza E. Fusing fluorescent proteins and ferritin for protein cage based lighting devices. NANOSCALE 2025; 17:10793-10800. [PMID: 40184033 PMCID: PMC11970472 DOI: 10.1039/d4nr05261g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 03/20/2025] [Indexed: 04/05/2025]
Abstract
Ferritin cages are an effective platform to encapsulate and stabilize a range of active cargoes and present a promising stepping stone towards a wide range of applications. They have been explored for optoelectronic applications in combination with fluorescent proteins towards bio-hybrid light-emitting diodes (Bio-HLEDs) only recently. However, protein integration within the cage or coassembled ferritin cages relies on electrostatic interactions and requires the supercharging of the fluorescent protein that easily compromises functionality and stability. To address this limitation, we have developed a fusion protein combining the Thermotoga maritima apoferritin (TmaFt) with a green fluorescent protein named mGreenlantern (mGL). This approach avoids jeopardizing both the cage assembly capability of TmaFt and the photophysical features of mGL. After optimizing the fusion protein mGL-TmaFt with respect to the linker length, assembling efficiency, and mGL payload into the cage (mGL@TmaFt), our findings reveal that they exhibited enhanced thermal and structural stabilities in both solution and when embedded into a polymer matrix. This enables effective mGL shielding, reducing H-transfer deactivation of the chromophore and water-assisted heat transfer across the polymer network. Indeed, the photo-induced heat generation in Bio-HLEDs operating at high currents was significantly reduced, resulting in a 30- and 15-fold higher device stability compared to references with either mGL or mGL-TmaFt proteins, respectively. Overall, this work sets in the potential of protein cage design for photon manipulation in protein lighting devices.
Collapse
Affiliation(s)
- Alba Sanz-Velasco
- Department of Bioproducts and Biosystems, Aalto University, 02150 Espoo, Finland.
| | - Marta Patrian
- Technical University of Munich, Campus Straubing for Biotechnology and Sustainability, Chair of Biogenic Functional Materials, Schulgasse 22, 94377, Straubing, Germany.
| | - Mattia Nieddu
- Technical University of Munich, Campus Straubing for Biotechnology and Sustainability, Chair of Biogenic Functional Materials, Schulgasse 22, 94377, Straubing, Germany.
| | - Boxuan Shen
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Juan Pablo Fuenzalida Werner
- Technical University of Munich, Campus Straubing for Biotechnology and Sustainability, Chair of Biogenic Functional Materials, Schulgasse 22, 94377, Straubing, Germany.
| | - Mauri A Kostiainen
- Department of Bioproducts and Biosystems, Aalto University, 02150 Espoo, Finland.
| | - Rubén D Costa
- Technical University of Munich, Campus Straubing for Biotechnology and Sustainability, Chair of Biogenic Functional Materials, Schulgasse 22, 94377, Straubing, Germany.
| | - Eduardo Anaya-Plaza
- Department of Bioproducts and Biosystems, Aalto University, 02150 Espoo, Finland.
| |
Collapse
|
80
|
Liu X, Zhao Y, Wu X, Zhou Y, Liu Y, Wang S, Zhang Y, Yang H, Song F, Huang C. Spatiotemporally Programming Microenvironment to Recapitulate Endochondral Ossification via Greenhouse-Inspired Bionic Niche. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2504057. [PMID: 40317581 DOI: 10.1002/adma.202504057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 03/31/2025] [Indexed: 05/07/2025]
Abstract
Various biomaterials have been developed to address challenging critical-sized bone defects. However, most of them focus on intramembranous ossification (IMO) rather than endochondral ossification (ECO), often resulting in suboptimal therapeutic outcomes. Drawing inspiration from the functionality of the greenhouse ecosystem, herein a bionic niche is innovatively crafted to recapitulate the ECO process. This niche consists of three hierarchical components: an embedded microchannel network that facilitates cell infiltration and matter exchange, a polydopamine surface modification layer with immunomodulatory functions, and an ECO-targeted delivery system based on mesoporous silica nanoparticles. Through spatiotemporally programming of the microenvironment, the bionic niche effectively recapitulates the key stages of ECO. Notably, even in the rat calvaria, a region well-known for IMO, the bionic niche is capable of initiating ECO, evident by cartilage template formation, leading to efficient bone regeneration. Taken together, this study introduces prospective concepts for designing next-generation ECO-driven biomaterials for bone tissue engineering.
Collapse
Affiliation(s)
- Xuzheng Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Yaning Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Xiaoyi Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Yueli Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Yingheng Liu
- Dental Materials Science, Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong
| | - Shilei Wang
- Key Laboratory of Resources and Compound of Traditional Chinese Medicine, Ministry of Education, Hubei University of Traditional Chinese Medicine, Wuhan, 430065, China
| | - Yufeng Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Hongye Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Fangfang Song
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Cui Huang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| |
Collapse
|
81
|
Hou Z, Wang K, Liu G, Yuan Z, Peng H, Yuan Y, Wei H, Wang T, Li P. Nitric Oxide-Mediated Dual-Functional Smart Titanium Implant Coating for Antibacterial and Osseointegration Promotion in Implant-Associated Infections. Adv Healthc Mater 2025; 14:e2500997. [PMID: 40195820 DOI: 10.1002/adhm.202500997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2025] [Revised: 03/22/2025] [Indexed: 04/09/2025]
Abstract
The balance of antibacterial and osseointegration-promoting properties on titanium (Ti) implants in a simple and efficient manner is crucial for the management of implant-associated infections, a condition that has become a significant global health concern. Herein, a nitric oxide (NO)-mediated dual-function smart coating with antibacterial and osseointegration-promoting properties is developed for Ti implants. The coating leverages the distinct properties of NO at high and low concentrations to enable an on-demand functional switch. Specifically, antibacterial function is achieved through a rapid release of high-dose NO in response to the infection microenvironment and near-infrared stimulation. Once the infection is resolved and normal physiological conditions are restored, the coating gradually releases low-dose NO to promote osseointegration. In vitro tests confirm that the coating exhibits antibacterial ratio of 97.84% and 97.18% against methicillin-resistant Staphylococcus aureus and its biofilms, respectively, and demonstrates the ability to activate osteoblasts. The rat femoral implant-associated infection model further certifies that the responsive NO release mechanism of the coating efficiently facilitates the on-demand functional switch between antibacterial and osseointegration-promoting properties. Notably, the use of the dual-functional nonantibiotic agent, NO, significantly mitigates the risk of bacterial resistance.
