1
|
Zhu S, Diao S, Liu X, Zhang Z, Liu F, Chen W, Lu X, Luo H, Cheng X, Liao Q, Li Z, Chen J. Biomaterial-based strategies: a new era in spinal cord injury treatment. Neural Regen Res 2025; 20:3476-3500. [PMID: 40095657 PMCID: PMC11974648 DOI: 10.4103/nrr.nrr-d-24-00844] [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: 07/29/2024] [Revised: 09/02/2024] [Accepted: 12/16/2024] [Indexed: 03/19/2025] Open
Abstract
Enhancing neurological recovery and improving the prognosis of spinal cord injury have gained research attention recently. Spinal cord injury is associated with a complex molecular and cellular microenvironment. This complexity has prompted researchers to elucidate the underlying pathophysiological mechanisms and changes and to identify effective treatment strategies. Traditional approaches for spinal cord injury repair include surgery, oral or intravenous medications, and administration of neurotrophic factors; however, the efficacy of these approaches remains inconclusive, and serious adverse reactions continue to be a concern. With advancements in tissue engineering and regenerative medicine, emerging strategies for spinal cord injury repair now involve nanoparticle-based nanodelivery systems, scaffolds, and functional recovery techniques that incorporate biomaterials, bioengineering, stem cell, and growth factors as well as three-dimensional bioprinting. Ideal biomaterial scaffolds should not only provide structural support for neuron migration, adhesion, proliferation, and differentiation but also mimic the mechanical properties of natural spinal cord tissue. Additionally, these scaffolds should facilitate axon growth and neurogenesis by offering adjustable topography and a range of physical and biochemical cues. The three-dimensionally interconnected porous structure and appropriate physicochemical properties enabled by three-dimensional biomimetic printing technology can maximize the potential of biomaterials used for treating spinal cord injury. Therefore, correct selection and application of scaffolds, coupled with successful clinical translation, represent promising clinical objectives to enhance the treatment efficacy for and prognosis of spinal cord injury. This review elucidates the key mechanisms underlying the occurrence of spinal cord injury and regeneration post-injury, including neuroinflammation, oxidative stress, axon regeneration, and angiogenesis. This review also briefly discusses the critical role of nanodelivery systems used for repair and regeneration of injured spinal cord, highlighting the influence of nanoparticles and the factors that affect delivery efficiency. Finally, this review highlights tissue engineering strategies and the application of biomaterial scaffolds for the treatment of spinal cord injury. It discusses various types of scaffolds, their integrations with stem cells or growth factors, and approaches for optimization of scaffold design.
Collapse
Affiliation(s)
- Shihong Zhu
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Sijun Diao
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Xiaoyin Liu
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan Province, China
| | - Zhujun Zhang
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Fujun Liu
- Department of Ophthalmology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Wei Chen
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Xiyue Lu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Huiyang Luo
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Xu Cheng
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Qiang Liao
- Department of Pharmacy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Zhongyu Li
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Jing Chen
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
2
|
Sonar S, Das A, Kalele K, Subramaniyan V. Exosome-based cancer vaccine: a cell-free approach. Mol Biol Rep 2025; 52:421. [DOI: 10.1007/s11033-025-10519-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 04/15/2025] [Indexed: 05/04/2025]
|
3
|
Abdelsattar M, Ramadan AM, Eltayeb AE, Saleh OM, Abdel-Tawab FM, Fahmy EM, Hassanein SE, Ali HM, Al-Saud NBS, Alameldin HF, Hassan SM, Mohamed NG, Abdel Azeiz AZ, Bahieldin A, Eissa HF. Development of transgenic wheat plants withstand salt stress via the MDAR1 gene. GM CROPS & FOOD 2025; 16:173-187. [PMID: 39927641 PMCID: PMC11812330 DOI: 10.1080/21645698.2025.2463139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 01/09/2025] [Accepted: 02/02/2025] [Indexed: 02/11/2025]
Abstract
In light of the fact that climate change has emerged as one of the difficulties confronting the global food system, researchers are obligated to work toward developing fundamental crops, particularly wheat, to combat environmental stress, including drought and salt. In the present study, genetic engineering was used to transfer the Arabidopsis MDAR1 gene, which controls the buildup of ascorbic acid (AsA) to make bread wheat less likely to be sensitive to salt stress. The biolistic bombardment was used to transfer cDNA from the Arabidopsis thaliana plant that encodes MDAR1 into Bobwhite 56 cultivar wheat plants. A molecular investigation was performed on six different transgenic lines to confirm the integration of the transgene, the copy number, and the expression of the transgene. There were one to three copies of the transgene, and there was no association found between the number of copies of the transgene and All the data generated or analyzed during this study are included in this published article [and its supplementary information files].the presence of its expression. Compared to plants that were not transgenic, the amount of ascorbic acid (AsA) that accumulated in the transgenic plants was twice as high. ROS concentrations are significantly lower in transgenic plants compared to non-transgenic plants under both control and salt stress conditions, effectively reducing oxidative stress. By cultivating transgenic T2 plants in a greenhouse, we were able to determine whether they were able to tolerate the potentially damaging effects of salt stress (200 mm). The study concluded that transgenic wheat plants that consistently expressed the MDAR1 gene become tolerant to salt stress with improvement in growth characteristics.
Collapse
Affiliation(s)
- Mohamed Abdelsattar
- Agricultural Genetic Engineering Research Institute (AGERI), Agricultural Research Center (ARC), Giza, Egypt
| | - Ahmed M. Ramadan
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University (KAU), Jeddah, Saudi Arabia
- Princess Najla Bint Saud Al-Saud Center for Excellence Research in Biotechnology, King Abdulaziz, Saudi Arabia
| | - Amin E. Eltayeb
- Arid Land Research Center, Tottori University, Tottori, Japan
| | - Osama M. Saleh
- Department of Natural Products Research, National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority
| | | | - Eman M. Fahmy
- Department of Genetics, Faculty of Agriculture, Ain Shams University, Cairo, Egypt
| | - Sameh E. Hassanein
- Agricultural Genetic Engineering Research Institute (AGERI), Agricultural Research Center (ARC), Giza, Egypt
- Bioinformatics Program, School of Biotechnology, Nile University, Giza, Egypt
| | - Hani M. Ali
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University (KAU), Jeddah, Saudi Arabia
- Princess Najla Bint Saud Al-Saud Center for Excellence Research in Biotechnology, King Abdulaziz, Saudi Arabia
| | - Najla B. S. Al-Saud
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University (KAU), Jeddah, Saudi Arabia
- Princess Najla Bint Saud Al-Saud Center for Excellence Research in Biotechnology, King Abdulaziz, Saudi Arabia
| | - Hussien. F. Alameldin
- Agricultural Genetic Engineering Research Institute (AGERI), Agricultural Research Center (ARC), Giza, Egypt
- DOE-Plant Research Lab, Michigan State University, East Lansing, Michigan, USA
| | - Sabah M. Hassan
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University (KAU), Jeddah, Saudi Arabia
- Princess Najla Bint Saud Al-Saud Center for Excellence Research in Biotechnology, King Abdulaziz, Saudi Arabia
| | - Nermin G. Mohamed
- College of Biotechnology, Misr University for Science and Technology
| | | | - Ahmed Bahieldin
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University (KAU), Jeddah, Saudi Arabia
| | - Hala F. Eissa
- Agricultural Genetic Engineering Research Institute (AGERI), Agricultural Research Center (ARC), Giza, Egypt
- College of Biotechnology, Misr University for Science and Technology
| |
Collapse
|
4
|
Liang H, Zhou B, Li P, Zhang X, Zhang S, Zhang Y, Yao S, Qu S, Chen J. Stemness regulation in prostate cancer: prostate cancer stem cells and targeted therapy. Ann Med 2025; 57:2442067. [PMID: 39711287 DOI: 10.1080/07853890.2024.2442067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 11/07/2024] [Accepted: 11/22/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND Increasing evidence indicates that cancer stem cells (CSCs) and cancer stem-like cells form a special subpopulation of cells that are ubiquitous in tumors. These cells exhibit similar characteristics to those of normal stem cells in tissues; moreover, they are capable of self-renewal and differentiation, as well as high tumorigenicity and drug resistance. In prostate cancer (PCa), it is difficult to kill these cells using androgen signaling inhibitors and chemotherapy drugs. Consequently, the residual prostate cancer stem cells (PCSCs) mediate tumor recurrence and progression. OBJECTIVE This review aims to provide a comprehensive and up-to-date overview of PCSCs, with a particular emphasis on potential therapeutic strategies targeting these cells. METHODS After searching in PubMed and Embase databases using 'prostate cancer' and 'cancer stem cells' as keywords, studies related were compiled and examined. RESULTS In this review, we detail the origin and characteristics of PCSCs, introduce the regulatory pathways closely related to CSC survival and stemness maintenance, and discuss the link between epithelial-mesenchymal transition, tumor microenvironment and tumor stemness. Furthermore, we introduce the currently available therapeutic strategies targeting CSCs, including signaling pathway inhibitors, anti-apoptotic protein inhibitors, microRNAs, nanomedicine, and immunotherapy. Lastly, we summarize the limitations of current CSC research and mention future research directions. CONCLUSION A deeper understanding of the regulatory network and molecular markers of PCSCs could facilitate the development of novel therapeutic strategies targeting these cells. Previous preclinical studies have demonstrated the potential of this treatment approach. In the future, this may offer alternative treatment options for PCa patients.
Collapse
Affiliation(s)
- Hao Liang
- Department of Urology, Qilu Hospital of Shandong University (Qingdao), Qingdao, China
| | - Bin Zhou
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Peixin Li
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaoyi Zhang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Shijie Zhang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Yaozhong Zhang
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Shengwen Yao
- Department of Urology, Qilu Hospital of Shandong University, Jinan, China
| | - Sifeng Qu
- Department of Urology, Qilu Hospital of Shandong University (Qingdao), Qingdao, China
| | - Jun Chen
- Department of Urology, Qilu Hospital of Shandong University (Qingdao), Qingdao, China
| |
Collapse
|
5
|
Ji J, Gong C, Lu G, Zhang J, Liu B, Liu X, Lin J, Wang P, Thomas BB, Humayun MS, Zhou Q. Potential of ultrasound stimulation and sonogenetics in vision restoration: a narrative review. Neural Regen Res 2025; 20:3501-3516. [PMID: 39688549 PMCID: PMC11974640 DOI: 10.4103/nrr.nrr-d-24-00841] [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: 07/27/2024] [Revised: 11/04/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
Vision restoration presents a considerable challenge in the realm of regenerative medicine, while recent progress in ultrasound stimulation has displayed potential as a non-invasive therapeutic approach. This narrative review offers a comprehensive overview of current research on ultrasound-stimulated neuromodulation, emphasizing its potential as a treatment modality for various nerve injuries. By examining of the efficacy of different types of ultrasound stimulation in modulating peripheral and optic nerves, we can delve into their underlying molecular mechanisms. Furthermore, the review underscores the potential of sonogenetics in vision restoration, which involves leveraging pharmacological and genetic manipulations to inhibit or enhance the expression of related mechanosensitive channels, thereby modulating the strength of the ultrasound response. We also address how methods such as viral transcription can be utilized to render specific neurons or organs highly responsive to ultrasound, leading to significantly improved therapeutic outcomes.
Collapse
Affiliation(s)
- Jie Ji
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Chen Gong
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
- USC Roski Eye Institute, Keck School of Medicine of University of Southern California, Los Angeles, CA, USA
| | - Gengxi Lu
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
- USC Roski Eye Institute, Keck School of Medicine of University of Southern California, Los Angeles, CA, USA
| | - Junhang Zhang
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
- USC Roski Eye Institute, Keck School of Medicine of University of Southern California, Los Angeles, CA, USA
| | - Baoqiang Liu
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Xunan Liu
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| | - Junhao Lin
- Thomas Lord Department of Computer Science, University of Southern California, Los Angeles, CA, USA
| | | | - Biju B. Thomas
- USC Roski Eye Institute, Keck School of Medicine of University of Southern California, Los Angeles, CA, USA
| | - Mark S. Humayun
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
- USC Roski Eye Institute, Keck School of Medicine of University of Southern California, Los Angeles, CA, USA
| | - Qifa Zhou
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
- USC Roski Eye Institute, Keck School of Medicine of University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
6
|
Simberg D, Barenholz Y, Roffler SR, Landfester K, Kabanov AV, Moghimi SM. PEGylation technology: addressing concerns, moving forward. Drug Deliv 2025; 32:2494775. [PMID: 40264371 PMCID: PMC12020137 DOI: 10.1080/10717544.2025.2494775] [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: 02/28/2025] [Revised: 04/01/2025] [Accepted: 04/11/2025] [Indexed: 04/24/2025] Open
Abstract
PEGylation technology, that is grafting of poly(ethylene glycol)(PEG) to biologics, vaccines and nanopharmaceuticals, has become a cornerstone of modern medicines with over thirty products used in the clinic. PEGylation of therapeutic proteins, nucleic acids and nanopharmaceuticals improves their stability, pharmacokinetic and biodistribution. While PEGylated medicines are safe in the majority of patients, there are growing concerns about the emergence of anti-PEG antibodies and their impact on the therapeutic efficacy of PEGylated medicines as well as broader immune responses, particularly in complement activation and hypersensitivity reactions. These concerns are beginning to scrutinize the future viability of PEGylation technology in medicine design. Here, we outline these concerns, encourage more efforts into looking for comprehensive scientific evidence on the role of anti-PEG antibodies in hypersensitivity reactions, discuss alternatives to PEG and propose strategies for moving PEGylation technology forward.
Collapse
Affiliation(s)
- Dmitri Simberg
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences and Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Translational Bio-Nanosciences Laboratory, Department of Pharmaceutical Sciences, Skaggs School of Pharmacy, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Yechezkel Barenholz
- Department of Biochemistry and Molecular Biology, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Steve R. Roffler
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Katharina Landfester
- Department of Physical Chemistry of Polymers, Max Planck Institute for Polymer Research, Mainz, Germany
| | | | - Seyed M. Moghimi
- Department of Pharmaceutical Sciences, The Skaggs School of Pharmacy and Pharmaceutical Sciences and Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- School of Pharmacy, Newcastle University, Newcastle upon Tyne, UK
- Translational and Clinical Research Institute, Faculty of Health and Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
7
|
Zhang J, Zeng F, Li Y, Mu C, Liu C, Wang L, Peng X, He L, Su Y, Li H, Wang A, Feng L, Gao D, Zhang Z, Xu G, Wang Y, Yue R, Si J, Zheng L, Zhang X, He F, Yi H, Tang Z, Li G, Ma K, Li Q. The characterization of technical design of a virus-like structure (VLS) nanodelivery system as vaccine candidate against SARS-CoV-2 variants. Hum Vaccin Immunother 2025; 21:2473183. [PMID: 40045463 PMCID: PMC11901403 DOI: 10.1080/21645515.2025.2473183] [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: 01/15/2025] [Revised: 02/11/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
The constant mutation of SARS-CoV-2 has led to the continuous appearance of viral variants and their pandemics and has improved the development of vaccines with a broad spectrum of antigens to curb the spread of the virus. The work described here suggested a novel vaccine with a virus-like structure (VLS) composed of combined mRNA and protein that is capable of stimulating the immune system in a manner similar to that of viral infection. This VLS vaccine is characterized by its ability to specifically target dendritic cells and/or macrophages through S1 protein recognition of the DC-SIGN receptor in cells, which leads to direct mRNA delivery to these innate immune cells for activation of robust immunity with a broad spectrum of neutralizing antibodies and immune protective capacity against variants. Research on its composition characteristics and structural features has suggested its druggability. Compared with the current mRNA vaccine, the VLS vaccine was identified as having no cytotoxicity at its effective application dosage, while the results of safety observations in animals revealed fewer adverse reactions during immunization.
Collapse
Affiliation(s)
- Jingjing Zhang
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
- Shandong Weigao Litong Biological Products Co, Ltd, Weihai, China
| | - Fengyuan Zeng
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Yanmei Li
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Changyong Mu
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Change Liu
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Lichun Wang
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Xiaowu Peng
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Liping He
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Yanrui Su
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Hongbing Li
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - An Wang
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Lin Feng
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Dongxiu Gao
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Zhixiao Zhang
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Gang Xu
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Yixuan Wang
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Rong Yue
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Junbo Si
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Lichun Zheng
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Xiong Zhang
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Fuyun He
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Hongkun Yi
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Zhongshu Tang
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Gaocan Li
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| | - Kaili Ma
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
- Shandong Weigao Litong Biological Products Co, Ltd, Weihai, China
| | - Qihan Li
- Weirui Biotechnology (Kunming) Co. Ltd, Ciba Biotechnology Innovation Center, Kunming, Yunnan, China
| |
Collapse
|
8
|
Steć A, Targońska M, Jaikishan S, Chen R, Mucha P, Czyrski GS, Jasiecki J, Płoska A, Heinz A, Wiedmer SK, Kalinowski L, Waleron K, Wielgomas B, Dziomba S. Incorporation of doxorubicin into plant-derived nanovesicles: process monitoring and activity assessment. Drug Deliv 2025; 32:2439272. [PMID: 39663752 PMCID: PMC11639057 DOI: 10.1080/10717544.2024.2439272] [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: 08/15/2024] [Revised: 11/27/2024] [Accepted: 12/03/2024] [Indexed: 12/13/2024] Open
Abstract
Extracellular vesicles (EVs) are an experimental class of drug carriers. Alternative sources of EVs are currently being explored to overcome limitations related to their manufacturing from mesenchymal stem cells. In this work, Citrus limon-derived EVs were tested as carriers for the widely used chemotherapeutic drug - doxorubicin (DOX). Capillary electrophoresis (CE) and nanoplasmonic sensing (NPS) were developed for the quality control of DOX-EV preparations. It was found that the CE method enables simultaneous detection of free and incorporated DOX and allows assessing the stability of the preparations and the drug leakage. NPS, on the other hand, demonstrated that DOX is accumulated in the interfacial region of the carrier. The activity of DOX-loaded EVs was tested on HeLa (cervical cancer cells) and HEK293T (human embryonic kidney cells) cell lines. It was found that DOX incorporation into plant-derived EVs virtually does not affect the drug's cytotoxicity to HeLa cells but significantly decreases DOX activity against HEK293T cell line.
Collapse
Affiliation(s)
- Aleksandra Steć
- Department of Toxicology, Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland
| | - Monika Targońska
- Department of Biology and Medical Genetics, Medical University of Gdansk, Gdansk, Poland
| | | | - Rui Chen
- Department of Chemistry, University of Helsinki, Helsinki, Finland
| | - Piotr Mucha
- Faculty of Chemistry, Laboratory of Chemistry of Biologically Active Compounds, University of Gdansk, Gdansk, Poland
| | - Grzegorz S. Czyrski
- Department of Pharmacy, LEO Foundation Center for Cutaneous Drug Delivery, University of Copenhagen, Copenhagen, Denmark
| | - Jacek Jasiecki
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland
| | - Agata Płoska
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland
| | - Andrea Heinz
- Department of Pharmacy, LEO Foundation Center for Cutaneous Drug Delivery, University of Copenhagen, Copenhagen, Denmark
| | | | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics—Fahrenheit Biobank BBMRI.pl, Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland
- Department of Mechanics of Materials and Structures, BioTechMed Centre, Gdansk University of Technology, Gdansk, Poland
| | - Krzysztof Waleron
- Department of Pharmaceutical Microbiology, Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland
| | - Bartosz Wielgomas
- Department of Toxicology, Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland
| | - Szymon Dziomba
- Department of Toxicology, Faculty of Pharmacy, Medical University of Gdansk, Gdansk, Poland
| |
Collapse
|
9
|
Pham DL, Gillette AA, Riendeau J, Wiech K, Guzman EC, Datta R, Skala MC. Perspectives on label-free microscopy of heterogeneous and dynamic biological systems. JOURNAL OF BIOMEDICAL OPTICS 2025; 29:S22702. [PMID: 38434231 PMCID: PMC10903072 DOI: 10.1117/1.jbo.29.s2.s22702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/22/2023] [Accepted: 12/14/2023] [Indexed: 03/05/2024]
Abstract
Significance Advancements in label-free microscopy could provide real-time, non-invasive imaging with unique sources of contrast and automated standardized analysis to characterize heterogeneous and dynamic biological processes. These tools would overcome challenges with widely used methods that are destructive (e.g., histology, flow cytometry) or lack cellular resolution (e.g., plate-based assays, whole animal bioluminescence imaging). Aim This perspective aims to (1) justify the need for label-free microscopy to track heterogeneous cellular functions over time and space within unperturbed systems and (2) recommend improvements regarding instrumentation, image analysis, and image interpretation to address these needs. Approach Three key research areas (cancer research, autoimmune disease, and tissue and cell engineering) are considered to support the need for label-free microscopy to characterize heterogeneity and dynamics within biological systems. Based on the strengths (e.g., multiple sources of molecular contrast, non-invasive monitoring) and weaknesses (e.g., imaging depth, image interpretation) of several label-free microscopy modalities, improvements for future imaging systems are recommended. Conclusion Improvements in instrumentation including strategies that increase resolution and imaging speed, standardization and centralization of image analysis tools, and robust data validation and interpretation will expand the applications of label-free microscopy to study heterogeneous and dynamic biological systems.
Collapse
Affiliation(s)
- Dan L. Pham
- University of Wisconsin—Madison, Department of Biomedical Engineering, Madison, Wisconsin, United States
| | | | | | - Kasia Wiech
- University of Wisconsin—Madison, Department of Biomedical Engineering, Madison, Wisconsin, United States
| | | | - Rupsa Datta
- Morgridge Institute for Research, Madison, Wisconsin, United States
| | - Melissa C. Skala
- University of Wisconsin—Madison, Department of Biomedical Engineering, Madison, Wisconsin, United States
- Morgridge Institute for Research, Madison, Wisconsin, United States
| |
Collapse
|
10
|
Zhang K, Yin Q, Ma Y, Cao M, Li L, Jin X, Leng J. Nanovaccine loaded with seno-antigen target senescent cells to improve metabolic disorders of adipose tissue and cardiac dysfunction. Hum Vaccin Immunother 2025; 21:2479229. [PMID: 40088037 PMCID: PMC11916409 DOI: 10.1080/21645515.2025.2479229] [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: 01/03/2025] [Revised: 02/21/2025] [Accepted: 03/09/2025] [Indexed: 03/17/2025] Open
Abstract
The buildup of senescent cells exacerbates metabolic disorders in adipose tissue and contributes to aging-related cardiac dysfunction. Targeted clearance of senescent cells can markedly ameliorate these aging-related diseases. Here, we developed a novel nanovaccine (GK-NaV) loaded with seno-antigen that is self-assembled from the fusion of cationic protein (K36) and seno-antigen peptide (Gpnmb). The GK-NaV could be highly engulfed by bone marrow-derived dendritic cells (BMDCs) and efficiently present antigens on the cellular surface, thereby promoting DCs maturation and activation of CD8+T cells in vitro. Following subcutaneous immunization, GK-NaV not only exhibited a noticeable antigen depot effect but also markedly activated specific cellular immune responses, enhancing the immunoreactivity and cytotoxic effects of CD8+T cells. Consequently, the targeted anti-aging immunity triggered by GK-NaV demonstrated the ability to selectively eliminate senescent adipocytes and cardiomyocytes in high-fat diet (HFD)-induced progeroid mice, leading to a significant improvement in age-related metabolic disorders in adipose tissue and cardiac dysfunction. Hence, our findings indicate that immunization with GK-NaV targeting seno-antigens may represent a promising strategy for novel senolytic therapies.
Collapse
Affiliation(s)
- Kexin Zhang
- Department of Cadre Ward, The First Hospital of Jilin University, Changchun, China
| | - Qiliang Yin
- Department of Cadre Ward, The First Hospital of Jilin University, Changchun, China
| | - Yucen Ma
- Department of Cadre Ward, The First Hospital of Jilin University, Changchun, China
| | - Mengyu Cao
- Department of Cadre Ward, The First Hospital of Jilin University, Changchun, China
| | - Lingwei Li
- Department of Cadre Ward, The First Hospital of Jilin University, Changchun, China
| | - Xinliang Jin
- Department of General Surgery, First Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Jiyan Leng
- Department of Cadre Ward, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
11
|
Mykchaylova O, Negriyko A, Matvieieva N, Lopatko K, Poyedinok N. Photoregulation of the biosynthetic activity of fungus Inonotus obliquus using colloidal solutions of biogenic metal nanoparticles and low-intensity laser radiation. Bioengineered 2025; 16:2458371. [PMID: 39873594 PMCID: PMC11776471 DOI: 10.1080/21655979.2025.2458371] [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: 06/05/2024] [Revised: 11/10/2024] [Accepted: 11/20/2024] [Indexed: 01/30/2025] Open
Abstract
This article presents new data on the integrated use of colloidal solutions of nanoparticles and low-intensity laser radiation on the biosynthetic activity of the medicinal mushroom Inonotus obliquus in vitro. Traditional mycological methods, colloidal solutions of biogenic metals, and unique photobiological methods have also been used. It was found that colloidal solutions of nanoparticles of all metals used increased the growth characteristics of I. obliquus (55-60%), while irradiation of the fungal inoculum with laser light in a medium with nanoparticles reduced the growth activity of I. obliquus mycelia by 12.3-35.4%. Silver nanoparticles (AgNPs) in a nutrient medium suppressed the biosynthesis of extracellular polysaccharides, whereas laser irradiation in the same medium increased the synthesis of intracellular polysaccharides by 9.7 times. Magnesium nanoparticles (MgNPs) and iron nanoparticles (FeNPs) inhibited the synthesis of intracellular polysaccharides in the mycelial mass of I. obliquus. At the same time, laser irradiation of the inoculum with MgNPs, on the contrary, induced a sharp increase in the amount of polysaccharides in the culture liquid (20 times). Treatment of the inoculum in a medium with nanoparticles with a laser caused an intensification of the synthesis of flavonoids in the mycelial mass and an increase in the synthesis of melanin pigments (25-140%). The results obtained suggest the possibility of the complex use of colloidal solutions of Fe, Ag, and Mg nanoparticles and low-intensity laser radiation as environmentally friendly factors for regulating biosynthetic activity in the biotechnology of cultivating the valuable medicinal mushroom I. obliquus.
Collapse
Affiliation(s)
- Oksana Mykchaylova
- Department of Translational Medical Bioengineering, National Technical University of Ukraine “Igor Sikorsky Kyiv Polytechnic Institute”, Kyiv, Ukraine
- M.G. Kholodny Institute of Botany, National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Anatoliy Negriyko
- Department of Laser Spectroscopy, Institute of Physics of the National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Nadiia Matvieieva
- Institute of Cell Biology and Genetic Engineering, National Academy of Science of Ukraine, Kyiv, Ukraine
| | - Kostiantyn Lopatko
- National Academy of Science of Ukraine, National University of Life and Environmental Sciences, Kyiv, Ukraine
| | - Natalia Poyedinok
- Department of Translational Medical Bioengineering, National Technical University of Ukraine “Igor Sikorsky Kyiv Polytechnic Institute”, Kyiv, Ukraine
| |
Collapse
|
12
|
Girase R, Gujarathi NA, Sukhia A, Kota SSN, Patil TS, Aher AA, Agrawal YO, Ojha S, Sharma C, Goyal SN. Targeted nanoliposomes for precision rheumatoid arthritis therapy: a review on mechanisms and in vivo potential. Drug Deliv 2025; 32:2459772. [PMID: 39891600 PMCID: PMC11789225 DOI: 10.1080/10717544.2025.2459772] [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: 10/04/2024] [Revised: 12/26/2024] [Accepted: 01/23/2025] [Indexed: 02/03/2025] Open
Abstract
Rheumatoid arthritis (RA) is an inflammatory immune-triggered disease that causes synovitis, cartilage degradation, and joint injury. In nanotechnology, conventional liposomes were extensively investigated for RA. However, they frequently undergo rapid clearance, reducing circulation time and therapeutic efficacy. Additionally, their stability in the bloodstream is often compromised, resulting in premature drug release. The current review explores the potential of targeted liposomal-based nanosystems in the treatment of RA. It highlights the pathophysiology of RA, explores selective targeting sites, and elucidates diverse mechanisms of novel liposomal types and their applications. Furthermore, the targeting strategies of pH-sensitive, flexible, surface-modified, PEGylated, acoustic, ROS-mediated, and biofunctionalized liposomes are addressed. Targeted nanoliposomes showed potential in precisely delivering drugs to CD44, SR-A, FR-β, FLS, and toll-like receptors through the high affinity of ligands. In vitro studies interpreted stable release profiles and improved stability. Ex vivo studies on skin demonstrated that ultradeformable and glycerol-conjugated liposomes enhanced drug penetrability. In vivo experiments for liposomal types in the arthritis rat model depicted remarkable efficacy in reducing joint swelling, pro-inflammatory cytokines, and synovial hyperplasia. In conclusion, these targeted liposomes represented a significant leap forward in drug delivery, offering effective therapeutic options for RA. In the future, integrating these advanced liposomes with artificial intelligence, immunotherapy, and precision medicine holds great promise.
Collapse
Affiliation(s)
- Rushikesh Girase
- Shri Vile Parle Kelavani Mandal’s Institute of Pharmacy, Dhule, India
| | | | - Amey Sukhia
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sri Sai Nikitha Kota
- Department of Molecular Pharmaceutics and Drug Delivery, The University of Texas at Austin, Austin, TX, USA
| | | | - Abhijeet A. Aher
- Shri Vile Parle Kelavani Mandal’s Institute of Pharmacy, Dhule, India
| | | | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Charu Sharma
- Department of Internal Medicine, College of Medicine and Health Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Sameer N. Goyal
- Shri Vile Parle Kelavani Mandal’s Institute of Pharmacy, Dhule, India
| |
Collapse
|
13
|
Zuo CJ, Tian J. Advancing the understanding of the role of apoptosis in lung cancer immunotherapy: Global research trends, key themes, and emerging frontiers. Hum Vaccin Immunother 2025; 21:2488074. [PMID: 40186454 PMCID: PMC11980473 DOI: 10.1080/21645515.2025.2488074] [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: 01/24/2025] [Revised: 03/12/2025] [Accepted: 03/30/2025] [Indexed: 04/07/2025] Open
Abstract
Apoptosis is vital for improving the efficacy of lung cancer (LC) immunotherapy by targeting cancer cell elimination. Despite its importance, there is a lack of comprehensive bibliometric studies analyzing global research on apoptosis in LC immunotherapy. This analysis aims to address this gap by highlighting key trends, contributors, and future directions. A total of 969 publications from 1996 to 2024 were extracted from the Web of Science Core Collection. Analysis was conducted using VOSviewer, CiteSpace, and the R package 'bibliometrix.' The study included contributions from 6,894 researchers across 1,469 institutions in 61 countries, with research published in 356 journals. The volume of publications has steadily increased, led by China and the United States, with Sichuan University as the top contributor. The journal Cancers published the most articles, while Cancer Research had the highest co-citations. Yu-Quan Wei was the leading author, and Jemal, A. was the most frequently co-cited. Key research themes include "cell death mechanisms," "immune regulation," "combination therapies," "gene and nanomedicine applications," and "traditional Chinese medicine (TCM)." Future research is likely to focus on "coordinated regulation of multiple cell death pathways," "modulation of the tumor immune microenvironment," "optimization of combination therapies," "novel strategies in gene regulation," and the "integration of TCM" for personalized treatment. This is the first bibliometric analysis on the role of apoptosis in LC immunotherapy, providing an landscape of global research patterns and emerging therapeutic strategies. The findings offer insights to guide future research and optimize treatment approaches.