Collapse
Affiliation(s)
- Zishuo Hou
- State Key Laboratory of Flexible Electronics (LOFE) & Institute of Flexible Electronics (IFE), Shaanxi Key Laboratory of Flexible Electronics & MIIT Key Laboratory of Flexible Electronics (KLoFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), Xi'an, Shaanxi, 710072, P. R. China
| | - Kun Wang
- State Key Laboratory of Flexible Electronics (LOFE) & Institute of Flexible Electronics (IFE), Shaanxi Key Laboratory of Flexible Electronics & MIIT Key Laboratory of Flexible Electronics (KLoFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), Xi'an, Shaanxi, 710072, P. R. China
| | - Guming Liu
- State Key Laboratory of Flexible Electronics (LOFE) & Institute of Flexible Electronics (IFE), Shaanxi Key Laboratory of Flexible Electronics & MIIT Key Laboratory of Flexible Electronics (KLoFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), Xi'an, Shaanxi, 710072, P. R. China
| | - Zhang Yuan
- Chongqing Innovation Center, Northwestern Polytechnical University, Chongqing, 401135, P. R. China
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an, Shaanxi, 710072, P. R. China
| | - Haowei Peng
- State Key Laboratory of Flexible Electronics (LOFE) & Institute of Flexible Electronics (IFE), Shaanxi Key Laboratory of Flexible Electronics & MIIT Key Laboratory of Flexible Electronics (KLoFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), Xi'an, Shaanxi, 710072, P. R. China
| | - Yue Yuan
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Hongbo Wei
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Tengjiao Wang
- State Key Laboratory of Flexible Electronics (LOFE) & Institute of Flexible Electronics (IFE), Shaanxi Key Laboratory of Flexible Electronics & MIIT Key Laboratory of Flexible Electronics (KLoFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), Xi'an, Shaanxi, 710072, P. R. China
- Chongqing Innovation Center, Northwestern Polytechnical University, Chongqing, 401135, P. R. China
| | - Peng Li
- State Key Laboratory of Flexible Electronics (LOFE) & Institute of Flexible Electronics (IFE), Shaanxi Key Laboratory of Flexible Electronics & MIIT Key Laboratory of Flexible Electronics (KLoFE), Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), Xi'an, Shaanxi, 710072, P. R. China
| |
Collapse
|
82
|
Li Y, Wang W, Wang W, Zhang X, Chen J, Gao H. Unveiling Structural Heterogeneity and Evolutionary Adaptations of Heteromultimeric Bacterioferritin Nanocages. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409957. [PMID: 40167232 PMCID: PMC12120770 DOI: 10.1002/advs.202409957] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 02/24/2025] [Indexed: 04/02/2025]
Abstract
Iron-storage bacterioferritins (Bfrs), existing in either homo- or hetero-multimeric form, play a crucial role in iron homeostasis. While the structure and function of homo-multimeric bacterioferritins (homo-Bfrs) have been extensively studied, little is known about the assembly, distinctive characteristics, or evolutionary adaptations of hetero-multimeric bacterioferritins (hetero-Bfrs). Here, the cryo-EM structure and functional characterization of a bacterial hetero-Bfr (SoBfr12) are reported. Compared to homo-Bfrs, although SoBfr12 exhibits a conserved spherical cage-like dodecahedron, its pores through which ions traverse exhibit substantially increased diversity. Importantly, the heterogeneity has significant impacts on sites for ion entry, iron oxidation, and reduction. Moreover, evolutionary analyses reveal that hetero-Bfrs may represent a new class within the Bfr subfamily, consisting of two different types that may have evolved from homo-Bfr through tandem duplication and directly from ferritin (Ftn) via dispersed duplication, respectively. These results reveal remarkable structural and functional features of a hetero-Bfr, enabling the rational design of nanocages for enhanced iron-storing efficiency and for other specific purposes, such as drug delivery vehicles and nanozymes.
Collapse
Affiliation(s)
- Yingxi Li
- State Key Laboratory for Vegetation StructureFunction and Construction (VegLab)Institute of Microbiology and College of Life SciencesZhejiang UniversityHangzhou310058China
| | - Weiwei Wang
- State Key Laboratory for Vegetation StructureFunction and Construction (VegLab)Institute of Microbiology and College of Life SciencesZhejiang UniversityHangzhou310058China
| | - Wei Wang
- State Key Laboratory for Vegetation StructureFunction and Construction (VegLab)Institute of Microbiology and College of Life SciencesZhejiang UniversityHangzhou310058China
| | - Xing Zhang
- Center of Cryo Electron MicroscopyZhejiang University School of MedicineHangzhou310058China
| | - Jinghua Chen
- State Key Laboratory for Vegetation StructureFunction and Construction (VegLab)Institute of Microbiology and College of Life SciencesZhejiang UniversityHangzhou310058China
| | - Haichun Gao
- State Key Laboratory for Vegetation StructureFunction and Construction (VegLab)Institute of Microbiology and College of Life SciencesZhejiang UniversityHangzhou310058China
| |
Collapse
|
83
|
Sen S, Kumar R, Tomar RS, Roy S. Designing Short Cardin-Motif Peptide and Biopolymer-Based Multicomponent Hydrogels for Developing Advanced Composite Scaffolds for Improving Cellular Behavior. Macromol Biosci 2025; 25:e2400555. [PMID: 39838741 DOI: 10.1002/mabi.202400555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 01/01/2025] [Indexed: 01/23/2025]
Abstract
Multicomponent self-assembly represents a cutting-edge strategy in peptide nanotechnology, enabling the creation of nanomaterials with enhanced physical and biological characteristics. This approach draws inspiration from the highly complex nature of the native extracellular matrix (ECM) constituting multicomponent biomolecular entities. In recent years, the combination of bioactive peptide with polymer has gained significant attention for the fabrication of novel biomaterials due to their inherent specificity, tunable physiochemical properties, biocompatibility, and biodegradability. This advanced strategy can address the limitation of the lower mechanical strength of the individual peptide hydrogel by incorporating the biopolymer, resulting in the formation of a composite scaffold. In this direction, this advanced strategy is explored using noncovalent interactions between cellulose nano-fiber (CNF) and cationic Cardin-motif peptide to develop advanced composite scaffolds. The bioactive cationic peptide otherwise failed to form hydrogel at physiological conditions. Interestingly, the differential mixing ratio of CNF and peptide modulated the surface charge, functionality, and mechanical properties of the composite scaffolds, resulting in diverse cellular responses. 10:1 (w/w) ratio of CNF and peptide-based composite scaffold demonstrates improved cellular survival and proliferation in 2D culture conditions. Notably, in 3D cultures, cell proliferation on the 10:1 matrix is comparable to Matrigel, highlighting its potential for advanced tissue engineering applications.
Collapse
Affiliation(s)
- Sourav Sen
- Institute of Nano Science and Technology (INST), Sector 81, Knowledge City, Mohali, Punjab, 140306, India
| | - Rakesh Kumar
- Institute of Nano Science and Technology (INST), Sector 81, Knowledge City, Mohali, Punjab, 140306, India
| | - Rahul Singh Tomar
- Institute of Nano Science and Technology (INST), Sector 81, Knowledge City, Mohali, Punjab, 140306, India
| | - Sangita Roy
- Institute of Nano Science and Technology (INST), Sector 81, Knowledge City, Mohali, Punjab, 140306, India
| |
Collapse
|
84
|
Li X, Zhang X, Wang Y, Ji S, Zhao Z, Yin J, Yang T, Feng X, Chen H, Li W, Wang X, Jing C, Ding D, Zhao L. Preparation and Evaluation of RGD-Conjugated Crosslinked PVA Tissue Engineered Vascular Scaffold with Endothelial Differentiation and Its Impact on Vascular Regeneration In Vivo. Macromol Biosci 2025; 25:e2400554. [PMID: 39985427 DOI: 10.1002/mabi.202400554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 02/03/2025] [Indexed: 02/24/2025]
Abstract
PVA has emerged as a prevalent material for the construction of vascular tissue engineering scaffolds. Nonetheless, the integration of 3D crosslinked polyvinyl alcohol (PVA) scaffolds featuring arginine-glycine-aspartate (RGD) binding remains a rarity in tissue engineering. In the present study, a PVA-4-azidobenzoic acid (AZ)-RGD scaffold is prepared based on cross-linking of two distinct PVA derivatives: one featuring photoreactive azides for ultraviolet (UV)-crosslinking and the other incorporating RGD peptides. The results show that the PVA-AZ-RGD scaffold has good blood compatibility and biomechanical properties, with hydrophilic properties, and a hydrolysis rate of 27.31% at 12 weeks. Notably, the incorporation of RGD peptides significantly bolsters the attachment and proliferation of mesenchymal stem cells (MSCs) on the scaffolds, compared to non-RGD-conjugated controls. Furthermore, RGD conjugation markedly accelerates endothelialization of MSCs following 15 days of endothelial culture. Post-transplantation, the PVA-AZ-RGD scaffold exhibits favorable blood flow patency, minimal immune rejection, promotes endothelialization and smooth muscle cell proliferation, and facilitates the development of extracellular matrix, ultimately contributing to the formation of regenerative artificial blood vessels. These comprehensive findings underscore the promising potential of RGD-integrated, crosslinked PVA scaffolds for applications in vascular tissue engineering.