Collapse
Affiliation(s)
- Chun-Jian Zuo
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jie Tian
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
14
|
Aldali F, Deng C, Nie M, Chen H. Advances in therapies using mesenchymal stem cells and their exosomes for treatment of peripheral nerve injury: state of the art and future perspectives. Neural Regen Res 2025; 20:3151-3171. [PMID: 39435603 PMCID: PMC11881730 DOI: 10.4103/nrr.nrr-d-24-00235] [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: 02/26/2024] [Revised: 07/26/2024] [Accepted: 08/26/2024] [Indexed: 10/23/2024] Open
Abstract
"Peripheral nerve injury" refers to damage or trauma affecting nerves outside the brain and spinal cord. Peripheral nerve injury results in movements or sensation impairments, and represents a serious public health problem. Although severed peripheral nerves have been effectively joined and various therapies have been offered, recovery of sensory or motor functions remains limited, and efficacious therapies for complete repair of a nerve injury remain elusive. The emerging field of mesenchymal stem cells and their exosome-based therapies hold promise for enhancing nerve regeneration and function. Mesenchymal stem cells, as large living cells responsive to the environment, secrete various factors and exosomes. The latter are nano-sized extracellular vesicles containing bioactive molecules such as proteins, microRNA, and messenger RNA derived from parent mesenchymal stem cells. Exosomes have pivotal roles in cell-to-cell communication and nervous tissue function, offering solutions to changes associated with cell-based therapies. Despite ongoing investigations, mesenchymal stem cells and mesenchymal stem cell-derived exosome-based therapies are in the exploratory stage. A comprehensive review of the latest preclinical experiments and clinical trials is essential for deep understanding of therapeutic strategies and for facilitating clinical translation. This review initially explores current investigations of mesenchymal stem cells and mesenchymal stem cell-derived exosomes in peripheral nerve injury, exploring the underlying mechanisms. Subsequently, it provides an overview of the current status of mesenchymal stem cell and exosome-based therapies in clinical trials, followed by a comparative analysis of therapies utilizing mesenchymal stem cells and exosomes. Finally, the review addresses the limitations and challenges associated with use of mesenchymal stem cell-derived exosomes, offering potential solutions and guiding future directions.
Collapse
Affiliation(s)
- Fatima Aldali
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Chunchu Deng
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Mingbo Nie
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Hong Chen
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
15
|
Bartolomé-Nafría A, García-Pardo J, Ventura S. Beyond neurodegeneration: engineering amyloids for biocatalysis. Neural Regen Res 2025; 20:2915-2916. [PMID: 39610101 PMCID: PMC11826452 DOI: 10.4103/nrr.nrr-d-24-00711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/06/2024] [Accepted: 08/27/2024] [Indexed: 11/30/2024] Open
Affiliation(s)
- Andrea Bartolomé-Nafría
- Institut de Biotecnologia i de Biomedicina (IBB) and Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Javier García-Pardo
- Institut de Biotecnologia i de Biomedicina (IBB) and Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
| | - Salvador Ventura
- Institut de Biotecnologia i de Biomedicina (IBB) and Departament de Bioquímica i Biologia Molecular, Universitat Autònoma de Barcelona, Bellaterra, Barcelona, Spain
- Hospital Universitari Parc Taulí, Institut d’Investigació i Innovació Parc Taulí (I3PT-CERCA), Universitat Autònoma de Barcelona, Sabadell, Spain
| |
Collapse
|
16
|
Zhai L, Gao Y, Yang H, Wang H, Liao B, Cheng Y, Liu C, Che J, Xia K, Zhang L, Guan Y. A ROS-Responsive nanoparticle for nuclear gene delivery and autophagy restoration in Parkinson's disease therapy. Biomaterials 2025; 321:123345. [PMID: 40245457 DOI: 10.1016/j.biomaterials.2025.123345] [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: 01/11/2025] [Revised: 03/12/2025] [Accepted: 04/13/2025] [Indexed: 04/19/2025]
Abstract
Parkinson's disease (PD) is characterized by the pathological aggregation of α-synuclein (α-syn) and neuroinflammation. Current gene therapies face challenges in nuclear delivery and resolving pre-existing α-syn aggregates. Here, we developed glucose-and trehalose-functionalized carbonized polymer dots (GT-PCDs) loaded with plasmid DNA (pDNA) for targeted gene delivery and autophagy restoration. The GT-PCDs@pDNA nanoparticles exhibit reactive oxygen species (ROS)-responsive behavior, enabling efficient nuclear entry under oxidative stress conditions. Both in vitro and in vivo studies demonstrated that GT-PCDs@pDNA effectively silenced SNCA gene expression, reduced α-syn aggregates, and restored autophagic flux by promoting transcription factor EB (TFEB) nuclear translocation. Moreover, GT-PCDs@pDNA enhanced blood-brain barrier (BBB) permeability via glucose transporter 1 (Glut-1)-mediated transcytosis, significantly improving motor deficits and reducing neuroinflammation in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD mouse model. This multifunctional nanocarrier system offers a promising strategy for combined gene therapy and autophagy modulation in neurodegenerative diseases.
Collapse
Affiliation(s)
- Limin Zhai
- School of Life Sciences, South China Normal University, Guangzhou, 510631, China
| | - Yifei Gao
- School of Life Sciences, South China Normal University, Guangzhou, 510631, China
| | - Hao Yang
- School of Life Sciences, South China Normal University, Guangzhou, 510631, China
| | - Haoyuan Wang
- School of Life Sciences, South China Normal University, Guangzhou, 510631, China
| | - Beining Liao
- School of Life Sciences, South China Normal University, Guangzhou, 510631, China
| | - Yuxue Cheng
- School of Life Sciences, South China Normal University, Guangzhou, 510631, China
| | - Chao Liu
- School of Life Sciences, South China Normal University, Guangzhou, 510631, China
| | - Jingfeng Che
- School of Life Sciences, South China Normal University, Guangzhou, 510631, China
| | - Kunwen Xia
- School of Life Sciences, South China Normal University, Guangzhou, 510631, China
| | - Lingkun Zhang
- School of Life Sciences, South China Normal University, Guangzhou, 510631, China
| | - Yanqing Guan
- School of Life Sciences, South China Normal University, Guangzhou, 510631, China; Institute for Advanced Materials and Guangdong Provincial Key Laboratory of Quantum Engineering and Quantum Materials South China Academy of Advanced Optoelectronics, South China Normal University, Guangzhou, 510006, China; MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, College of Biophotonics, South China Normal University, Guangzhou, 510631, China.
| |
Collapse
|
17
|
Zhao Z, Zhang Y, Li J, Huang S, Xing G, Zhang K, Ma X, Zhang X, Zhang Y. A remotely controlled nanotherapeutic with immunomodulatory property for MRSA-induced bone infection. Biomaterials 2025; 321:123298. [PMID: 40164042 DOI: 10.1016/j.biomaterials.2025.123298] [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: 12/18/2024] [Revised: 03/10/2025] [Accepted: 03/27/2025] [Indexed: 04/02/2025]
Abstract
Osteomyelitis is a deep bone tissue infection caused by pathogenic microorganisms, with the primary pathogen being methicillin-resistant Staphylococcus aureus (MRSA). Due to the tendency of the infection site to form biofilms that shield drugs and immune cells to kill bacteria, combined with the severe local inflammatory response causing bone tissue destruction, the treatment of osteomyelitis poses a significant challenge. Herein, we developed a remotely controlled nanotherapeutic (TLBA) with immunomodulatory to treat MRSA-induced osteomyelitis. TLBA, combined with baicalin and gold nanorods, is positively charged to actively target and penetrate biofilms. Near-infrared light (808 nm) triggers spatiotemporal, controllable drug release, while bacteria are eliminated through synergistic interaction of non-antibiotic drugs and photothermal therapy, enhancing bactericidal efficiency and minimizing drug resistance. TLBA eliminated nearly 100 % of planktonic bacteria and dispersed 90 % of biofilms under NIR light stimulation. In MRSA-induced osteomyelitis rat models, laser irradiation raised the infection site temperature to 50 °C, effectively eradicating bacteria, promoting M2 macrophage transformation, inhibiting bone inflammation, curbing bone destruction, and fostering bone tissue repair. In summary, TLBA proposes a more comprehensive treatment strategy for the two characteristic pathological changes of bacterial infection and bone tissue damage in osteomyelitis.
Collapse
Affiliation(s)
- Zhe Zhao
- Department of Orthopedics, Tianjin Hospital, No. 406 Jiefangnan Road, Hexi District, Tianjin, 300211, China
| | - Yufei Zhang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Jie Li
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Siyuan Huang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China
| | - Guosheng Xing
- Laboratory of Biochemistry and Molecular Biology, Institute of Orthopedics, Tianjin Hospital, Tianjin, 300050, China
| | - Kai Zhang
- Department of Transfusion, Tianjin Hospital, No. 406 Jiefangnan Road, Hexi District, Tianjin, 300211, China
| | - Xinlong Ma
- Department of Orthopedics, Tianjin Hospital, No. 406 Jiefangnan Road, Hexi District, Tianjin, 300211, China.
| | - Xinge Zhang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin, 300071, China.
| | - Yingze Zhang
- The School of Medicine, Nankai University, Tianjin, 300071, China; Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, China
| |
Collapse
|
18
|
Meng N, Lu J, Zhou J, Yang S, Zhang C, Jia R, Ding Y, Bao Y, Wang J, Ma X, Chen R, Jiang Z, Xie C, Lu L, Lu W. Improved immunocompatibility of active targeting liposomes by attenuating nucleophilic attack of cyclic RGD peptides on complement 3. Biomaterials 2025; 321:123350. [PMID: 40267598 DOI: 10.1016/j.biomaterials.2025.123350] [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: 02/11/2025] [Revised: 03/31/2025] [Accepted: 04/16/2025] [Indexed: 04/25/2025]
Abstract
One of the challenges for the clinical translation of active targeting nanomedicines is the adverse interactions between targeting ligands and blood components. Herein, a novel regularity, which reveals the interactions between cyclic RGD (Arg-Gly-Asp) peptide-modified liposomes and complement components in blood, is reported. As the nucleophilicity of arginine guanidine group within the cyclic RGD-like peptide increases, targeting liposomes potentiate complement cascade via the amplification loop of complement 3 (C3), ultimately leading to accelerated blood clearance, increased deposition in the reticuloendothelial system (RES) organs, enhanced immune responses, and potential side effects. By appropriately reducing the nucleophilicity of guanidine group, cyclic R2 peptide is designed for modification of liposomes to target integrin αvβ3. Compared to the widely used targeting molecule c(RGDyK), R2 eliminates the negative effects of C3 opsonization and specific antibody production, significantly improves the in vivo immunocompatibility of targeting liposomes, and demonstrates superior anti-tumor efficacy in mouse models of orthotopic breast cancer and glioma. Thus, the proposed regularity of interactions between guanidine nucleophilicity and C3, along with the successful application of the low complement activation capacity targeting ligand R2, provides new insights for addressing challenges related to complement activation in the clinical translation of active targeting nanomedicines.
Collapse
Affiliation(s)
- Nana Meng
- Department of Pharmaceutics, School of Pharmacy, Fudan University & National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, Shanghai, 201203, China
| | - Jiasheng Lu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, Shanghai, 201203, China
| | - Jianfen Zhou
- Department of Pharmaceutics, School of Pharmacy, Fudan University & National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, Shanghai, 201203, China
| | - Shengmin Yang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, Shanghai, 201203, China
| | - Chen Zhang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, Shanghai, 201203, China
| | - Ruiyi Jia
- Department of Pharmaceutics, School of Pharmacy, Fudan University & National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, Shanghai, 201203, China
| | - Yuan Ding
- Department of Pharmaceutics, School of Pharmacy, Fudan University & National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, Shanghai, 201203, China
| | - Yanning Bao
- Department of Pharmaceutics, School of Pharmacy, Fudan University & National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, Shanghai, 201203, China
| | - Jun Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, Shanghai, 201203, China
| | - Xiaopei Ma
- Department of Pharmaceutics, School of Pharmacy, Fudan University & National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, Shanghai, 201203, China
| | - Ruohan Chen
- Department of Pharmaceutics, School of Pharmacy, Fudan University & National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, Shanghai, 201203, China
| | - Zhixuan Jiang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, Shanghai, 201203, China
| | - Cao Xie
- Department of Pharmaceutics, School of Pharmacy, Fudan University & National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, Shanghai, 201203, China
| | - Linwei Lu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China; Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, China.
| | - Weiyue Lu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & National Key Laboratory of Advanced Drug Formulations for Overcoming Delivery Barriers, Shanghai, 201203, China; Institutes of Integrative Medicine, Fudan University, Shanghai, 200040, China; Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, and Shanghai Frontiers Science Center for Druggability of Cardiovascular Non-coding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai, 201620, China.
| |
Collapse
|
19
|
Xie R, Fan D, Cheng X, Yin Y, Li H, Wegner SV, Chen F, Zeng W. Living therapeutics: Precision diagnosis and therapy with engineered bacteria. Biomaterials 2025; 321:123342. [PMID: 40252271 DOI: 10.1016/j.biomaterials.2025.123342] [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: 02/02/2025] [Revised: 04/02/2025] [Accepted: 04/12/2025] [Indexed: 04/21/2025]
Abstract
Bacteria-based therapy has emerged as a promising strategy for cancer treatment, offering the potential for targeted tumor delivery, immune activation, and modulation of the tumor microenvironment. However, the unpredictable behavior, safety concerns, and limited efficacy of wild-type bacteria pose significant challenges to their clinical translation. Recent advancements in synthetic biology and chemical engineering have enabled the development of precisely engineered bacterial platforms with enhanced controllability, targeted delivery, and reduced toxicity. This review summarize the current progress of engineered bacteria in cancer therapy. We first introduce the theoretical underpinnings and key advantages of bacterial therapies in cancer. Subsequently, we delve into the applications of genetic engineering and chemical modification techniques to enhance their therapeutic potential. Finally, we address critical challenges and future prospects, with a focus on improving safety and efficacy. This review aims to stimulate further research and provide valuable insights into the development of engineered bacterial therapies for precision oncology.
Collapse
Affiliation(s)
- Ruyan Xie
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China; Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410078, China
| | - Duoyang Fan
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China; Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410078, China
| | - Xiang Cheng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China; Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410078, China
| | - Ying Yin
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China; Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410078, China
| | - Haohan Li
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China; Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410078, China
| | - Seraphine V Wegner
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, 48149, Germany
| | - Fei Chen
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China; Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410078, China.
| | - Wenbin Zeng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China; Hunan Key Laboratory of Diagnostic and Therapeutic Drug Research for Chronic Diseases, Changsha, 410078, China.
| |
Collapse
|
20
|
Hu R, Jiang X, Zhu L, Meng R, Yang R, Sun W, Zhao Z, Lyu Y, Huang R, Xue F, Shi M, Zhou Z, Shen J, Xie C. Overcoming radiation-induced PD-L1 and COX-2 upregulation by nitric oxide gas nanogenerator to sensitize radiotherapy of lung cancer. Biomaterials 2025; 321:123335. [PMID: 40222258 DOI: 10.1016/j.biomaterials.2025.123335] [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: 11/19/2024] [Revised: 03/20/2025] [Accepted: 04/08/2025] [Indexed: 04/15/2025]
Abstract
Currently, certain lung cancer patients exhibit resistance to radiotherapy due to reduced DNA damage under hypoxic conditions and the cytoprotective and immune-resistance effect caused by increased programmed death ligand-1 (PD-L1) and Cyclooxygenase 2 (COX-2) expression after radiotherapy. At present, existing nanoparticles or drugs could hardly effectively, and easily address these obstacles faced by highly effective radiotherapy simultaneously, especially the simultaneous depression of PD-L1 and COX-2. In this study, it is newly proved that some typical nitric oxide (NO) gas donors could co-inhibit PD-L1 and COX-2 expression, revealing the possible not fully proven role of NO in reversing tumor immunotherapy resistance. Then, to realize selective NO generation in tumors, a simple tumor glutathione (GSH) responsive NO gas nanogenerator named SAB-NO nanoparticles was designed and prepared, which was composed of the NO donor Isoamyl Nitrite conjugated with serum albumin (SAB). By doing this, SAB-NO nanoparticles more effectively sensitized radiotherapy through breaking the cytoprotective effects faced by radiotherapy in vitro by generating more DNA damage through reversing tumor hypoxia and impairing the DNA damage repair process through decreasing PD-L1 expression. Then, the combination therapy of SAB-NO nanoparticles and radiotherapy effectively transformed cold tumors into hot ones through avoiding some potential immune-resistance effects induced by radiotherapy treatment alone through PD-L1 and COX-2 co-inhibition. In conclusion, the combined treatment of radiotherapy and SAB-NO nanoparticles finally almost completely suppressed the growth of lung tumors, revealing the novel role of NO donors in sensitizing tumor immunotherapy by avoiding the potential cytoprotective and immune-resistance effects.
Collapse
Affiliation(s)
- Rui Hu
- Department of Radiation and Medical Oncology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Xin Jiang
- Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Lijie Zhu
- The Pharmaceutical Department of the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325030, Zhejiang, China
| | - Rui Meng
- Department of Radiation and Medical Oncology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Rongbo Yang
- Department of Radiation and Medical Oncology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Wenjie Sun
- Department of Radiation and Medical Oncology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China; Zhejiang Engineering Research Center for Innovation and Application of Intelligent Radiotherapy Technology, Zhejiang-Hong Kong Precision Theranostics of Thoracic Tumors Joint Laboratory, Wenzhou Key Laboratory of Basic Science and Translational Research of Radiation Oncology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Zhenzhou Zhao
- Department of Radiation and Medical Oncology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Yuehua Lyu
- Department of Radiation and Medical Oncology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Ruoyuan Huang
- Department of Radiation and Medical Oncology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Fei Xue
- Department of Radiotherapy, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Mengke Shi
- Department of Radiotherapy, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Zaigang Zhou
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China.
| | - Jianliang Shen
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China; Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, Zhejiang, China.
| | - Congying Xie
- Department of Radiation and Medical Oncology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China; Department of Radiation Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China; Zhejiang Engineering Research Center for Innovation and Application of Intelligent Radiotherapy Technology, Zhejiang-Hong Kong Precision Theranostics of Thoracic Tumors Joint Laboratory, Wenzhou Key Laboratory of Basic Science and Translational Research of Radiation Oncology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| |
Collapse
|
21
|
Lu J, Cai J, Zhou Z, Ma J, Han T, Lu N, Zhu L. Gel@CAT-L hydrogel mediates mitochondrial unfolded protein response to regulate reactive oxygen species and mitochondrial homeostasis in osteoarthritis. Biomaterials 2025; 321:123283. [PMID: 40222260 DOI: 10.1016/j.biomaterials.2025.123283] [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] [Revised: 03/03/2025] [Accepted: 03/20/2025] [Indexed: 04/15/2025]
Abstract
OBJECTIVE This study investigates the role of Gelatin-Catalase (Gel@CAT)-L hydrogel in mediating reactive oxygen species (ROS) production and maintaining mitochondrial homeostasis through SIRT3-mediated unfolded protein response (UPRmt), while exploring its involvement in the molecular mechanism of osteoarthritis (OA). METHODS Self-assembled Gel@CAT-L hydrogels were fabricated and characterized using transmission electron microscopy, mechanical testing, external release property evaluation, and oxygen production measurement. Biocompatibility was assessed via live/dead cell staining and CCK8 assays. An OA mouse model was established using destabilization of the medial meniscus (DMM) surgery. X-ray and micro-CT imaging were employed to evaluate the structural integrity of the mouse knee joints, while histological staining was used to assess cartilage degeneration. Immunohistochemistry was performed to analyze the expression of proteins including Col2a1, Aggrecan, MMP13, ADAMTS5, SIRT3, PINK1, and Parkin. Multi-omics analyses-encompassing high-throughput sequencing, proteomics, and metabolomics-were conducted to identify key genes and metabolic pathways targeted by Gel@CAT-L hydrogel intervention in OA. Immunofluorescence techniques were utilized to measure ROS levels, mitochondrial membrane potential, and the expression of SIRT3, PINK1, Parkin, LYSO, LC3B, Col2a1, and MMP13 in primary mouse chondrocytes and mouse knee joints. Flow cytometry was applied to quantify ROS-positive cells. RT-qPCR analysis was conducted to determine mRNA levels of Aggrecan, Col2a1, ADAMTS5, MMP13, SIRT3, mtDNA, HSP60, LONP1, CLPP, and Atf5 in primary mouse chondrocytes, mouse knee joints, and human knee joints. Western blotting was performed to measure protein expression levels of SIRT3, HSP60, LONP1, CLPP, and Atf5 in both primary mouse chondrocytes and mouse knee joints. Additionally, 20 samples each from the control (CON) and OA groups were collected for analysis. Hematoxylin and eosin staining was used to evaluate cartilage degeneration in human knee joints. The Mankin histological scoring system quantified the degree of cartilage degradation, while immunofluorescence analyzed SIRT3 protein expression in human knee joints. RESULTS In vitro experiments demonstrated that self-assembled Gel@CAT-L hydrogels exhibited excellent biodegradability and oxygen-releasing capabilities, providing a stable three-dimensional environment conducive to cell viability and proliferation while reducing ROS levels. Multi-omics analysis identified SIRT3 as a key regulatory gene in mitigating OA and revealed its central role in the UPRmt pathway. Furthermore, Gel@CAT-L was confirmed to regulate mitochondrial homeostasis. Both in vitro experiments and in vivo mouse model studies confirmed that Gel@CAT-L significantly reduced ROS levels and regulated mitochondrial autophagy by activating the SIRT3-mediated UPRmt pathway, thereby improving the pathological state of OA. Clinical trials indicated downregulation of SIRT3 and UPRmt-related proteins in OA patients. CONCLUSION Gel@CAT-L hydrogel activates SIRT3-mediated UPRmt to regulate ROS and mitochondrial homeostasis, providing potential therapeutic benefits for OA.
Collapse
Affiliation(s)
- Jiajia Lu
- Department of Orthopedic Trauma, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China; Department of Orthopedic Trauma, Shanghai Changzheng Hospital, Shanghai, 200434, China
| | - Jiao Cai
- Department of Medical Administration, Shanghai Changzheng Hospital, Shanghai, 200434, China
| | - Zhibin Zhou
- Department of Orthopaedics, General Hospital of Northern Theater Command, Shenyang, 110016, Liaoning, China
| | - Jun Ma
- Department of Orthopedic Trauma, Shanghai Changzheng Hospital, Shanghai, 200434, China; Department of Orthopaedic Trauma, Naval Medical Center of PLA, Naval Medical University, Shanghai, 200001, China
| | - Tianyu Han
- Department of Orthopaedics, General Hospital of Northern Theater Command, Shenyang, 110016, Liaoning, China.
| | - Nan Lu
- Department of Orthopedic Trauma, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China.
| | - Lei Zhu
- Department of Orthopedic Trauma, Shanghai Changzheng Hospital, Shanghai, 200434, China.
| |
Collapse
|
22
|
Liu J, Tang W, Chen L, Zhang Q, Liu T, Qin L, Zhang Y, Chen X. Engineered gold nanoparticles for accurate and full-scale tumor treatment via pH-dependent sequential charge-reversal and copper triggered photothermal-chemodynamic-immunotherapy. Biomaterials 2025; 321:123322. [PMID: 40222257 DOI: 10.1016/j.biomaterials.2025.123322] [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: 10/13/2024] [Revised: 03/12/2025] [Accepted: 04/03/2025] [Indexed: 04/15/2025]
Abstract
Current anti-tumor strategies majorly rely on the targeted delivery of functional nanomedicines to tumor region, neglecting the importance of effective infiltration of these nanomedicines in whole tumor tissue. This process normally causes the quick endocytosis by the tumor cells at surface layer of tumor tissue, resulting in the restriction of the penetration of these nanomedicines and limited therapeutic region, which would not be able to treat the entire tumor tissue. Herein, we prepared a series of engineered gold nanoparticles (Au-MBP NPs) with step-wise charge reversal in different acid environments that could entirely infiltrate into the whole tumor tissue and then perform tumor-specific photothermal-chemodynamic-immunotherapy to achieve the complete and accurate tumor treatment. These Au-MBP NPs consisted of AuNPs, thiol modified piperidine (SH-PD, charge reversal group), thiol modified benzoyl thiourea (SH-BTU, copper chelator) and 11-mercaptoundecanoic acid (MUA) with different proportions. Once these Au-MBP NPs arrived tumor tissue, the decreasing pH values from shallow to deep region of tumor tissue separately induced the charge reversal of these nanoparticles from negative to positive, allowing them to bind with negatively charged tumor cells at designed area to occupy the whole tumor for further therapy. Following with the internalization by tumor cells, these Au-MBP NPs would selectively capture the excessive Cu2+ to decrease the available copper in tumor cells, resulting in the inhibition of tumor metastasis via the copper metabolism blockade. On one hand, the captured Cu2+ also induced the aggregation of Au-MBP NPs, which in situ generated the photothermal agents in tumor cells for tumor-specific photothermal therapy (PTT). On the other hand, the chelated Cu2+ ions were reduced to Cu+, which catalyzed the high concentration of intracellular H2O2 to produce cytotoxic hydroxyl radical (•OH), exerting tumor-specific chemodynamic therapy (CDT). Furthermore, the immune-associated tumor antigens were also generated during PTT and CDT processes via immunogenic cell death (ICD), which further matured the dendritic cells (DCs) and then activated CD4+ and CD8+ T cells to turn on the immunotherapy, resulting in additional anti-tumor and anti-metastasis effects. Both in vitro and in vivo results indicated that these Au-MBP NPs possessed enormous potential for effectively suppressing primary and metastatic tumors.
Collapse
Affiliation(s)
- Jie Liu
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Wenjuan Tang
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China; School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Li Chen
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Qianqian Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Tao Liu
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Longyu Qin
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yanmin Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, 710061, China.
| | - Xin Chen
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
| |
Collapse
|
23
|
Wang Y, Yan T, Cai J, Dou H, Zhu Y, Geng B, Pan D, Shen L. A heterojunction-engineering nanodrug with tumor microenvironment responsiveness for tumor-specific cuproptosis and chemotherapy amplified sono-immunotherapy. Biomaterials 2025; 321:123319. [PMID: 40187098 DOI: 10.1016/j.biomaterials.2025.123319] [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: 02/04/2025] [Revised: 03/24/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Cuproptosis has recently identified as a unique copper-dependent cell death mechanism that may provide new opportunities for improving the therapeutic effect of tumor therapy through triggering efficient adaptive immune responses. However, the poor delivery efficiency and non-tumor-specific release of Cu ions would restrict the potential clinical applications of cuproptosis inducers. Herein, we report for the first time the development of hollow Cu2-xSe nanocubes as the tumor microenvironment (TME)-responsive drug delivery systems and cuproptosis inducers for tumor-specific chemotherapy and cuproptosis. The presence of Cu vacancy endows Cu2-xSe with excellent sonodynamic and chemodynamic activity. The hollow Cu2-xSe nanocubes with TME-responsive degradation behaviors are further utilized to load graphene quantum dot (GQD) nanodrugs to form GQD/Cu2-xSe heterojunctions for achieving tumor-specific chemotherapy. The heterojunction-fabrication GQD/Cu2-xSe exhibits amplified ROS generation capabilities and improved TME regulation ability owing to the optimized electron-hole separation kinetics. More importantly, the significant increase in ROS levels and efficient cuproptosis could reverse the immunosuppressive TME and induce immunogenic cell death that stimulates strong systemic immune responses to eliminate tumors. Collectively, this work presents an innovative strategy for the utilization of TME-responsive cuproptosis inducers for tumor-specific chemotherapy and cuproptosis augmented sono-immunotherapy.
Collapse
Affiliation(s)
- Yang Wang
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Tingting Yan
- Department of Breast Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Jinming Cai
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Hongjing Dou
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yu Zhu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Bijiang Geng
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China.
| | - Dengyu Pan
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China.
| | - Longxiang Shen
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| |
Collapse
|
24
|
Shi H, Yang H, Wu C, Wang S, He S, Chen L, Chan YK, Lai S, Liang K, Deng Y. Glucose-triggered NO-evolving coating bestows orthopedic implants with enhanced anti-bacteria and angiectasis for safeguarding diabetic osseointegration. Biomaterials 2025; 321:123334. [PMID: 40239593 DOI: 10.1016/j.biomaterials.2025.123334] [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: 02/19/2025] [Revised: 04/08/2025] [Accepted: 04/08/2025] [Indexed: 04/18/2025]
Abstract
As a common chronic metabolic disease, diabetes mellitus (DM) features a hyperglycemic micromilieu around implants, resulting in the critical implantation failure and high complications such as peri-implantitis and angiectasis disorder. To address the plaguing issue, we devise and develop a glucose-unlocked NO-evolving orthopedic implant consisted of polyetheretherketone (PEEK), glucose oxidase (GOx) and l-arginine (Arg) with enhanced angiogenesis for boosting diabetic osseointegration. Upon hyperglycemic niche, GOx on implants catalytically exhaust glucose to H2O2, which immediately reacts with Arg to in situ liberate nitric oxide (NO), resulting in enhanced angiogenesis and angiectasis around PEEK implant. Besides, the engineered implant exhibits great anti-bacterial properties against both Gram-positive and Gram-negative bacteria, as well as fortifies osteogenicity of osteoblasts in terms of cell proliferation, alkaline phosphatase activity and calcium matrix mineralization. Intriguingly, in vivo evaluations utilizing diabetic infectious bone defect models of rat further authenticate that the engineered implants substantially augment bone remodeling and osseointegration at weeks 4 and 8 through dampening pathogens, anti-inflammatory as well as promoting angiectasis. Altogether, this work proposed a new tactic to remedy stalled diabetic osseointegration with hyperglycemic micromilieu-responsive therapeutic gas-evolving orthopedic implants.