Collapse
Affiliation(s)
- Xiafei Li
- College of Medical Engineering, Xinxiang Medical University, Xinxiang, 453003, China
| | - Xuewei Zhang
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
| | - Yameng Wang
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
| | - Shenglu Ji
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
| | - Ziwei Zhao
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
| | - Jianshen Yin
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
| | - Tuo Yang
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
| | - Xin Feng
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
- Huaihe hospital, Henan University, Kaifeng, 475004, China
| | - Hongli Chen
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
| | - Wenbin Li
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
| | - Xianwei Wang
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, 453003, China
| | - Changqin Jing
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
| | - Dan Ding
- Frontiers Science Center for Cell Responses, State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials Ministry of Education, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Liang Zhao
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, 453003, China
| |
Collapse
|
85
|
Wu JLY, Ji Q, Blackadar C, Nguyen LNM, Lin ZP, Sepahi Z, Stordy BP, Granda Farias A, Sindhwani S, Ngo W, Chan K, Habsid A, Moffat J, Chan WCW. The pathways for nanoparticle transport across tumour endothelium. NATURE NANOTECHNOLOGY 2025; 20:672-682. [PMID: 40097646 DOI: 10.1038/s41565-025-01877-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 01/30/2025] [Indexed: 03/19/2025]
Abstract
The active transport and retention principle is an alternative mechanism to the enhanced permeability and retention effect for explaining nanoparticle tumour delivery. It postulates that nanoparticles actively transport across tumour endothelial cells instead of passively moving through gaps between these cells. How nanoparticles transport across tumour endothelial cells remains unknown. Here we show that nanoparticles cross tumour endothelial cells predominantly using the non-receptor-based macropinocytosis pathway. We discovered that tumour endothelial cell membrane ruffles capture circulating nanoparticles, internalize them in intracellular vesicles and release them into the tumour interstitium. Tumour endothelial cells have a higher membrane ruffle density than healthy endothelium, which may partially explain the elevated nanoparticle tumour accumulation. Receptor-based endocytosis pathways such as clathrin-mediated endocytosis contribute to nanoparticle transport to a lesser extent. Nanoparticle size determines the degree of contribution for each pathway. Elucidating the nanoparticle transport mechanism is crucial for developing strategies to control nanoparticle tumour delivery.
Collapse
Affiliation(s)
- Jamie L Y Wu
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Qin Ji
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Colin Blackadar
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Luan N M Nguyen
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
- David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Zachary P Lin
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Zahra Sepahi
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Benjamin P Stordy
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
| | - Adrian Granda Farias
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Shrey Sindhwani
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
- Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Adult Hematology and Medical Oncology Fellowship Program, Section of Hematology/Oncology, University of Chicago, Chicago, IL, USA
| | - Wayne Ngo
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, CA, USA
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA, USA
| | - Katherine Chan
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Andrea Habsid
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Jason Moffat
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Genetics and Genome Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Warren C W Chan
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada.
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario, Canada.
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada.
- School of Chemistry, Chemical Engineering, and Biotechnology, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
86
|
Lu M, Lu P, Liu S, Peng Y, Yang Z, Chu Z, Hong Z, Yang Z, Hu Y. Chitosan/acrylic rosin-based superhydrophobic coatings inspired by Pickering emulsion template and lotus leaf surface structure for paper-based food packaging. Int J Biol Macromol 2025; 308:142375. [PMID: 40120890 DOI: 10.1016/j.ijbiomac.2025.142375] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 03/05/2025] [Accepted: 03/19/2025] [Indexed: 03/25/2025]
Abstract
The superhydrophobic coated papers have attracted much attention as the promising alternatives for disposable plastic packaging materials in food packaging fields. Despite recent visible progress in constructing superhydrophobic coated papers, it is still a challenge to realize superhydrophobicity, high oil-resistant behavior, biodegradability and recyclability at the same time in a coated paper. Herein, we have effectively constructed the superhydrophobic coated (PE&SP-CP) paper with the imitated lotus leaf surface structures by firstly dip-coating cellulose nanocrystals stabilized Pickering emulsion containing chitosan in water phase and acrylic rosin (AAR) in oil phase, and then spraying ethanol suspension of hydrophobic nano-silica (hSiO2) and polydimethylsiloxane (PDMS). In this aspect, the micro-nano hierarchical surface of chitosan-based coating film consists of AAR microcapsules and hSiO2 nanoparticles, as well as covered a hydrophobic PDMS polymer layer. And the prepared PE&SP-CP paper with surface superhydrophobicity demonstrates the highly water barrier performance with the water contact angle of 155.8° and water vapor permeability rate of 1.42 × 10-10 gm-1s-1Pa-1, the favorable oil repellency with the kit rating of 9/12, the clearly improved tensile strengths of 21.67 MPa. Interestingly, the PE&SP-CP paper also shows superior antifouling performance, self-cleaning ability, biodegradability of soil burial and recyclability. This work provides a universal approach for the effective construction of oil-resistant, self-cleaning, antifouling, recyclable, biodegradable superhydrophobic coated paper with the imitated lotus leaf surface structure, which has great potential application in the food packaging industry.
Collapse
Affiliation(s)
- Manzhi Lu
- Key Laboratory for Bio-based Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China; State Key Laboratory of Pulp and Paper Engineering, South China University of Technology, Guangzhou 510640, China
| | - Pan Lu
- Key Laboratory for Bio-based Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
| | - Song Liu
- Key Laboratory for Bio-based Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
| | - Yingying Peng
- Key Laboratory for Bio-based Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
| | - Ziming Yang
- Key Laboratory of Tropical Fruit Biology, Ministry of Agriculture & Rural Affairs, South Subtropical Crops Research Institute, Chinese Academy of Tropical Agricultural Sciences, Zhanjiang 524091, China.
| | - Zhuangzhuang Chu
- Key Laboratory for Bio-based Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
| | - Ziyin Hong
- Key Laboratory for Bio-based Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China
| | - Zhuohong Yang
- Key Laboratory for Bio-based Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China.
| | - Yang Hu
- Key Laboratory for Bio-based Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou 510642, China; State Key Laboratory of Pulp and Paper Engineering, South China University of Technology, Guangzhou 510640, China.
| |
Collapse
|
87
|
Gong G, Wan W, Zhang X, Chen X, Yin J. Management of ROS and Regulatory Cell Death in Myocardial Ischemia-Reperfusion Injury. Mol Biotechnol 2025; 67:1765-1783. [PMID: 38852121 DOI: 10.1007/s12033-024-01173-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 04/02/2024] [Indexed: 06/10/2024]
Abstract
Myocardial ischemia-reperfusion injury (MIRI) is fatal to patients, leading to cardiomyocyte death and myocardial remodeling. Reactive oxygen species (ROS) and oxidative stress play important roles in MIRI. There is a complex crosstalk between ROS and regulatory cell deaths (RCD) in cardiomyocytes, such as apoptosis, pyroptosis, autophagy, and ferroptosis. ROS is a double-edged sword. A reasonable level of ROS maintains the normal physiological activity of myocardial cells. However, during myocardial ischemia-reperfusion, excessive ROS generation accelerates myocardial damage through a variety of biological pathways. ROS regulates cardiomyocyte RCD through various molecular mechanisms. Targeting the removal of excess ROS has been considered an effective way to reverse myocardial damage. Many studies have applied antioxidant drugs or new advanced materials to reduce ROS levels to alleviate MIRI. Although the road from laboratory to clinic has been difficult, many scholars still persevere. This article reviews the molecular mechanisms of ROS inhibition to regulate cardiomyocyte RCD, with a view to providing new insights into prevention and treatment strategies for MIRI.