Collapse
Affiliation(s)
- Hongxing Shi
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
| | - Hao Yang
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
| | - Chao Wu
- Institute of Digital Medicine, Zigong Academy of Big Data for Medical Science and Artificial Intelligence, Department of Orthopedics, Zigong Fourth People's Hospital, Zigong, 643000, China
| | - Song Wang
- Department of Orthopaedics, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Shuai He
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
| | - Lin Chen
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
| | - Yau Kei Chan
- Department of Ophthalmology, The University of Hong Kong, 999077, Hong Kong, China
| | - Shuangquan Lai
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
| | - Kunneng Liang
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China; Department of Cardiology and Endodontics, State Key Laboratory of Oral Disease, West China Hospital, Sichuan University, Chengdu, 610065, China.
| | - Yi Deng
- School of Chemical Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China; National Key Laboratory of Advanced Polymer Materials, Sichuan University, Chengdu, 610065, China; Department of Mechanical Engineering, The University of Hong Kong, 999077, Hong Kong, China.
| |
Collapse
|
25
|
Xue Y, Zhang L, Chen J, Ma D, Zhang Y, Han Y. An "all-in-one" therapeutic platform for programmed antibiosis, immunoregulation and neuroangiogenesis to accelerate diabetic wound healing. Biomaterials 2025; 321:123293. [PMID: 40179815 DOI: 10.1016/j.biomaterials.2025.123293] [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] [Revised: 03/01/2025] [Accepted: 03/26/2025] [Indexed: 04/05/2025]
Abstract
Pathological microenvironment of diabetes induces a high risk of bacterial invasion, aggressive inflammatory response, and hindered neuroangiogenesis, leading to retarded ulcer healing. To address this, an "all-in-one" therapeutic platform, named MZZ, was constructed by loading maltodextrin onto a MOF-on-MOF structure (with ZIF-67 as the core and ZIF-8 as the shell) through a hybrid process of solvent treatment and electrostatic adsorption. Maltodextrin acts as a target to bind surrounding bacteria, and ZIF-8 as well as ZIF-67 responsively release Zn and Co ions, which not only kill most bacteria, but also improve the phagocytosis and xenophagy of M1 macrophages by up-regulating the expression levels of ATG5, Bcl1 and FLT4, helping the residual bacterial clearance. In inflammatory stage, MZZ scavenges extracellular and intracellular ROS by valence transition between Co2+ and Co3+, and promote M1 macrophages to transform into M2 phenotype. In tissue reconstruction stage, the synergistic effect of Zn and Co ions as well as cytokines secreted by macrophages up-regulates cell vitality and biofunctions of endotheliocytes, neurocytes and fibroblasts. The programmed effects of MZZ on antibiosis, anti-inflammatory and neuroangiogenesis to accelerate wound repair are further confirmed in an infected diabetic model, and this "all-in-one" platform shows great clinical application potential.
Collapse
Affiliation(s)
- Yang Xue
- State-key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an, 710049, China; Department of Orthopaedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Lan Zhang
- State-key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an, 710049, China.
| | - Jun Chen
- Department of Osteology, Xi'an People's Hospital (Xi'an No. 4 Hospital), Xi'an, 710100, China
| | - Dayan Ma
- State-key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Yingang Zhang
- Department of Orthopaedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Yong Han
- State-key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an, 710049, China; Department of Orthopaedics, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| |
Collapse
|
26
|
Moyo B, Brown LBC, Khondaker II, Bao G. Engineering adeno-associated viral vectors for CRISPR/Cas based in vivo therapeutic genome editing. Biomaterials 2025; 321:123314. [PMID: 40203649 DOI: 10.1016/j.biomaterials.2025.123314] [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: 12/23/2024] [Revised: 03/30/2025] [Accepted: 04/01/2025] [Indexed: 04/11/2025]
Abstract
The recent approval of the first gene editing therapy for sickle cell disease and transfusion-dependent beta-thalassemia by the U.S. Food and Drug Administration (FDA) demonstrates the immense potential of CRISPR (clustered regularly interspaced short palindromic repeats) technologies to treat patients with genetic disorders that were previously considered incurable. While significant advancements have been made with ex vivo gene editing approaches, the development of in vivo CRISPR/Cas gene editing therapies has not progressed as rapidly due to significant challenges in achieving highly efficient and specific in vivo delivery. Adeno-associated viral (AAV) vectors have shown great promise in clinical trials as vehicles for delivering therapeutic transgenes and other cargos but currently face multiple limitations for effective delivery of gene editing machineries. This review elucidates these challenges and highlights the latest engineering strategies aimed at improving the efficiency, specificity, and safety profiles of AAV-packaged CRISPR/Cas systems (AAV-CRISPR) to enhance their clinical utility.
Collapse
Affiliation(s)
- Buhle Moyo
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
| | - Lucas B C Brown
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA; Graduate Program in Systems, Synthetic, and Physical Biology, Rice University, Houston, TX, 77030, USA
| | - Ishika I Khondaker
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA; Medical Scientist Training Program, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Gang Bao
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA.
| |
Collapse
|
27
|
Liu S, Ren Z, Yan M, Ye W, Hu Y. Strategies to enhance the penetration of nanomedicine in solid tumors. Biomaterials 2025; 321:123315. [PMID: 40185056 DOI: 10.1016/j.biomaterials.2025.123315] [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: 12/22/2024] [Revised: 03/16/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Nanomedicine was previously regarded as a promising solution in the battle against cancer. Over the past few decades, extensive research has been conducted to exploit nanomedicine for overcoming tumors. Unfortunately, despite these efforts, nanomedicine has not yet demonstrated its ability to cure tumors, and the research on nanomedicine has reached a bottleneck. For a significant period of time, drug delivery strategies have primarily focused on targeting nanomedicine delivery to tumors while neglecting its redistribution within solid tumors. The uneven distribution of nanomedicine within solid tumors results in limited therapeutic effects on most tumor cells and significantly hampers the efficiency of drug delivery and treatment outcomes. Therefore, this review discusses the challenges faced by nanomedicine in penetrating solid tumors and provides an overview of current nanotechnology strategies (alleviating penetration resistance, size regulation, tumor cell transport, and nanomotors) that facilitate enhanced penetration of nanomedicine into solid tumors. Additionally, we discussed the potential role of nanobionics in promoting effective penetration of nanomedicine.
Collapse
Affiliation(s)
- Sen Liu
- Jiangsu Provincial Engineering Research Center for Biomedical Materials and Advanced Medical Devices, Faculty of Mechanical and Material Engineering, Huaiyin Institute of Technology, Huaian, 223003, China
| | - Zhendong Ren
- Faculty of Chemical Engineering, Huaiyin Institute of Technology, Huaian, 223003, China
| | - Manqi Yan
- College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Wei Ye
- Faculty of Chemical Engineering, Huaiyin Institute of Technology, Huaian, 223003, China.
| | - Yong Hu
- College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu, 210023, China.
| |
Collapse
|
28
|
Cho H, Ju H, Ahn Y, Jang J, Cho J, Park E, Kang SM, Lee J, Seo D, Baek MC, Yea K. Engineered extracellular vesicles with surface FGF21 and enclosed miR-223 for treating metabolic dysfunction-associated steatohepatitis. Biomaterials 2025; 321:123321. [PMID: 40209593 DOI: 10.1016/j.biomaterials.2025.123321] [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: 10/28/2024] [Revised: 02/22/2025] [Accepted: 04/03/2025] [Indexed: 04/12/2025]
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is a progressive liver disorder with a complex pathogenesis that requires combination therapies rather than monotherapies. Extracellular vesicles (EVs) exhibit inherently efficient delivery to the liver and can be engineered to carry various therapeutic substances, making them promising agents. In this study, EVs were engineered to display fibroblast growth factor 21 (FGF21) on their surface and encapsulate miR-223 (223/F-EVs), aiming to improve steatosis and alleviate inflammation and fibrosis, respectively. Introducing the 223/F-EVs into human liver cell lines significantly reduced both basal and induced levels of lipid storage, inflammation, and fibrosis markers. Furthermore, using an FGF21-blocking antibody or miR-223 inhibitor effectively diminished the efficacy of the 223/F-EVs, confirming the essential roles of FGF21 and miR-223 in these processes. In a Choline-Deficient, l-Amino acid-defined, High-Fat Diet (CDAHFD)-fed mouse model, intravenously administered 223/F-EVs demonstrated liver-preferential delivery and a marked reduction in the MASH phenotype without compromising bone density, unlike conventional FGF21 treatment. Collectively, 223/F-EVs convey FGF21 and miR-223 exclusively to the liver, offering strategic advantages by mitigating MASH progression via multiple pathways. This study lays a solid foundation for further investigation of engineered EVs as a transformative therapeutic approach for treating MASH.
Collapse
Affiliation(s)
- Hanchae Cho
- Department of Biomedical Science, Kyungpook National University School of Medicine, Daegu, 41944, Republic of Korea
| | - Hyunji Ju
- Department of Molecular Medicine, CMRI, Kyungpook National University School of Medicine, Daegu, 41944, Republic of Korea
| | - Yongdeok Ahn
- Department of Physics and Chemistry, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 42988, Republic of Korea
| | - Juhee Jang
- Department of Physics and Chemistry, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 42988, Republic of Korea
| | - Juhyeong Cho
- Department of Physics and Chemistry, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 42988, Republic of Korea
| | - Eunju Park
- Department of New Biology, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 42988, Republic of Korea
| | - Sung-Min Kang
- Department of Molecular Medicine, CMRI, Kyungpook National University School of Medicine, Daegu, 41944, Republic of Korea
| | - Jaemin Lee
- Department of New Biology, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 42988, Republic of Korea
| | - Daeha Seo
- Department of Physics and Chemistry, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 42988, Republic of Korea
| | - Moon-Chang Baek
- Department of Molecular Medicine, CMRI, Kyungpook National University School of Medicine, Daegu, 41944, Republic of Korea.
| | - Kyungmoo Yea
- Department of New Biology, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 42988, Republic of Korea; New Biology Research Center, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 43024, Republic of Korea.
| |
Collapse
|
29
|
Liu Y, Ren H, Wu Z, Wu Y, Zhou X, Ji D. Advances in the application of smart materials in the treatment of ophthalmic diseases. Biomaterials 2025; 321:123316. [PMID: 40194319 DOI: 10.1016/j.biomaterials.2025.123316] [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: 12/15/2024] [Revised: 03/30/2025] [Accepted: 04/01/2025] [Indexed: 04/09/2025]
Abstract
Smart materials dynamically sense and respond to physiological signals like reactive oxygen species (ROS), pH, and light, surpassing traditional materials such as poly(lactic-co-glycolic acid), which have high drug loss rates and limited spatiotemporal control. These innovative materials offer new strategies for ophthalmic treatments, with core advantages including targeted delivery via ROS-sensitive nanocarriers, precise regulation through microvalves, and multifunctional integration, such as glucose-responsive contact lenses that create a "sensing-treatment" loop. However, challenges remain, like pathological microenvironment interference with material response specificity, and the need to address long-term biocompatibility and energy dependence issues. This article systematically examines three key treatment barriers: the blood-ocular barrier, immune rejection, and physiological fluctuations, while reviewing innovative smart material design strategies. Future research should focus on biomimetic interface engineering, for example, cornea mimicking nanostructures, AI-driven dynamic optimization like causal network-regulated drug release, and multidisciplinary approaches combining gene editing with smart materials. These efforts aim to shift from structural replacement to physiological function simulation, enabling precise treatment of ophthalmic diseases. Clinical translation must balance innovation with safety, prioritizing clinical value to ensure reliable, widespread application of smart materials in ophthalmology.
Collapse
Affiliation(s)
- Yida Liu
- Basic Medical College of the Army Medical University (Third Military Medical University), Chongqing, China
| | - Hong Ren
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Ophthalmology, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhenkai Wu
- Department of Ophthalmology, Changde Hospital, Xiangya School of Medicine, Central South University(The First People's Hospital of Changde City), Changde, Hunan Province, China
| | - Yukun Wu
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Ophthalmology, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xuezhi Zhou
- Department of Ophthalmology, Xiangya Hospital, Central South University, Changsha, China; Hunan Key Laboratory of Ophthalmology, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Dan Ji
- Hunan Key Laboratory of Ophthalmology, Central South University, Changsha, China; Department of Ophthalmology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University, Changsha, China.
| |
Collapse
|
30
|
Chen R, Bhavsar C, Lourie R, Li S, Wu SY. Development of an innovative extracellular vesicle mimetic delivery platform for efficient miRNA delivery to tumours. Biomaterials 2025; 321:123282. [PMID: 40156978 DOI: 10.1016/j.biomaterials.2025.123282] [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: 12/04/2024] [Revised: 03/20/2025] [Accepted: 03/20/2025] [Indexed: 04/01/2025]
Abstract
Extracellular vesicles (EVs) display high degree of tissue tropism and therefore represent promising carriers for tissue-specific delivery of genes or drugs for the treatment of human diseases. However, current approaches for the loading of therapeutics into EVs have low entrapment efficiency and also do not adequately deplete endogenous EV content; thus, more effective approaches are needed. Here, we report an innovative EXtraCElluar vesicle surface Ligand-NanoParticles (EXCEL NPs), generated by transferring moieties of EVs onto the surface of synthetic nanoparticles. EXCEL NPs facilitate the efficient entrapment of therapeutics (89 % efficiency) and are completely devoid of pre-existing unwanted EV internal content. Importantly, we show that EXCEL NPs formulated using EVs derived from endothelial cells, astrocytes and macrophages retain the delivery characteristics of the original EVs. Using miRNA-146a as a model anti-cancer therapeutic, we further demonstrated successful delivery of miRNA-146a to IG10 orthotopic ovarian tumours in immune competent mice using EXCEL NPs formulated with macrophage-derived EVs. Our findings establish a new clinically translatable approach to leverage characteristics of endogenous EVs for therapeutic delivery. The versatility of the platform enables future application to different target cell types and therapeutic modalities.
Collapse
Affiliation(s)
- Rui Chen
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Chintan Bhavsar
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Rohan Lourie
- Mater Health Services, South Brisbane, QLD, 4101, Australia
| | - Shuying Li
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Sherry Y Wu
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
31
|
Wang K, Jiang S, Wang W, Chen W, Kai T. Dual-miRNA guided in-vivo imaging and multimodal nanomedicine approaches for precise hepatocellular carcinoma differentiation and synergistic cancer theranostics using DNA hairpins and dual-ligand functionalized zirconium-MOF nanohybrids. Biomaterials 2025; 321:123330. [PMID: 40222259 DOI: 10.1016/j.biomaterials.2025.123330] [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: 02/26/2025] [Accepted: 04/07/2025] [Indexed: 04/15/2025]
Abstract
As one of the most common and heterogeneous liver malignancies, hepatocellular carcinoma (HCC) remains a significant clinical challenge due to the lack of biomarkers for early diagnosis, challenges in accurate subtyping, and limitations of current therapeutic strategies with poor efficacy. Herein, based on DNA hairpin probes and dual-ligand zirconium (Zr)-based metal-organic frameworks (DMOFs), the multifunctional nanohybrids (DMOF@MnCO@CuS@Hairpin probe, DMCH) were developed to overcome these diagnostic and therapeutic obstacles. Two improved DNA molecular beacons and APE1 enzyme within HCC cells were utilized for sensitive miRNA imaging in vivo with high accuracy to differentiate HCC subtypes precisely. Furthermore, this "all-in-one" theranostic platform not only facilitates the generation of active oxygen species and conversion of near-infrared light into heat, but also releases carbon monoxide to inhibit the expression of HSP70 protein to improve photothermal (PTT) therapy efficiency during laser radiation, which enables PTT, photodynamic (PDT), chemodynamic (CDT), and gas therapy (GAT) for HCC treatment simultaneously. The developed nano-theranostics platform provides a novel way for efficient early screening, diagnosis, and intervention of HCC, and paves the path for future "bench-to-bedside" design of theranostics.