Collapse
Affiliation(s)
- Ge Gong
- Department of Geriatrics, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 211002, China
| | - Wenhui Wan
- Department of Geriatrics, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 211002, China
| | - Xinghu Zhang
- Department of Geriatrics, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 211002, China
| | - Xiangxuan Chen
- Department of Cardiology, the Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, 211100, China.
| | - Jian Yin
- Department of Orthopedics, the Affiliated Jiangning Hospital with Nanjing Medical University, Nanjing, 211100, China.
- Department of Orthopedics, Jiangning Clinical Medical College of Jiangsu Medical Vocational College, Nanjing, 211100, China.
- Department of Orthopedics, Jiangning Clinical Medical College of Nanjing Medical University Kangda College, Nanjing, 211100, China.
| |
Collapse
|
88
|
Li L, Tian Y, Liu Y, Teng F, Li Y, Geng M. Insights into the co-loading capacity of Pickering W/O/W systems for different types of soy protein isolated-pectin particles: Affected by biopolymer complexation sequence. Int J Biol Macromol 2025; 308:142593. [PMID: 40157683 DOI: 10.1016/j.ijbiomac.2025.142593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 03/20/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
This study developed a Pickering water-in-oil-in-water (W/O/W) emulsion (PWE) for co-delivering vitamins C and E using soy protein isolate (SPI) and pectin (PEC) particles as emulsifiers. Two particle types were synthesized via distinct nucleation mechanisms: (1) heat-treated SPI-PEC complexes formed at pH 4.0 and (2) SPI-PEC complexes formed through pH regulation followed by heating. Type 2 particles exhibited smaller sizes, more regular spherical structures, and stronger intermolecular interactions than type 1. Higher PEC concentrations further enhanced particle compactness. Additionally, type 2 particles demonstrated superior interfacial protein adsorption, forming stable and elastic interfacial membranes in the PWE. As a result, PWEs stabilized with type 2 particles achieved controlled release and high bioaccessibility of vitamins C (90.18 %) and E (85.24 %). These findings provide insights into the development of heat-induced biopolymer particles as effective PWE stabilizers.
Collapse
Affiliation(s)
- Lijia Li
- College of Food Science, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Yang Tian
- College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China; Puer University, Puer 665000, Chinae Pu'er University, Pu'er 665000, Yunnan, People's Republic of China
| | - Yue Liu
- College of Food Science, Northeast Agricultural University, Harbin, Heilongjiang 150030, China
| | - Fei Teng
- College of Food Science, Northeast Agricultural University, Harbin, Heilongjiang 150030, China.
| | - Yang Li
- College of Food Science, Northeast Agricultural University, Harbin, Heilongjiang 150030, China.
| | - Mengjie Geng
- College of Food Science, Northeast Agricultural University, Harbin, Heilongjiang 150030, China; College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China.
| |
Collapse
|
89
|
Miao J, Yong Y, Zheng Z, Zhang K, Li W, Liu J, Zhou S, Qin J, Sun H, Wang Y, Fu X, Luo X, Chen S, She Z, Cai J, Zhu P. Artesunate Inhibits Neointimal Hyperplasia by Promoting IRF4 Associated Macrophage Polarization. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408992. [PMID: 40126336 PMCID: PMC12097016 DOI: 10.1002/advs.202408992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 02/20/2025] [Indexed: 03/25/2025]
Abstract
Vascular restenosis is a serious clinical issue initiated and aggravated by macrophage inflammation, with no effective treatments available, in cardiovascular and autoimmune diseases. However, the untapped mechanisms and new targets that can regulate macrophage polarization and vascular restenosis remain elusive. The research identifies interferon regulatory factor 4 (IRF4) expression as crucial in macrophage polarization during arterial restenosis. Myeloid-specific Irf4 deficiency and overexpression experiments showed that IRF4 promoted M2 macrophage polarization, inhibited M1 macrophage transitions, and disrupted the interaction between macrophages and vascular smooth muscle cells to reduce neointimal hyperplasia by directly upregulating krüppel like factor 4 (KLF4) expression. Artesunate, an FDA-approved drug, is screened as a potent activator of IRF4 expression in M2 polarization, and its treatment attenuated arterial restenosis in rodents and non-human primates. The findings reveal a significant protective role of IRF4 in the development of neointimal hyperplasia by regulating macrophage polarization, and artesunate may be proposed as a novel therapy for vascular restenosis.
Collapse
Affiliation(s)
- Jinlin Miao
- Department of Clinical Immunology of Xijing Hospital and Department of Cell Biology of National Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'an710032China
| | - Yule Yong
- Department of Clinical Immunology of Xijing Hospital and Department of Cell Biology of National Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'an710032China
| | - Zhaohui Zheng
- Department of Clinical Immunology of Xijing Hospital and Department of Cell Biology of National Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'an710032China
| | - Kui Zhang
- Department of Clinical Immunology of Xijing Hospital and Department of Cell Biology of National Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'an710032China
| | - Wei Li
- Department of CardiologyRenmin HospitalWuhan UniversityWuhan430060China
- Institute of Model AnimalWuhan UniversityWuhan430071China
| | - Jiayi Liu
- Department of CardiologyRenmin HospitalWuhan UniversityWuhan430060China
- Institute of Model AnimalWuhan UniversityWuhan430071China
| | - Siyi Zhou
- Institute of Model AnimalWuhan UniversityWuhan430071China
- School of Basic Medical ScienceWuhan UniversityWuhan430071China
| | - Juan‐juan Qin
- Department of GeriatricsZhongnan HospitalWuhan UniversityWuhan430070China
| | - Haoyang Sun
- Department of Clinical Immunology of Xijing Hospital and Department of Cell Biology of National Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'an710032China
| | - Yatao Wang
- Department of Clinical Immunology of Xijing Hospital and Department of Cell Biology of National Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'an710032China
| | - Xianghui Fu
- Department of Clinical Immunology of Xijing Hospital and Department of Cell Biology of National Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'an710032China
| | - Xing Luo
- Department of Clinical Immunology of Xijing Hospital and Department of Cell Biology of National Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'an710032China
| | - Siyu Chen
- Department of Clinical Immunology of Xijing Hospital and Department of Cell Biology of National Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'an710032China
| | - Zhi‐Gang She
- Department of CardiologyRenmin HospitalWuhan UniversityWuhan430060China
- Institute of Model AnimalWuhan UniversityWuhan430071China
| | - Jingjing Cai
- Department of CardiologyThe Third Xiangya HospitalCentral South UniversityChangsha410013China
| | - Ping Zhu
- Department of Clinical Immunology of Xijing Hospital and Department of Cell Biology of National Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'an710032China
| |
Collapse
|
90
|
Branco F, Cunha J, Mendes M, Sousa JJ, Vitorino C. 3D Bioprinting Models for Glioblastoma: From Scaffold Design to Therapeutic Application. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2501994. [PMID: 40116532 DOI: 10.1002/adma.202501994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Indexed: 03/23/2025]
Abstract
Conventional in vitro models fail to accurately mimic the tumor in vivo characteristics, being appointed as one of the causes of clinical attrition rate. Recent advances in 3D culture techniques, replicating essential physical and biochemical cues such as cell-cell and cell-extracellular matrix interactions, have led to the development of more realistic tumor models. Bioprinting has emerged to advance the creation of 3D in vitro models, providing enhanced flexibility, scalability, and reproducibility. This is crucial for the development of more effective drug treatments, and glioblastoma (GBM) is no exception. GBM, the most common and deadly brain cancer, remains a major challenge, with a median survival of only 15 months post-diagnosis. This review highlights the key components needed for 3D bioprinted GBM models. It encompasses an analysis of natural and synthetic biomaterials, along with crosslinking methods to improve structural integrity. Also, it critically evaluates current 3D bioprinted GBM models and their integration into GBM-on-a-chip platforms, which hold noteworthy potential for drug screening and personalized therapies. A versatile development framework grounded on Quality-by-Design principles is proposed to guide the design of bioprinting models. Future perspectives, including 4D bioprinting and machine learning approaches, are discussed, along with the current gaps to advance the field further.
Collapse
Affiliation(s)
- Francisco Branco
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, 3000-548, Portugal
| | - Joana Cunha
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, 3000-548, Portugal
| | - Maria Mendes
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, 3000-548, Portugal
- Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Faculty of Sciences and Technology, University of Coimbra, Coimbra, 3004-535, Portugal
| | - João J Sousa
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, 3000-548, Portugal
- Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Faculty of Sciences and Technology, University of Coimbra, Coimbra, 3004-535, Portugal
| | - Carla Vitorino
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, 3000-548, Portugal
- Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Faculty of Sciences and Technology, University of Coimbra, Coimbra, 3004-535, Portugal
| |
Collapse
|
91
|
Eggenhofer E, Proneth B. Ferroptosis Inhibition: A Key Opportunity for the Treatment of Ischemia/Reperfusion Injury in Liver Transplantation. Transplantation 2025; 109:e228-e236. [PMID: 39294870 DOI: 10.1097/tp.0000000000005199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2024]
Abstract
The outcome after liver transplantation has improved in recent years, which can be attributed to superior storage and transportation conditions of the organs, as well as better peri- and postoperative management and advancements in surgical techniques. Nevertheless, there is an increasing discrepancy between the need for organs and their availability. Consequently, the mortality rate on the waiting list is high and continues to rise. One way of counteracting this trend is to increase the use of "expanded criteria donors." This means that more and more donors will be included, especially those who are older and having additional comorbidities (eg, steatosis). A major complication of any transplantation is the occurrence of ischemia/reperfusion injury (IRI), which often leads to liver dysfunction and failure. However, there have been various promising approaches to minimize IRI in recent years, but an effective and clinically applicable method to achieve a better outcome for patients after liver transplantation is still missing. Thereby, the so-called marginal organs are predominantly affected by IRI; thus, it is crucial to develop suitable and effective treatment options for patients. Recently, regulated cell death mechanisms, particularly ferroptosis, have been implicated to play a major role in IRI, including the liver. Therefore, inhibiting this kind of cell death modality presents a promising therapeutic approach for the management of this yet untreatable condition. Thus, this review provides an overview of the role of ferroptosis in liver IRI and transplantation and discusses possible therapeutic solutions based on ferroptosis inhibition to restrain IRI in marginal organs (especially steatosis and donation after circulatory death organs).
Collapse
Affiliation(s)
- Elke Eggenhofer
- Department of Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Bettina Proneth
- Institute of Metabolism and Cell Death, Helmholtz Munich, Neuherberg, Germany
| |
Collapse
|
92
|
Afshari R, Roy A, Jain S, Lum K, Huang J, Denton S, Annabi N. One-Pot Synthesis of Antibacterial and Antioxidant Self-Healing Bioadhesives Using Ugi Four-Component Reactions. J Biomed Mater Res B Appl Biomater 2025; 113:e35584. [PMID: 40317897 DOI: 10.1002/jbm.b.35584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 03/14/2025] [Accepted: 04/05/2025] [Indexed: 05/07/2025]
Abstract
Bioadhesive materials are extensively utilized as alternatives to surgical sutures and wound dressings. Despite significant advancements in their synthesis, current bioadhesives suffer from inadequate mechanical stability, suboptimal wet tissue adhesion, and a lack of inherent antibacterial and antioxidant properties, while requiring multistep synthesis processes, complicating their production for biomedical applications. To address these limitations, we developed a new bioadhesive, named UgiGel, synthesized through a one-pot Ugi four-component reaction (Ugi-4CR). Our strategy utilized gelatin as the backbone, 4-formylphenylboronic acid (4-FPBA) as an aldehyde source for improved adhesion and antibacterial activity, gallic acid (GA) as a carboxylic acid source for improved antioxidant activity and wound healing, and cyclohexyl isocyanide (CyIso) to induce pseudopeptide structures. The internal crosslinking between GA and 4-FPBA via dynamic boronate ester bond formation, triggered by slight pH changes (7.4-7.8) and temperature elevation (25°C-40°C), resulted in the formation of viscoelastic and self-healing hydrogels with water as the only byproduct without the need for initiator/light activation. UgiGel showed higher adhesion to porcine skin tissue (139.8 ± 8.7 kPa) as compared to commercially available bioadhesives, Evicel (26.3 ± 2.6 kPa) and Coseal (19.3 ± 9.9 kPa). It also demonstrated effective antibacterial properties against both Gram-negative and Gram-positive bacteria, as well as antioxidant activity. Additionally, the in vitro studies using NIH-3T3 cells confirmed the biocompatibility of the UgiGel over 7 days of culture. Moreover, in vivo biocompatibility and biodegradation of UgiGel were confirmed via subcutaneous implantation in rats for up to 28 days. Our results demonstrated that UgiGel outperformed commercially available bioadhesives in terms of adhesion, self-healing, and antibacterial activity, without compromising biocompatibility or physical properties, representing a promising multifunctional bioadhesive for wound sealing and repair.
Collapse
Affiliation(s)
- Ronak Afshari
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, California, USA
| | - Arpita Roy
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, California, USA
| | - Saumya Jain
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, California, USA
| | - Kaimana Lum
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, California, USA
| | - Joyce Huang
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California, USA
| | - Sam Denton
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California, USA
| | - Nasim Annabi
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, California, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
93
|
Yuan Z, Wu S, Fu L, Wang X, Wang Z, Shafiq M, Feng H, Han L, Song J, EL-Newehy M, Abdulhameed MM, Xu Y, Mo X, Jiang S. A natural biological adhesive from slug mucus for wound repair. Bioact Mater 2025; 47:513-527. [PMID: 40034409 PMCID: PMC11874230 DOI: 10.1016/j.bioactmat.2025.01.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/29/2024] [Accepted: 01/22/2025] [Indexed: 03/05/2025] Open
Abstract
Slugs could secrete mucus with multifunctional characteristics, such as reversible gelation, mucoadhesiveness, and viscoelasticity, which can be harnessed for multifaceted biotechnological and healthcare applications. The dried mucus (DM) was prepared using slug, which can be adhered to the tissue surface through different types of interactions (lap-shear force, 1.1 N for DM-3 group). The DM-3 further exhibited the highest hemostatic ability as discerned in a liver trauma injury model (hemostasis time, <15 s), biocompatibility and biodegradability (an insignificant residue at 4 weeks) in vivo, and considerably improved skin repair in full-thickness excisional wounds (wound closure, 96.2 % at day 14). Taken together, slug's mucus can be easily prepared with an economic and an eco-friendly method, which may have broad biotechnological and healthcare implications and potential utility in other related disciplines. This transition from natural components to the biomaterial may provide an invaluable platform for different types of applications.