Collapse
Affiliation(s)
- Kun Wang
- Xiangya School of Public Health, Central South University, Changsha, Hunan, 410013, China
| | - Siyu Jiang
- Xiangya School of Public Health, Central South University, Changsha, Hunan, 410013, China
| | - Wanli Wang
- Xiangya School of Public Health, Central South University, Changsha, Hunan, 410013, China
| | - Wansong Chen
- College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, 410083, China
| | - Tianhan Kai
- Xiangya School of Public Health, Central South University, Changsha, Hunan, 410013, China; Furong Laboratory, Changsha, Hunan, 410008, China.
| |
Collapse
|
32
|
Lu Z, Yan J, Zeng J, Zhang R, Xu M, Liu J, Sun L, Zu G, Chen X, Zhang Y, Pei R, Cao Y. Time-resolved T 1 and T 2 contrast for enhanced accuracy in MRI tumor detection. Biomaterials 2025; 321:123313. [PMID: 40187097 DOI: 10.1016/j.biomaterials.2025.123313] [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: 01/26/2025] [Revised: 03/30/2025] [Accepted: 03/31/2025] [Indexed: 04/07/2025]
Abstract
Stimuli-responsive contrast agents (CAs) have shown great promise in enhancing magnetic resonance imaging (MRI) for more accurate tumor diagnosis. However, current CAs still face challenges in achieving high accuracy due to their low specificity and contrast signals being confounded by potential endogenous MRI artifacts. Herein, an extremely small iron oxide nanoparticle (ESIONP)-based smart responsive MRI contrast agent (LESPH) is proposed, which is meticulously designed with sequential dual biochemical stimuli-initiated, time-resolved T1 and T2 contrast presentation, ensuring high tumor specificity while minimizing interference from endogenous artifacts. LESPH is constructed using emulsion solvent evaporation by assembling poly(2-(hexamethyleneimino) ethyl methacrylate) terminally conjugated with a disulfide bond-linked catechol group (DSPH)-modified ESIONPs, with lauryl betaine serving as a surfactant. When LESPH undergoes sequential responses to the weak acidity and high-concentration glutathione (GSH) in the tumor microenvironment, it experiences an extremely rapid transition from sparse ESIONP assemblies to dispersed ESIONPs, followed by a slower transition to closely aggregated ones, concomitantly providing distinguishable brightening and darkening contrast enhancement at the tumor location on different time scales. By virtue of its sequential dual responsiveness and time-resolved distinguishable contrast enhancements, LESPH successfully detects tumors with extremely high accuracy, providing a novel paradigm for the precise medical diagnosis of cancer.
Collapse
Affiliation(s)
- Zhongzhong Lu
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230000, China; CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Jincong Yan
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230000, China; CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Jianxian Zeng
- Department of Radiology, First Affiliated Hospital of Soochow University, Suzhou, 215026, China
| | - Ruihao Zhang
- Department of Oncology, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, 215026, China
| | - Mingsheng Xu
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Jihuan Liu
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Lina Sun
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Guangyue Zu
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Xiaomin Chen
- Department of Stomatology, Traditional Chinese Medicine Hospital of Kunshan, Kunshan, 215300, China
| | - Ye Zhang
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China.
| | - Renjun Pei
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230000, China; CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China.
| | - Yi Cao
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230000, China; CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China.
| |
Collapse
|
33
|
Lan X, Johnston E, Ning T, Chen G, Haglund L, Li J. Immunomodulatory bioadhesive technologies. Biomaterials 2025; 321:123274. [PMID: 40156979 DOI: 10.1016/j.biomaterials.2025.123274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/20/2025] [Accepted: 03/17/2025] [Indexed: 04/01/2025]
Abstract
Bioadhesives have found significant use in medicine and engineering, particularly for wound care, tissue engineering, and surgical applications. Compared to traditional wound closure methods such as sutures and staples, bioadhesives offer advantages, including reduced tissue damage, enhanced healing, and ease of implementation. Recent progress highlights the synergy of bioadhesives and immunoengineering strategies, leading to immunomodulatory bioadhesives capable of modulating immune responses at local sites where bioadhesives are applied. They foster favorable therapeutic outcomes such as reduced inflammation in wounds and implants or enhanced local immune responses to improve cancer therapy efficacy. The dual functionalities of bioadhesion and immunomodulation benefit wound management, tissue regeneration, implantable medical devices, and post-surgical cancer management. This review delves into the interplay between bioadhesion and immunomodulation, highlighting the mechanobiological coupling involved. Key areas of focus include the modulation of immune responses through chemical and physical strategies, as well as the application of these bioadhesives in wound healing and cancer treatment. Discussed are remaining challenges such as achieving long-term stability and effectiveness, necessitating further research to fully harness the clinical potential of immunomodulatory bioadhesives.
Collapse
Affiliation(s)
- Xiaoyi Lan
- Department of Surgery, McGill University, 1650 Cedar Avenue, Montreal, Quebec, H3G 1A3, Canada; Department of Mechanical Engineering, McGill University, 817 Sherbrooke St W, Montreal, Quebec, H3A 0C3, Canada
| | - Evan Johnston
- Department of Mechanical Engineering, McGill University, 817 Sherbrooke St W, Montreal, Quebec, H3A 0C3, Canada
| | - Tianqin Ning
- Department of Mechanical Engineering, McGill University, 817 Sherbrooke St W, Montreal, Quebec, H3A 0C3, Canada; Department of Biomedical Engineering, McGill University, 3775 Rue University, Montreal, Quebec, H3A 2B4, Canada
| | - Guojun Chen
- Department of Biomedical Engineering, McGill University, 3775 Rue University, Montreal, Quebec, H3A 2B4, Canada; Rosalind & Morris Goodman Cancer Institute, McGill University, 1160 Pine Ave W, Montreal, Quebec, H3A 1A3, Canada
| | - Lisbet Haglund
- Department of Surgery, McGill University, 1650 Cedar Avenue, Montreal, Quebec, H3G 1A3, Canada; Shriners Hospital for Children, 1003 Decarie Blvd, Montreal, Quebec, H4A 0A9, Canada.
| | - Jianyu Li
- Department of Surgery, McGill University, 1650 Cedar Avenue, Montreal, Quebec, H3G 1A3, Canada; Department of Mechanical Engineering, McGill University, 817 Sherbrooke St W, Montreal, Quebec, H3A 0C3, Canada; Department of Biomedical Engineering, McGill University, 3775 Rue University, Montreal, Quebec, H3A 2B4, Canada.
| |
Collapse
|
34
|
Zhang P, Ran Y, Han L, Li Y, Tian W, Sun X, Jiao M, Jing L, Luo X. Nanomaterial technologies for precision diagnosis and treatment of brain hemorrhage. Biomaterials 2025; 321:123269. [PMID: 40174300 DOI: 10.1016/j.biomaterials.2025.123269] [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: 12/02/2024] [Revised: 02/19/2025] [Accepted: 03/17/2025] [Indexed: 04/04/2025]
Abstract
Brain hemorrhage events present complex clinical challenges due to their rapid progression and the intricate interplay of oxidative stress, inflammation, and neuronal damage. Traditional diagnostic and therapeutic approaches often struggle to meet the demands for timely and effective intervention. This review explores the cutting-edge role of nanomaterials in transforming cerebral hemorrhage management, focusing on both diagnostic and therapeutic advancements. Nanomaterial-enabled imaging techniques, such as optical imaging, magnetic resonance imaging, and magnetic particle imaging, significantly enhance the accuracy of hemorrhage detection by providing real-time, high-resolution assessments of blood-brain barrier (BBB) integrity, cerebral perfusion, and hemorrhage progression, which is critical for guiding intervention strategies. On the therapeutic front, nanomaterial-based systems enable the precise delivery of drugs and bioactive molecules, fostering neural repair and functional recovery while minimizing systemic side effects. Furthermore, multifunctional nanomaterials not only address the primary injury but also offer precise control over secondary injuries, such as edema and oxidative stress. Their ability to enhance neuroprotection, prevent re-bleeding, and stimulate brain tissue regeneration provides a holistic approach and marks a significant advancement in brain hemorrhage therapy. As the field continues to advance, nanotechnology is set to fundamentally reshape the clinical management and long-term outcomes of brain hemorrhages, presenting a paradigm shift towards personalized and highly effective neurological care.
Collapse
Affiliation(s)
- Peisen Zhang
- Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Zhengzhou Road 53, Qingdao, 266042, China
| | - Yi'an Ran
- Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Zhengzhou Road 53, Qingdao, 266042, China
| | - Lei Han
- Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Zhengzhou Road 53, Qingdao, 266042, China
| | - Yao Li
- Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Zhengzhou Road 53, Qingdao, 266042, China
| | - Wanru Tian
- Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Zhengzhou Road 53, Qingdao, 266042, China
| | - Xiao Sun
- Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Zhengzhou Road 53, Qingdao, 266042, China
| | - Mingxia Jiao
- Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Zhengzhou Road 53, Qingdao, 266042, China.
| | - Lihong Jing
- CAS Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Beijing National Laboratory for Molecular Sciences, Center for Carbon Neutral Chemistry, Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing, 100190, China.
| | - Xiliang Luo
- Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Zhengzhou Road 53, Qingdao, 266042, China.
| |
Collapse
|
35
|
Lyu Y, Yin Q, Liao X, Xie Y, Yang H, Cui Y, Han Y, Yao K, Wang C, Shentu X. Aminated fullerene for comprehensive dry eye therapy: Promoting epithelial-barrier reconstruction and nerve regeneration by suppressing oxidation and inflammation. Biomaterials 2025; 321:123329. [PMID: 40273474 DOI: 10.1016/j.biomaterials.2025.123329] [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: 03/14/2025] [Accepted: 04/07/2025] [Indexed: 04/26/2025]
Abstract
Dry eye disease (DED) affects up to 50 % of the global population, leading to serious discomforts that affect patients' quality of life. In the multifactorial etiology of DED, oxidative stress is at the core, initiating a sequence of inflammatory responses and surface damage via a vicious cycle. However, current therapies merely have a narrow focus on inflammation. In this study, we developed a novel antioxidative eye drop, ethylenediamine (EDA)-modified C70 fullerene derivatives (abbreviated as FN-EDA), to break this vicious cycle. FN-EDA was successfully synthesized by modifying C70 fullerene with multiple ethylenediamine (EDA) groups, resulting in enhanced water solubility and a positive charge. This modification significantly improved ocular surface retention time, cellular uptake, and lysosomal escape in vitro. Therapeutically, FN-EDA significantly alleviated dry eye disease (DED) in a mouse model. It reduced corneal epithelial damage by 3.8-fold compared to 0.05 % cyclosporine A (CsA) and restored tear secretion to approximately 65 % of the normal level. Mechanistically, both in vivo and in vitro results demonstrate that FN-EDA is endowed with superior biological activity in effectively scavenging excessive oxidative stress, down-regulating proinflammatory cytokines expression, and promoting epithelial barrier reconstruction, even recovering corneal innervation. Thus, our findings open an avenue to make this multi-functional eye drop a promising candidate for DED.
Collapse
Affiliation(s)
- Ying Lyu
- Zhejiang University, Eye Center of Second Affiliated Hospital, School of Medicine, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, China.
| | - Qichuan Yin
- Zhejiang University, Eye Center of Second Affiliated Hospital, School of Medicine, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, China.
| | - Xiaodan Liao
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China.
| | - Youxuan Xie
- Zhejiang University, Eye Center of Second Affiliated Hospital, School of Medicine, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, China.
| | - Hao Yang
- Zhejiang University, Eye Center of Second Affiliated Hospital, School of Medicine, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, China.
| | - Yilei Cui
- Zhejiang University, Eye Center of Second Affiliated Hospital, School of Medicine, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, China.
| | - Yuqi Han
- Zhejiang University, Eye Center of Second Affiliated Hospital, School of Medicine, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, China.
| | - Ke Yao
- Zhejiang University, Eye Center of Second Affiliated Hospital, School of Medicine, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, China.
| | - Chunru Wang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, Nanostructure and Nanotechnology, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China.
| | - Xingchao Shentu
- Zhejiang University, Eye Center of Second Affiliated Hospital, School of Medicine, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou, 310009, China.
| |
Collapse
|
36
|
Yang Y, Zeng Q, Zhao C, Shi J, Wang W, Liang Y, Li C, Guan Q, Chen B, Li W. Metal-free antioxidant nanozyme incorporating bioactive hydrogel as an antioxidant scaffold for accelerating bone reconstruction. Biomaterials 2025; 320:123285. [PMID: 40127506 DOI: 10.1016/j.biomaterials.2025.123285] [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: 12/17/2024] [Revised: 01/30/2025] [Accepted: 03/20/2025] [Indexed: 03/26/2025]
Abstract
Oxidative stress at bone defect sites mediates inflammation and even osteoblast apoptosis, severely hindering the repair process. While current antioxidant bone tissue engineering (BTE) scaffolds lack broad-spectrum reactive oxygen species (ROS) scavenging capability and structure-activity elucidation. Herein, we report a three-dimensional nitrogen-doped carbon antioxidant nanozyme (ZIFC) derived from metal-organic frameworks, which exhibits cascading superoxide dismutase- and catalase-like activities, along with the ability to scavenge other harmful free radicals. Through the experimental studies and theoretical calculations, we reveal that the catalase-like activity arises from the synergistic catalytic interaction between graphitized pyridinic nitrogen and its adjacent carbon atom. Moreover, hybrid double network hydrogel integrated with ZIFC is utilized to construct composite scaffold (Gel/ZIFC) by 3D printing. In vivo transcriptome analysis confirms that Gel/ZIFC can rapidly activate antioxidant defense system and suppress local inflammation under oxidative stress microenvironment, thereby protecting cells from oxidative damage. Subsequently, owing to the unique osteoinductive property of carbon nanomaterials and the osteoconductive property of 3D-printed scaffold, Gel/ZIFC composite scaffold exhibits desirable bone repair efficacy. The elucidation of structure-activity relationship and therapeutic mechanism provides new insights and guidance for devising antioxidant BTE scaffolds, and demonstrates their feasibility for clinical application.
Collapse
Affiliation(s)
- Yang Yang
- State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Advanced Energy Materials Chemistry (Ministry of Education), College of Chemistry, Nankai University, Tianjin 300071, PR China
| | - Qianrui Zeng
- State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Advanced Energy Materials Chemistry (Ministry of Education), College of Chemistry, Nankai University, Tianjin 300071, PR China
| | - Chaoyue Zhao
- Ningbo Key Laboratory of Agricultural Germplasm Resources Mining and Environmental Regulation, College of Science and Technology, Ningbo University, Ningbo, 315000, PR China
| | - Jie Shi
- The School of Medicine, Nankai University, Tianjin 300071, PR China
| | - Wanmeng Wang
- Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin Medical University Institute of Stomatology, Tianjin 300070, PR China
| | - Yunkai Liang
- Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin Medical University Institute of Stomatology, Tianjin 300070, PR China
| | - Changyi Li
- Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin Medical University Institute of Stomatology, Tianjin 300070, PR China
| | - Qingxin Guan
- State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Advanced Energy Materials Chemistry (Ministry of Education), College of Chemistry, Nankai University, Tianjin 300071, PR China.
| | - Bo Chen
- Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin Medical University Institute of Stomatology, Tianjin 300070, PR China.
| | - Wei Li
- State Key Laboratory of Elemento-Organic Chemistry, Key Laboratory of Advanced Energy Materials Chemistry (Ministry of Education), College of Chemistry, Nankai University, Tianjin 300071, PR China.
| |
Collapse
|
37
|
Lavecchia di Tocco F, Cannistraro S, Bizzarri AR. A PEG-based strategy to improve detection of clinical microRNA 155 by bio-Field Effect Transistor in high ionic strength environment. Talanta 2025; 292:127881. [PMID: 40073819 DOI: 10.1016/j.talanta.2025.127881] [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: 12/12/2024] [Revised: 02/14/2025] [Accepted: 03/01/2025] [Indexed: 03/14/2025]
Abstract
microRNAs are small oligonucleotides involved in post-transcriptional gene regulation whose alteration is found in several diseases, including cancer, and therefore their detection is crucial for diagnosis, prognosis, and treatment purposes. Field-Effect Transistor-based biosensors (bioFETs) represent a promising technology for the clinical detection of microRNAs. However, one of the main challenges associated with this technology is the Debye screening, becoming significant at the high ionic strengths required for effective hybridization. We aimed at detecting oncogenic microRNA-155 by using a bioFET system using as capture element a complementary RNA probe (antimiR-155) combined with the introduction of PEG molecules (20 kDa, PEG20), at an ionic strength of 300 mM. We optimized the co-immobilization ratio between antimiR-155 and PEG20 and assessed its impact on the interactions between the oligonucleotides. The kinetics can be well described by the Langmuir-Freundlich isotherm with an affinity constant within the range typical of nucleic acid interactions. The introduction of PEG20 significantly enhanced the detection sensitivity of miR-155 by reaching a level of less than 200 pM, together with excellent discrimination against other clinically relevant microRNAs. Our findings demonstrate that the incorporation of PEG20 constitutes an effective strategy to mitigate the Debye screening effects and facilitates bioFET-based clinical applications at physiological ionic strengths.