Collapse
Affiliation(s)
- Zhengchao Yuan
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, 201620, Shanghai, PR China
| | - Siyuan Wu
- Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, PR China
| | - Liwen Fu
- Department of Orthopedic Oncology, Shanghai Bone Tumor Institute, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, PR China
| | - Xinyi Wang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, 201620, Shanghai, PR China
| | - Zewen Wang
- Department of Orthopaedics, Xinqiao Hospital, Army Military Medical University, No. 183, Xinqiao Street, Shapingba District, Chongqing, 400037, PR China
| | - Muhammad Shafiq
- Innovation Center of NanoMedicine (iCONM), Kawasaki Institute of Industrial Promotion, Kawasaki-ku, Kawasaki, 210-0821, Japan
| | - Hao Feng
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, 201620, Shanghai, PR China
| | - Lu Han
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, 201620, Shanghai, PR China
| | - Jiahui Song
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, 201620, Shanghai, PR China
| | - Mohamed EL-Newehy
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Meera Moydeen Abdulhameed
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Yuan Xu
- Department of Orthopaedics, Xinqiao Hospital, Army Military Medical University, No. 183, Xinqiao Street, Shapingba District, Chongqing, 400037, PR China
| | - Xiumei Mo
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, 201620, Shanghai, PR China
- Institute of Biomaterials and Biomedicine, School of Food and Pharmacy, Shanghai Zhongqiao Vocational and Technical University, Shanghai, 201514, PR China
| | - Shichao Jiang
- Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, PR China
| |
Collapse
|
94
|
Chen T, Zhang Y, Ding L, Xiong C, Mei C, Wei S, Jiang M, Huang Y, Chen J, Xie T, Zhu Q, Zhang Q, Huang X, Chen S, Li Y. Tripartite Motif Containing 65 Deficiency Confers Protection Against Acute Kidney Injury via Alleviating Voltage-Dependent Anion Channel 1-Mediated Mitochondrial Dysfunction. MedComm (Beijing) 2025; 6:e70149. [PMID: 40264575 PMCID: PMC12013732 DOI: 10.1002/mco2.70149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 01/02/2025] [Accepted: 02/17/2025] [Indexed: 04/24/2025] Open
Abstract
Acute kidney injury (AKI) is a prevalent and serious clinical disease with a high incidence rate and significant health burden. The limited understanding of the complex pathological mechanisms has hindered the development of efficacious therapeutics. Tripartite motif containing 65 (TRIM65) has recently been identified as a key regulator of acute inflammation. However, its role in AKI remains unclear. The present study observed that TRIM65 expression was upregulated in AKI. Moreover, the knockout of the Trim65 gene in mice exhibited a substantial protective impact against rhabdomyolysis, ischemia-reperfusion (I/R), and cisplatin-induced AKI. Mechanistically, TRIM65 directly binds and mediates K48/K63-linked polyubiquitination modifications of voltage-dependent anion channel 1 (VDAC1) at its K161 and K200 amino acid sites. TRIM65 plays a role in maintaining the stability of VDAC1 and preventing its degradation by the autophagy pathway. TRIM65 deficiency attenuates mitochondrial dysfunction in renal tubular epithelial cells during AKI. Conversely, the overexpression of VDAC1 in renal tissues has been demonstrated to negate the protective effect of TRIM65 deficiency on AKI. These findings suggest that TRIM65 may play a role regulating of AKI through the targeting of VDAC1-dependent mitochondrial function, offering potential avenues for the development of new drug targets and strategies for the treatment of AKI.
Collapse
Affiliation(s)
- Tao Chen
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Jiangxi Provincial Key Laboratory of Bioengineering Drugs, Institute of Translational Medicine, Jiangxi Medical CollegeNanchang UniversityNanchangChina
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of MedicineZhejiang UniversityHangzhouChina
| | - Yang Zhang
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Liting Ding
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Jiangxi Provincial Key Laboratory of Bioengineering Drugs, Institute of Translational Medicine, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Chenlu Xiong
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Chao Mei
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Sisi Wei
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Ming Jiang
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Jiangxi Provincial Key Laboratory of Bioengineering Drugs, Institute of Translational Medicine, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Yingjie Huang
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Jiangxi Provincial Key Laboratory of Bioengineering Drugs, Institute of Translational Medicine, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Jianrong Chen
- Department of Endocrinology, The First Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Tao Xie
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Jiangxi Provincial Key Laboratory of Bioengineering Drugs, Institute of Translational Medicine, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Qing Zhu
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Jiangxi Provincial Key Laboratory of Bioengineering Drugs, Institute of Translational Medicine, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Qi Zhang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Jiangxi Provincial Key Laboratory of Bioengineering Drugs, Institute of Translational Medicine, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Xuan Huang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Jiangxi Provincial Key Laboratory of Bioengineering Drugs, Institute of Translational Medicine, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Shibiao Chen
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Yong Li
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical CollegeNanchang UniversityNanchangChina
| |
Collapse
|
95
|
Romoli J, Chiodelli P, Signoroni PB, Vertua E, Ferrari C, Giuzzi E, Paini A, Scalvini E, Papait A, Stefani FR, Silini AR, Parolini O. Modeling Stromal Cells Inside the Tumor Microenvironment of Ovarian Cancer: In Vitro Generation of Cancer-Associated Fibroblast-Like Cells and Their Impact in a 3D Model. MedComm (Beijing) 2025; 6:e70172. [PMID: 40255916 PMCID: PMC12006666 DOI: 10.1002/mco2.70172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 01/31/2025] [Accepted: 02/13/2025] [Indexed: 04/22/2025] Open
Abstract
The tumor microenvironment (TME) is the combination of cells and factors that promotes tumor progression, and cancer-associated fibroblasts (CAFs) are a key component within TME. CAF originates from various stromal cells and is activated by factors such as transforming growth factor-beta (TGF-β) secreted by tumor cells, favoring chemoresistance and metastasis. Recent publications have underlined plasticity and heterogeneity and their strong contribution to the reactive stroma within the TME. Our study aimed to replicate the TME's structure by creating a 3D in vitro model of ovarian cancer (OC). By incorporating diverse tumor and stromal cells, we simulated a physiologically relevant environment for studying CAF-like cell behavior within tumor spheroids in a context-dependent manner. CAF-like cells were generated by exposing human dermal fibroblasts to OC cell line conditioned media in the presence or absence of TGF-β. Herein, we found that different stimuli induce the generation of heterogeneous populations of CAF-like cells. Notably, we observed the ability of CAF-like cells to shape the intratumoral architecture and to contribute to functional changes in tumor cell behavior. This study highlights the importance of precise assessment of CAF for potential therapeutic interventions and further provides a reliable model for investigating novel therapeutic targets in OC.
Collapse
Affiliation(s)
- Jacopo Romoli
- Department of Life Science and Public HealthUniversità Cattolica del Sacro CuoreRomeItaly
| | - Paola Chiodelli
- Department of Life Science and Public HealthUniversità Cattolica del Sacro CuoreRomeItaly
| | | | - Elsa Vertua
- Centro di Ricerca E. MenniFondazione Poliambulanza Istituto OspedalieroBresciaItaly
| | - Clarissa Ferrari
- Research and Clinical Trials UnitFondazione Poliambulanza Istituto OspedalieroBresciaItaly
| | - Elisabetta Giuzzi
- Centro di Ricerca E. MenniFondazione Poliambulanza Istituto OspedalieroBresciaItaly
| | - Alice Paini
- Centro di Ricerca E. MenniFondazione Poliambulanza Istituto OspedalieroBresciaItaly
| | - Elisa Scalvini
- Centro di Ricerca E. MenniFondazione Poliambulanza Istituto OspedalieroBresciaItaly
| | - Andrea Papait
- Department of Life Science and Public HealthUniversità Cattolica del Sacro CuoreRomeItaly
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCSRomeItaly
| | | | | | - Ornella Parolini
- Department of Life Science and Public HealthUniversità Cattolica del Sacro CuoreRomeItaly
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCSRomeItaly
| |
Collapse
|
96
|
Zhong L, Guo X, Deng L, Wang X, He H, Wu N, Tang R, Chen L, Chen Y, Li P. Dopant-Regulated Piezocatalysts Evoke Sonopiezoelectric and Enzymatic PANoptosis for Synergistic Cancer Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2500406. [PMID: 40056039 PMCID: PMC12061309 DOI: 10.1002/advs.202500406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/17/2025] [Indexed: 05/10/2025]
Abstract
Piezocatalyst-enabled sonopiezoelectric therapy offers noninvasive treatment with high spatiotemporal selectivity, yet existing piezocatalysts are limited by suboptimal efficacy, cancer cell resistance to oxidative stress, and biosafety concerns. Here, hafnia (HfO2), one of the only few FDA-approved inorganic nanomaterials for clinical trials, is identified as a promising piezocatalyst with high translational potential for sonopiezoelectric and enzymatic PANoptosis-boosted nanocatalytic therapy. Specifically, engineered transition metal-substituted HfO2 nanocatalysts are synthesized to optimize piezoelectric and enzyme-mimicking activities. Among these, Mn-substituted HfO2 with a 20% Mn ratio (HMO) demonstrates superior performance in sono-triggered reactive oxygen species generation, attributed to its reduced bandgap and increased oxygen vacancies. HMO also exhibits multiple enzyme-mimicking activities, including peroxidase (POD), catalase (CAT), and glutathione peroxidase (GPx), amplifying oxidative stress through tumor-specific catalytic reactions. These dual catalytic effects enable the activation of cancer cell PANoptosis to elicit a robust antitumor immune response. Biological evaluations show significant tumor suppression and antitumor immune responses by HMO-mediated nanocatalytic therapy. Unlike utilizing the radiosensitization ability of HfO2 in the clinic, this work unveils the distinctive sonopiezoelectric effect and multienzymatic activities of HfO2-based nanocatalysts for biomedical applications, holding the potential to overcome the challenges of radiation damage associated with radiotherapy.
Collapse
Affiliation(s)
- Linhong Zhong
- Ultrasound Department of the Second Affiliated Hospital of Chongqing Medical UniversityChongqing Key Laboratory of Ultrasound Molecular ImagingChongqing400010P. R. China
| | - Xun Guo
- Ultrasound Department of the Second Affiliated Hospital of Chongqing Medical UniversityChongqing Key Laboratory of Ultrasound Molecular ImagingChongqing400010P. R. China
| | - Liming Deng
- The First Affiliated Hospital of Chongqing Medical UniversityChongqing400010P. R. China
| | - Xiaoting Wang
- Ultrasound Department of the Second Affiliated Hospital of Chongqing Medical UniversityChongqing Key Laboratory of Ultrasound Molecular ImagingChongqing400010P. R. China
| | - Hongye He
- Ultrasound Department of the Second Affiliated Hospital of Chongqing Medical UniversityChongqing Key Laboratory of Ultrasound Molecular ImagingChongqing400010P. R. China
| | - Nianhong Wu
- Ultrasound Department of the Second Affiliated Hospital of Chongqing Medical UniversityChongqing Key Laboratory of Ultrasound Molecular ImagingChongqing400010P. R. China
| | - Rui Tang
- Ultrasound Department of the Second Affiliated Hospital of Chongqing Medical UniversityChongqing Key Laboratory of Ultrasound Molecular ImagingChongqing400010P. R. China
| | - Liang Chen
- Materdicine LabSchool of Life SciencesShanghai UniversityShanghai200444P. R. China
| | - Yu Chen
- Materdicine LabSchool of Life SciencesShanghai UniversityShanghai200444P. R. China
- Shanghai Institute of MaterdicineShanghai200051P. R. China
| | - Pan Li
- Ultrasound Department of the Second Affiliated Hospital of Chongqing Medical UniversityChongqing Key Laboratory of Ultrasound Molecular ImagingChongqing400010P. R. China
| |
Collapse
|
97
|
Hang R, Yao X, Bai L, Hang R. Evolving biomaterials design from trial and error to intelligent innovation. Acta Biomater 2025; 197:29-47. [PMID: 40081552 DOI: 10.1016/j.actbio.2025.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 01/20/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
The design and exploration of biomaterials plays a pivotal role in many fields, including medical and engineering. The prevailing approach to biomaterials discovery relies on orthogonal experiments, with repeated attempts to optimize experimental conditions. This method has proven invaluable in gaining experience, but it is also inefficient and challenging to predict the behavior of complex systems. The advent of high-throughput screening (HTS) techniques has led to a notable enhancement in the efficiency of biomaterials development, enabling researchers to assess a vast array of material combinations within a relatively short timeframe. Nevertheless, the emergence of artificial intelligence (AI) has been the catalyst for a new era in biomaterials design. AI has markedly accelerated the development of new materials by enabling the prediction of material properties through machine learning (ML) and deep learning models, as well as optimizing the design pipeline. This review will present a systematic overview of the development of biomaterials design technology. It will also explore the integration of AI with HTS technology and envisage the potential of AI-driven materials design in biomaterials for the future. STATEMENT OF SIGNIFICANCE: The design and synthesis of biomaterials have undergone substantial shifts, reflecting evolving research paradigms. High-throughput screening has emerged as a broad and efficient alternative to traditional free-form combination methods in biomaterial design. The advent of artificial intelligence (AI) enables personalized biomaterial design and, as a transformative tool in biomaterial development, is poised to redefine the field and offer long-term solutions for its advancement. Building on these advancements, this review systematically summarizes the evolution of biomaterial design, offering insights into the future trajectory of the field.
Collapse
Affiliation(s)
- Ruiyue Hang
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, PR China
| | - Xiaohong Yao
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, PR China
| | - Long Bai
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, PR China.
| | - Ruiqiang Hang
- Shanxi Key Laboratory of Biomedical Metal Materials, College of Materials Science and Engineering, Taiyuan University of Technology, Taiyuan 030024, PR China.
| |
Collapse
|
98
|
Cao S, Pang Y, Wei Y, Wang D, Xiong A, Yang J, Zeng H. Nanozymes in biomedicine: Unraveling trends, research foci, and future trajectories via bibliometric insights (from 2007 to 2024). Int J Biol Macromol 2025; 309:142798. [PMID: 40185460 DOI: 10.1016/j.ijbiomac.2025.142798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/27/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Nanozymes, a new generation of artificial enzymes, have attracted significant attention in biomedical applications due to their multifunctional properties, multi-enzyme mimicking abilities, cost-effectiveness, and high stability. Leveraging these diverse catalytic activities, an increasing number of nanozyme-based therapeutic strategies have been developed for the treatment of various diseases. Despite substantial research efforts, a significant gap remains in comprehensive studies examining the progression, key areas, current trends, and future directions in this field. This study provides a comprehensive overview of nanozyme applications in biomedical research over the past 17 years, utilizing data from the Web of Science Core Collection, covering the period from January 1, 2007, to October 8, 2024. Advanced bibliometric and visualization tools were employed to facilitate a comprehensive analysis. The results highlight China's dominant role in this field, accounting for 76.83 % of total publications, significantly influencing the evolution of research in this area. Key contributions were made by institutions such as the Chinese Academy of Sciences, the University of Chinese Academy of Sciences, and the University of Science and Technology of China, with Qu Xiaogang as the leading author. The journal ACS Applied Materials & Interfaces has become the most prolific publisher in this field. Keyword analysis indicates that since 2022, research hotspots in this field have increasingly focused on areas such as photothermal therapy, chemodynamic therapy, and ferroptosis. Challenges such as obstacles to clinical translation, limitations in recyclability, and insufficient targeting ability were addressed. The potential applications of emerging interdisciplinary technologies, such as artificial intelligence, machine learning, and organoids, in advancing nanozyme development were explored. This study offers a data-driven roadmap for researchers to navigate the evolving landscape of nanozyme innovation, emphasizing interdisciplinary collaboration in impactful biomedical applications.