Collapse
Affiliation(s)
- Francesco Lavecchia di Tocco
- Biophysics and Nanoscience Centre, DEB, Università della Tuscia, Largo dell'Università, 01100, Viterbo, Italy; Department of Biomedical Sciences and Technologies, Università Roma Tre, Viale Guglielmo Marconi, 00144 Rome, Italy
| | - Salvatore Cannistraro
- Biophysics and Nanoscience Centre, DEB, Università della Tuscia, Largo dell'Università, 01100, Viterbo, Italy
| | - Anna Rita Bizzarri
- Biophysics and Nanoscience Centre, DEB, Università della Tuscia, Largo dell'Università, 01100, Viterbo, Italy.
| |
Collapse
|
38
|
Liu X, Li Y, Chen J, He S, Wang J, Zhang Z, Bai Z, Zhao Z, Wei H, Yu CY. A robust metal-organic framework-based nanozyme with multienzyme-like properties for synergistic tumor therapy via efficient and sustainable Nitric oxide generation. J Colloid Interface Sci 2025; 693:137603. [PMID: 40250114 DOI: 10.1016/j.jcis.2025.137603] [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: 01/21/2025] [Revised: 04/12/2025] [Accepted: 04/13/2025] [Indexed: 04/20/2025]
Abstract
Nitric oxide (NO)-based gas therapy represents an emerging strategy for cancer treatment, which, however, still suffers from insufficient intracellular NO production for compromised therapeutic efficiency due to limited endogenous hydrogen peroxide (H2O2) concentration and relatively slow NO generation rate. The use of metal-organic framework (MOF) with highly ordered porous structure and functional adjustability to design a multienzyme-like nanozyme provides a simple yet reliable approach for efficient and sustainable NO generation. Herein, a MOF-based nanozyme with multienzyme-like properties is constructed by depositing ultrasmall gold nanoparticles (Au NPs) and subsequently loading a NO donor, l-arginine (l-Arg) on an iron porphyrin integrated MOF, which enables efficient and sustainable NO generation for synergistic tumor therapy. Two notable merits of this design are (i) incorporation of Au NPs with glucose oxidase (GOx)-like property for effectively depleting intratumoral glucose, and simultaneously generating large amounts of H2O2, which is further utilized to initiate the effective production of NO and reactive oxygen species (ROS) by taking advantage of the peroxidase (POD)/NO synthase (NOS)-like activities of iron porphyrin, and (ii) deposition of ultrasmall Au NPs on the MOF for remarkably improving the structural stability of nanocomposites to circumvent the low catalytic efficiency associated with serious aggregation. This nanozyme achieves a high tumor inhibitory rate of 87.3 % with negligible systemic effects in a 4T1-tumor-bearing mice model by integrating NO, and ROS generation and starvation therapy with synergistic efficiency. Such an ingenious integration of multienzyme-like nanozyme and MOF paves a new way for NO-based gas therapy.
Collapse
Affiliation(s)
- Xinping Liu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China.
| | - Yi Li
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Jinjin Chen
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Suisui He
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Jun Wang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Zhijun Zhang
- Department of Chemistry, Key Laboratory of Surface & Interface Science of Polymer Materials of Zhejiang Province, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Zhiyuan Bai
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Zhike Zhao
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China
| | - Hua Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China.
| | - Cui-Yun Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang 421001, China; Affiliated Hospital of Hunan Academy of Chinese Medicine Hunan, Academy of Chinese Medicine, Changsha 410013, China.
| |
Collapse
|
39
|
Zhu L, Shen Z, Liu X, Tang R, Zhang Z, Zhao F, Wang J, Zhan W, Zhou L, Liang G, Wang R. Acid and phosphatase-triggered release and trapping of a prodrug on cancer cell enhance its chemotherapy. Biomaterials 2025; 320:123254. [PMID: 40088578 DOI: 10.1016/j.biomaterials.2025.123254] [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: 12/05/2024] [Revised: 02/22/2025] [Accepted: 03/10/2025] [Indexed: 03/17/2025]
Abstract
Using anticancer drug-encapsulated nanocarriers to actively target tumors is a promising chemotherapy strategy. Nevertheless, premature release of the drugs in tumor microenvironment (TME) or low tumor targeting efficiency of the nanocarriers significantly reduces its therapeutic efficiency. Herein, we propose a release-and-trapping strategy that significantly enhances the chemotherapeutic efficiency of an anticancer drug camptothecin. TME acid triggers the release of its prodrug from the nanocarrier and thereafter phosphatase instructs the prodrug to form hydrogel to trap the nanocarrier on cancer cell membrane. As trapped nanocarrier facilitates cell uptake of the prodrug and its intracellular carboxylesterase-mediated hydrolysis to release camptothecin. In vitro studies showed that the prodrug release from nanocarrier was maximized at pH 6.5. In tumor-bearing mice, our release-and-trapping strategy significantly prolonged the retention of the nanocarrier in tumor and significantly enhanced the anticancer efficacy of camptothecin. We propose that our release-and-trapping strategy be applied for more efficient cancer treatment in the future.
Collapse
Affiliation(s)
- Liangxi Zhu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 211189, China
| | - Zixiu Shen
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
| | - Xiaoyang Liu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 211189, China
| | - Runqun Tang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 211189, China
| | - Ziyi Zhang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 211189, China
| | - Furong Zhao
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 211189, China
| | - Jue Wang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, 211189, China
| | - Wenjun Zhan
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 211189, China
| | - Lei Zhou
- School of Science, China Pharmaceutical University, Nanjing, 210009, China
| | - Gaolin Liang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 211189, China.
| | - Rui Wang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 211189, China.
| |
Collapse
|
40
|
Fang J, Wang Y, Jiang Y, Li T, Qiu X. Advances in total antioxidant capacity detection based on nanozyme. Talanta 2025; 292:127941. [PMID: 40088770 DOI: 10.1016/j.talanta.2025.127941] [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: 01/24/2025] [Revised: 03/10/2025] [Accepted: 03/12/2025] [Indexed: 03/17/2025]
Abstract
Nanozymes, a class of nanomaterials mimicking natural enzymatic functions, have gained significant attention due to their exceptional biocatalytic properties and wide-ranging applications in biosensing. The total antioxidant capacity (TAC) can serve as a crucial parameter for assessing food quality, guiding dietary choices, and monitoring health conditions. In recent years, various nanomaterials with peroxidase (POD)-like and oxidase (OXD)-like activity have been widely used for TAC determination. This review discusses the enzyme-mimicking catalytic activities of nanozymes related to TAC determination, the construction principles of nanozyme-based TAC sensors and systematically classifies the application of nanozyme sensors in TAC determination. Furthermore, the potential opportunities and challenges in the development of nanozyme-based sensors are evaluated, aiming to provide valuable insights for researchers in related fields.
Collapse
Affiliation(s)
- Jiaoyuan Fang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, 471023, China
| | - Yun Wang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, 471023, China
| | - Yihan Jiang
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, 471023, China
| | - Tian Li
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, 471023, China.
| | - Xiangjun Qiu
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, 471023, China
| |
Collapse
|
41
|
Esmaeili H, Zhang Y, Ravi K, Neff K, Zhu W, Migrino RQ, Park JG, Nikkhah M. Development of an electroconductive Heart-on-a-chip model to investigate cellular and molecular response of human cardiac tissue to gold nanomaterials. Biomaterials 2025; 320:123275. [PMID: 40138961 DOI: 10.1016/j.biomaterials.2025.123275] [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: 10/23/2024] [Revised: 02/16/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025]
Abstract
To date, various strategies have been developed to construct biomimetic and functional in vitro cardiac tissue models utilizing human induced pluripotent stem cells (hiPSCs). Among these approaches, microfluidic-based Heart-on-a-chip (HOC) models are promising, as they enable the engineering of miniaturized, physiologically relevant in vitro cardiac tissues with precise control over cellular constituents and tissue architecture. Despite significant advancements, previously reported HOC models often lack the electroconductivity features of the native human myocardium. In this study, we developed a 3D electroconductive HOC (referred to as eHOC) model through the co-culture of isogenic hiPSC-derived cardiomyocytes (hiCMs) and cardiac fibroblasts (hiCFs), embedded within an electroconductive hydrogel scaffold in a microfluidic-based chip system. Functional and gene expression analyses demonstrated that, compared to non-conductive HOC, the eHOC model exhibited enhanced contractile functionality, improved calcium transients, and increased expression of structural and calcium handling genes. The eHOC model was further leveraged to investigate the underlying electroconduction-induced pathway(s) associated with cardiac tissue development through single-cell RNA sequencing (scRNA-seq). Notably, scRNA-seq analyses revealed a significant downregulation of a set of cardiac genes, associated with the fetal stage of heart development, as well as upregulation of sarcomere- and conduction-related genes within the eHOC model. Additionally, upregulation of the cardiac muscle contraction and motor protein pathways were observed in the eHOC model, consistent with enhanced contractile functionality of the engineered cardiac tissues. Comparison of scRNA-seq data from the 3D eHOC model with published datasets of adult human hearts demonstrated a similar expression pattern of fetal- and adult-like cardiac genes. Overall, this study provides a unique eHOC model with improved biomimcry and organotypic features, which could be potentially used for drug testing and discovery, as well as disease modeling applications.
Collapse
Affiliation(s)
- Hamid Esmaeili
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Yining Zhang
- Center for Personalized Diagnostics (CPD), Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA
| | - Kalpana Ravi
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Keagan Neff
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Wuqiang Zhu
- Department of Cardiovascular Medicine, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic, Scottsdale, AZ, 85259, USA
| | - Raymond Q Migrino
- Phoenix Veterans Affairs Health Care System, Phoenix, AZ, 85022, USA; University of Arizona College of Medicine, Phoenix, AZ, 85004, USA
| | - Jin G Park
- Center for Personalized Diagnostics (CPD), Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA
| | - Mehdi Nikkhah
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287, USA; Biodesign Virginia G. Piper Center for Personalized Diagnosis, Arizona State University, Tempe, AZ, 85287, USA.
| |
Collapse
|
42
|
Gan S, Wen X, Li L, Ao K, Qin J, Hao Y, Guo X. A cancer theranostic nanoplatform for second near-infrared fluorescence imaging-guided carbon monoxide-sensitized mild photothermal therapy with ICD induction. J Colloid Interface Sci 2025; 693:137652. [PMID: 40279848 DOI: 10.1016/j.jcis.2025.137652] [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: 12/24/2024] [Revised: 04/05/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
Mild-temperature photothermal therapy (mild PTT), utilizing photothermal agents to convert external light into mild heat (<45 °C), holds significant potential as a localized treatment modality to induce cellular thermal damage. This therapeutic strategy not only directly eliminates targeted cells but also induces immunogenic cell death (ICD), activating the immune response. However, the presence of heat shock proteins (HSPs) can significantly reduce the effectiveness of photothermal therapy. Therefore, it is crucial to inhibit HSP repair and minimize damage to surrounding normal cells in order to enhance the efficiency of low-temperature PTT. Additionally, carbon monoxide (CO) has been shown to suppress the upregulation of HSPs in cancer cells under heat treatment. Furthermore, the utilization of second near-infrared (NIR-II) fluorescence particles can improve the precision and suitability of PTT due to their increased penetration depth and novel imaging capabilities. In this study, we developed a NIR-light-activated CO release system using CO-loaded mesoporous organosilica nanoparticles (CO-MON) for enhancing the effectiveness of mild PTT by suppressing HSPs repair through selectively targeted CO delivery. Triiron dodecacarbonyl (Fe3(CO)12), as the source of CO was employed for encapsulation within the pores of the MON. These MON showed emission in the NIR-II range, while also displaying remarkable photostability and a high efficiency in photothermal conversion (34.7 %). Through intratumoral administration, the CO-MON platform demonstrated efficient tumor accumulation and localized photothermal efficacy in vivo. In vitro and in vivo studies demonstrated that this exceptional photothermal effect not only effectively eliminated tumor but also augmented tumor ICD.
Collapse
Affiliation(s)
- Shaoyan Gan
- Shenzhen Key Laboratory of Viral Oncology, Department of Science and Innovation, Shenzhen Hospital, Southern Medical University, Shenzhen, PR China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou, PR China
| | - Xiuli Wen
- Department of Ultrasound, South China Hospital, Medical School, Shenzhen University, Shenzhen, Guangdong 518116, PR China
| | - Li Li
- Shenzhen Key Laboratory of Viral Oncology, Department of Science and Innovation, Shenzhen Hospital, Southern Medical University, Shenzhen, PR China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou, PR China
| | - Keyi Ao
- Shenzhen Key Laboratory of Viral Oncology, Department of Science and Innovation, Shenzhen Hospital, Southern Medical University, Shenzhen, PR China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou, PR China
| | - Jiaqi Qin
- Shenzhen Key Laboratory of Viral Oncology, Department of Science and Innovation, Shenzhen Hospital, Southern Medical University, Shenzhen, PR China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou, PR China
| | - Yi Hao
- Department of Ultrasound, South China Hospital, Medical School, Shenzhen University, Shenzhen, Guangdong 518116, PR China.
| | - Xia Guo
- Shenzhen Key Laboratory of Viral Oncology, Department of Science and Innovation, Shenzhen Hospital, Southern Medical University, Shenzhen, PR China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou, PR China.
| |
Collapse
|
43
|
Wang J, Zou Z, Shi Z, Chen H, Lu J, Ren C, Zhu P, He J, Yang R. Structure modification of P2-Na 0.67Ni 0.15Fe 0.2Mn 0.65O 2 cathode by incorporation of cerium for high-performance sodium-ion batteries. J Colloid Interface Sci 2025; 693:137619. [PMID: 40253861 DOI: 10.1016/j.jcis.2025.137619] [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: 01/23/2025] [Revised: 04/12/2025] [Accepted: 04/15/2025] [Indexed: 04/22/2025]
Abstract
The P2-type transition metal oxides hold promise for the cathode of sodium-ion batteries (SIBs). However, the prevailing irreversible phase transition and Jahn-Teller effects of this kind of metal oxides lead to low capacity and poor cycling stability of SIBs. To address these challenges, in this study, the rare-earth metal element cerium (Ce) is successfully introduced into P2-Na0.67Ni0.15Fe0.2Mn0.65O2 (NFM), achieving a synergistic bulk doping and surface modification for NFM. Upon incorporating larger Ce3+ into the lattice, the interlayer spacing of P2- NFM is increased, facilitating the diffusion of Na+. The substitution of Ce for Mn sites suppresses the Jahn-Teller effect caused by Mn3+ in P2-NFM. The concurrently formed CeO2 on the surface effectively inhibits the corrosion and degradation of P2-NFM by the electrolyte. Benefitting from the dual-modification, P2-Na0.67Ni0.15Fe0.2Mn0.61Ce0.04O2 (NFMC-0.04) exhibits an increased discharge capacity of 171.6 mAh g-1 at 0.1C as compared to that of NFM (142.9 mAh g-1), and a higher capacity retention of 57.40 % after 200 cycles than that of NFM (37.97 %). Impressively, it can deliver a capacity retention of 58.41 % after 250 cycles at 1C, while the discharge capacity of un-modified NFM is close to zero. In-situ XRD analysis reveals that the successful doping of Ce into the bulk phase of NFM suppresses the structural distortion of NFM from P2 to OP4 phase. Density Functional Theory calculations disclose that the substitution of Mn sites by Ce elements is energetically most favorable in NFM. Ce doping not only improves the electronic conductivity of NFM by enhancing the degree of electron localization, but also suppresses the Jahn-Teller effect by modulating the electronic structure of Mn. This study provides a new approach for engineering of layered transition metal oxides toward the high-performance cathode of SIBs.