Collapse
Affiliation(s)
- Siyang Cao
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, People's Republic of China; Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, People's Republic of China; Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, People's Republic of China
| | - Yingchen Pang
- Department of Pulmonary and Critical Care Medicine, Peking University Shenzhen Hospital, Shenzhen, Guangdong, People's Republic of China; Department of Pulmonary and Critical Care Medicine, Shenzhen Xinhua Hospital, Shenzhen, Guangdong, People's Republic of China
| | - Yihao Wei
- Department of Rehabilitation Science, The Hong Kong Polytechnic University, Hong Kong; Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences (CAS), Shenzhen, Guangdong, People's Republic of China; Faculty of Pharmaceutical Sciences, Shenzhen University of Advanced Technology, Shenzhen, Guangdong, People's Republic of China
| | - Deli Wang
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, People's Republic of China; Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, People's Republic of China; Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, People's Republic of China
| | - Ao Xiong
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, People's Republic of China; Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, People's Republic of China; Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, People's Republic of China.
| | - Jun Yang
- Department of Radiology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, People's Republic of China.
| | - Hui Zeng
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, People's Republic of China; Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, People's Republic of China; Department of Orthopedics, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, People's Republic of China.
| |
Collapse
|
99
|
Teng Y, Zhang X, Song L, Yang J, Li D, Shi Z, Guo X, Wang S, Fan H, Jiang L, Hou S, Ramakrishna S, Lv Q, Shi J. Construction of anti-calcification small-diameter vascular grafts using decellularized extracellular matrix/poly (L-lactide-co-ε-caprolactone) and baicalin-cathepsin S inhibitor. Acta Biomater 2025; 197:184-201. [PMID: 40120837 DOI: 10.1016/j.actbio.2025.03.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 03/07/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
The long-term transplantation of small-diameter vascular grafts (SDVGs) is associated with a risk of calcification, which is a key factor limiting the clinical translation of SDVG. Hence, there is an urgency attached to the development of new SDVGs with anti-calcification properties. Here, we used decellularized extracellular matrix (dECM) and poly (L-lactide-co-ε-caprolactone) (PLCL) as base materials and combined these with baicalin, cathepsin S (Cat S) inhibitor to prepare PBC-SDVGs by electrospinning. Baicalin contains carboxyl and hydroxyl groups that can interact with chemical groups in dECM powder, potentially blocking calcium nucleation sites. Cat S inhibitor prevents elastin degradation and further reduces the risk of calcification. PBC-SDVGs were biocompatible and when implanted in rat abdominal aorta, accelerated endothelialization, enhanced vascular tissue regeneration, inhibited elastin degradation, and promoted macrophage polarization M2 phenotype to regulate inflammation. After 3 months of implantation, the results of Doppler ultrasound, MicroCT, and histological staining revealed a significant reduction in calcification. In summary, the developed anti-calcification SDVGs offer a promising strategy for long-term implantation with significant clinical application potential. STATEMENT OF SIGNIFICANCE: The dECM and PLCL were used as base materials, connected with baicalin, and loaded with Cat S inhibitor to prepare PBC-SDVGs. The baicalin and dECM powder formed hydrogen bonds to crosslink together reducing the calcium deposition. In vitro, the vascular graft downregulated the expression level of osteogenic genes and promoted macrophage polarization toward an anti-inflammatory M2 phenotype, thereby reducing calcification. The PBC-SDVGs implanted in rat abdominal aorta can accelerate endothelialization, enhance vascular tissue regeneration, inhibit elastin degradation, reduce inflammation response and calcification.
Collapse
Affiliation(s)
- Yanjiao Teng
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, PR China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, PR China; Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou 325026, PR China
| | - Xiaohai Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210000, PR China
| | - Lin Song
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210000, PR China
| | - Jianing Yang
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, PR China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, PR China; Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou 325026, PR China
| | - Duo Li
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, PR China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, PR China; Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou 325026, PR China
| | - Ziqi Shi
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, PR China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, PR China; Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou 325026, PR China
| | - Xiaoqin Guo
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, PR China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, PR China; Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou 325026, PR China
| | - Shufang Wang
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, PR China
| | - Haojun Fan
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, PR China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, PR China; Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou 325026, PR China
| | - Li Jiang
- Tianjin Eye Hospital, Nankai University Affiliated Eye Hospital, Tianjin 300021, PR China
| | - Shike Hou
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, PR China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, PR China; Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou 325026, PR China.
| | - Seeram Ramakrishna
- Department of Mechanical Engineering, College of Design and Engineering, National University of Singapore, Singapore 117575, Singapore.
| | - Qi Lv
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, PR China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, PR China; Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou 325026, PR China.
| | - Jie Shi
- School of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, PR China; Key Laboratory for Disaster Medicine Technology, Tianjin 300072, PR China; Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou 325026, PR China.
| |
Collapse
|
100
|
Choi M, Choi W, Hwang PTJ, Oh Y, Jun T, Ryu DY, Kim NK, Jang EH, Shin YR, Youn YN, Lee SH, Jung SY, Hong J. Engineered silk fibroin bio-hybrid artificial graft with releasing biological gas for enhanced circulatory stability and surgical performance. Int J Biol Macromol 2025; 309:142760. [PMID: 40185440 DOI: 10.1016/j.ijbiomac.2025.142760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 03/20/2025] [Accepted: 03/31/2025] [Indexed: 04/07/2025]
Abstract
Cardiovascular disease (CVD) compromises a range of conditions affecting the heart and blood vessels, and is the leading cause of mortality globally. Vascular grafts are essential in cardiovascular surgical interventions. In clinical treatment, low mechanical durability, thrombosis and hyperplasia are primary failure modes for vascular grafts, highlighting the challenge of developing small-diameter grafts that withstand stress and integrate. A lack of suitable autologous grafts is a main cause of surgery failures. Herein, we have engineered silk fibroin (SF)-based small-diameter artificial grafts (NOeGraft) using a biologically functional polyurethane (PU) template with cost-effectiveness and high feasibility. This template facilitates the generation of biological gases via S-nitrosylation and improves mechanical properties by modulating the secondary structure of SF. Nitric oxide (NO) is one of the most essential biological gases for the cardiovascular system. NO release from NOeGraft suppresses platelet adhesion and smooth muscle cell (SMC) proliferation while scavenging reactive oxygen species (ROS) and promoting epithelial cell growth. Additionally, the suture retention strength of the NOeGraft exceeds 3.4 N. We evaluated the circulatory performance of the NOeGraft using a blood pressure-controllable system, observing no leaks or failures over 2535 min. Cost-effective NOeGraft provides biologically functional and mechanically advantageous solutions for cardiovascular surgeries.
Collapse
Affiliation(s)
- Moonhyun Choi
- Department of Chemical & Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Woojin Choi
- Department of Chemical & Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Patrick T J Hwang
- Department of Biomedical Engineering, College of Engineering, Rowan University, 201 Mullica Hill Rd., Glassboro, NJ 08028, USA
| | - Yoogyeong Oh
- Department of Chemical & Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Taesuk Jun
- Department of Chemical & Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Du Yeol Ryu
- Department of Chemical & Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Nam Kyun Kim
- Division of Cardiovascular Surgery, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Eui Hwa Jang
- Division of Cardiovascular Surgery, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Yu Rim Shin
- Division of Cardiovascular Surgery, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Young-Nam Youn
- Division of Cardiovascular Surgery, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Seung Hyun Lee
- Division of Cardiovascular Surgery, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Se Yong Jung
- Division of Pediatric Cardiology, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.
| | - Jinkee Hong
- Department of Chemical & Biomolecular Engineering, College of Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea.
| |
Collapse
|