Collapse
Affiliation(s)
- Ji Wang
- College of Energy, Soochow Institute for Energy and Materials Innovations, Soochow University, Suzhou 215006, China
| | - Zhixiong Zou
- College of Energy, Soochow Institute for Energy and Materials Innovations, Soochow University, Suzhou 215006, China
| | - Zhangjing Shi
- College of Energy, Soochow Institute for Energy and Materials Innovations, Soochow University, Suzhou 215006, China; Institute of Nuclear and New Energy Technology, Tsinghua University, Beijing 100084, China
| | - Hong Chen
- College of Energy, Soochow Institute for Energy and Materials Innovations, Soochow University, Suzhou 215006, China
| | - Jiahao Lu
- College of Energy, Soochow Institute for Energy and Materials Innovations, Soochow University, Suzhou 215006, China
| | - Chaojie Ren
- College of Energy, Soochow Institute for Energy and Materials Innovations, Soochow University, Suzhou 215006, China
| | - Peijun Zhu
- College of Energy, Soochow Institute for Energy and Materials Innovations, Soochow University, Suzhou 215006, China
| | - Jie He
- College of Energy, Soochow Institute for Energy and Materials Innovations, Soochow University, Suzhou 215006, China
| | - Ruizhi Yang
- College of Energy, Soochow Institute for Energy and Materials Innovations, Soochow University, Suzhou 215006, China.
| |
Collapse
|
44
|
Zhao Z, Zhou J, Li X, Zhang T, Tian Z, Sun T, Jiang C. Manganese-based virus-mimicking nanomedicine with triple immunomodulatory functions inhibits breast cancer brain metastasis. Biomaterials 2025; 320:123262. [PMID: 40138963 DOI: 10.1016/j.biomaterials.2025.123262] [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: 12/09/2024] [Revised: 02/23/2025] [Accepted: 03/16/2025] [Indexed: 03/29/2025]
Abstract
Hindered by the challenges of blood-brain barrier (BBB) hindrance, tumor heterogeneity and immunosuppressive microenvironment, patients with breast cancer brain metastasis have yet to benefit from current clinical treatments, experiencing instead a decline in quality of life due to radiochemotherapy. While virus-mimicking nanosystems (VMN) mimicking viral infection processes show promise in treating peripheral tumors, the inability to modulate the immunosuppressive microenvironment limits the efficacy against brain metastasis. Accordingly, a VMN-based triple immunomodulatory strategy is initially proposed, aiming to activate innate and adaptive immune responses and reverse the immunosuppressive microenvironment. Here, manganese-based virus-mimicking nanomedicine (Vir-HD@HM) with intratumoral drug enrichment is engineered. Vir-HD@HM can induce the immune response through the activation of cGAS-STING by mimicking the in vivo infection process of herpesviruses. Meanwhile, DNAzyme mimicking the genome can rescue the epigenetic silencing of PTEN with the assistance of Mn2+, thus ameliorating the immunosuppressive metastatic microenvironment and achieving synergistic sensitizing therapeutic efficacy. In vivo experiments substantiate the efficacy of Vir-HD@HM in recruiting NK cells and CD8+ T cells to metastatic foci, inhibiting Treg cells infiltration, and prolonging murine survival without adjunctive radiochemotherapy. This study demonstrates that Vir-HD@HM with triple immunomodulation offers an encouraging therapeutic option for patients with brain metastasis.
Collapse
Affiliation(s)
- Zhenhao Zhao
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
| | - Jingyi Zhou
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
| | - Xuwen Li
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
| | - Tongyu Zhang
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
| | - Zonghua Tian
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
| | - Tao Sun
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China
| | - Chen Jiang
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai, 201203, China; Department of Digestive Diseases, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China.
| |
Collapse
|
45
|
Liu Y, Yin S, Hu L, Ma R, Fan M, Feng M, Fang R, Xiao Z, Zhang J, Gan Y, Huang H, He X, Xia X, Jin Y, Tao X, Zhang W, Xia Y. Silicified-montmorillonite assisted in-situ construction of LiF-rich phase for enhanced interfacial stability in solid-state lithium batteries. J Colloid Interface Sci 2025; 693:137585. [PMID: 40245828 DOI: 10.1016/j.jcis.2025.137585] [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: 02/11/2025] [Revised: 03/25/2025] [Accepted: 04/11/2025] [Indexed: 04/19/2025]
Abstract
The interfacial failure of solid polymer electrolytes (SPEs) with Li anode, particularly those containing succinonitrile (SN) types, has significantly hindered the practical development of solid-state lithium-metal batteries. Herein, we introduce silicified montmorillonite (SiO2-MMT) into polyethylene oxide (PEO)/SN-based SPEs to facilitate the in-situ formation of a LiF-rich phase, thereby significantly enhancing the electrolyte/Li anode interface stability. Specifically, the SiO2-MMT strongly anchors the SN molecules, preventing their migration to the Li anode side. Meanwhile, the TFSI- reacts competitively with Li to generate a dense LiF phase, further blocking the SN chemical corrosion of Li metal. Furthermore, the SiO2-MMT greatly accelerates the Li+ transportability and enhances the thermal stability and mechanical strength of the composite electrolyte. Consequently, the composite electrolyte has a high ionic conductivity of 1.2 × 10-4 S cm-1 at 30 °C, which almost twice as higher as the counterpart. As for the solid-state battery, the LiFePO4||Li cell demonstrates remarkable cycling stability, achieving a high discharge capacity of 141.7 mA h g-1 with a remarkable capacity retention of 90.2 % after 350 cycles at 0.5C. Additionally, the high electrochemical window (4.6 V) ensures compatibility with high-voltage cathode materials. The NCM83||Li cell demonstrates a satisfactory initial discharge capacity and excellent capacity retention of 179.2 mA h g-1 and 81.8 % at 0.2 C after 100 cycles. This work offers a new insight for achieving a highly stable electrolyte/Li anode interface in SN-containing SPEs, facilitating the practical application of solid-state lithium metal batteries with enhanced safety and lifespan.
Collapse
Affiliation(s)
- Yaning Liu
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, China; Moganshan Research Institute at Deqing County Zhejiang University of Technology, Huzhou 313000, China
| | - Shengwei Yin
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Liuyi Hu
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Ruojian Ma
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Min Fan
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, China.
| | - Meiqing Feng
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Ruyi Fang
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Zhen Xiao
- Key Laboratory of Rare Earth Optoelectronic Materials and Devices of Zhejiang Province, Institute of Optoelectronic Materials and Devices, China Jiliang University, Hangzhou 310018, China.
| | - Jun Zhang
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yongping Gan
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Hui Huang
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Xinping He
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Xinhui Xia
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yanxian Jin
- School of Pharmaceutical and Chemical Engineering, Taizhou University, Jiaojiang 318000, China
| | - Xinyong Tao
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Wenkui Zhang
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yang Xia
- College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou 310014, China.
| |
Collapse
|
46
|
Ren Y, Dong Y, Li Z, Xu K, Xu J, Li X, Zhang M, Xu C, Yang M, Lee M, Meng X, Wang J. Kidney-targeting DNA tetrahedral molecular cage synergistically inhibits acute kidney injury by clearing ROS and activating HO-1. Biomaterials 2025; 320:123237. [PMID: 40049024 DOI: 10.1016/j.biomaterials.2025.123237] [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: 11/12/2024] [Revised: 02/04/2025] [Accepted: 03/02/2025] [Indexed: 04/06/2025]
Abstract
Acute kidney injury (AKI) is a major cause of mortality in hospitalized patients, yet effective therapeutic interventions remain underdeveloped. To address this critical need, we have employed tetrahedral framework nucleic acid (tFNA) as a carrier to self-assemble a complex incorporating G-quadruplex and hemin (G4/Hemin). This novel formulation exhibits uniform particle size, targeted delivery, and significant therapeutic efficacy for AKI. In a chemotherapy-induced AKI model, G4/Hemin-tFNA preferentially accumulated in the renal tubules, significantly mitigating drug-induced renal tubular injury. In healthy mice, G4/Hemin-tFNA was rapidly cleared from circulation due to efficient renal filtration. Safety evaluations conducted over a continuous 30-day period indicated minimal side effects associated with G4/Hemin-tFNA administration. Mechanistic studies elucidated three primary molecular mechanisms through which G4/Hemin-tFNA exerts its therapeutic effects in AKI: 1) Enhanced Renal Targeting. G4/Hemin-tFNA facilitates effective renal targeting and protection during blood circulation, leading to significant accumulation of drug within the kidneys. 2) Reactive Oxygen Species (ROS) Clearance. The complex exhibits peroxidase-like activity, enabling the rapid clearance of ROS at the site of AKI lesions, thereby inhibiting the oxidative stress progression. 3) Activation of heme oxygenase-1 (HO-1). G4/Hemin-tFNA selectively activates HO-1, enhancing the concentration of anti-inflammatory factors at inflamed sites and promoting an anti-inflammatory microenvironment. Collectively, these findings demonstrate that G4/Hemin-tFNA is a safe and effective therapeutic agent for AKI. By activating HO-1 and clearing ROS, G4/Hemin-tFNA inhibits disease progression, offering a promising approach for the development of future AKI therapies.
Collapse
Affiliation(s)
- Yu Ren
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, 230032, PR China
| | - Yuhang Dong
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, 230032, PR China
| | - Zhi Li
- Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, CA, 90095, United States
| | - Keying Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, 230032, PR China
| | - Jiafeng Xu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, 230032, PR China
| | - Xiangyu Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, 230032, PR China
| | - Mengmeng Zhang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, 230032, PR China
| | - Changlu Xu
- Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, CA, 90095, United States
| | - Min Yang
- The Second Department of Critical. Care Medicine, The Second Affiliated Hospital of Anhui- Medical University, Anhui, Hefei, 230601, PR China
| | - Min Lee
- Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, CA, 90095, United States; Department of Bioengineering, University of California, Los Angeles, CA, 90095, United States.
| | - Xiaoming Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, Institute for Liver Diseases of Anhui Medical University, School of Pharmacy, Anhui Medical University, Hefei, 230032, PR China.
| | - Jie Wang
- The Second Department of Critical. Care Medicine, The Second Affiliated Hospital of Anhui- Medical University, Anhui, Hefei, 230601, PR China.
| |
Collapse
|
47
|
Wei D, Xu A, Chen X, Ma J, Huang F, Wu H, Liu Y, Ye R, Zhu M, Xu J. Single-atom catalysts confined in shell layer achieved by a modified top-down strategy for efficient CO 2 reduction. J Colloid Interface Sci 2025; 693:137566. [PMID: 40233689 DOI: 10.1016/j.jcis.2025.137566] [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: 11/27/2024] [Revised: 03/24/2025] [Accepted: 04/09/2025] [Indexed: 04/17/2025]
Abstract
High-temperature pyrolysis is a primary method for synthesizing single-atom catalysts (SACs). However, this method accelerates the migration of metal atoms within the solid support, leading to low atom utilization. Herein, we report a novel top-down synthesis strategy wherein surface-sintered nickel sulfide (NiS2) nanoparticles (NPs) are in situ atomized into single atoms, achieving confinement of the single-atom catalyst within the shell layer and synthesizing a high-performance single-atom catalyst. Systematic investigations indicate that driven by strong interactions between metal atoms and the support, the NiS2 NPs on the surface of the support atomize into single Ni atoms, which are predominantly distributed on the support surface, thus enhancing the accessibility of the active sites. Furthermore, theoretical calculations indicate that introducing S atoms into the second coordination shell around Ni atoms significantly reduces the activation energy of the CO2 reduction reaction, thereby enhancing the catalytic performance of the single-atom catalyst. In the flow cell, the Ni single-atom catalyst achieving nearly 100% Faradaic efficiency for CO (FECO) over a wide potential range of -0.5 to -1.3 V versus reversible hydrogen electrode (vs. RHE). At -1.6 V vs. RHE, the partial current density for CO reaches a maximum of 709 mA cm-2 (turnover frequency of 28.67 s-1) with a FECO of 95.9%.
Collapse
Affiliation(s)
- Dong Wei
- Guangxi Key Laboratory of Petrochemical Resource Processing and Process Intensification Technology, School of Chemistry and Chemical Engineering, Guangxi University, Nanning 530004, China
| | - Aihao Xu
- Guangxi Key Laboratory of Petrochemical Resource Processing and Process Intensification Technology, School of Chemistry and Chemical Engineering, Guangxi University, Nanning 530004, China; Guangxi Academy of Medical Sciences, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Xiangyu Chen
- Guangxi Key Laboratory of Petrochemical Resource Processing and Process Intensification Technology, School of Chemistry and Chemical Engineering, Guangxi University, Nanning 530004, China
| | - Junjie Ma
- Guangxi Key Laboratory of Petrochemical Resource Processing and Process Intensification Technology, School of Chemistry and Chemical Engineering, Guangxi University, Nanning 530004, China
| | - Fang Huang
- Guangxi Key Laboratory of Petrochemical Resource Processing and Process Intensification Technology, School of Chemistry and Chemical Engineering, Guangxi University, Nanning 530004, China
| | - Haoran Wu
- State Key Laboratory of Green Chemical Engineering and Industrial Catalysis, School of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, China
| | - Yong Liu
- Department of Chemistry, State Key Laboratory of Marine Pollution, City University of Hong Kong, 999077, Hong Kong, China
| | - Ruquan Ye
- Department of Chemistry, State Key Laboratory of Marine Pollution, City University of Hong Kong, 999077, Hong Kong, China
| | - Minghui Zhu
- State Key Laboratory of Green Chemical Engineering and Industrial Catalysis, School of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, China.
| | - Jing Xu
- Guangxi Key Laboratory of Petrochemical Resource Processing and Process Intensification Technology, School of Chemistry and Chemical Engineering, Guangxi University, Nanning 530004, China; State Key Laboratory of Green Chemical Engineering and Industrial Catalysis, School of Chemical Engineering, East China University of Science and Technology, Shanghai 200237, China.
| |
Collapse
|
48
|
Li W, Ma S, Zhang N, Yang Y, Fan S, Cong L, Xie H. Molecular fluorination towards deep eutectic amide-based electrolyte for stable high voltage lithium-metal batteries. J Colloid Interface Sci 2025; 693:137597. [PMID: 40253859 DOI: 10.1016/j.jcis.2025.137597] [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: 01/19/2025] [Revised: 03/29/2025] [Accepted: 04/12/2025] [Indexed: 04/22/2025]
Abstract
Currently, non-flammable deep eutectic electrolytes (DEEs), typically based on N-methylacetamide (NMAC), have been deemed as high-quality electrolytes employed in lithium-metal batteries (LMBs). However, the unstable interphase chemistry derived from high reactivity of amide groups towards aggressive electrodes (Li and NCM cathode) and tight Li+-amide coordination still exists as the unavoidable "sore point" for DEEs innovation as yet. Herein, inspired by fluorinated solvent strategy, N-Methyl-2,2,2-trifluoroacetamide (FNMAC), is proposed to design the FNMAC-based DEE (F-DEE-1:n, n = 2 ∼ 8) solely containing lithium bis(trifluoromethanesulphonyl)imide (LiTFSI) salt. Introducing electron-withdrawing -CF3 group is conducive to realizing excellent oxidation resistance as well as stable interphase chemistry, which impairs Li+-amide strong coordination bringing forth anion-rich solvation sheath and robust solid electrolyte interface (SEI) with high inorganic content, together with promoting the fast desolvation of Li+. Consequently, the F-DEE-1:4 endows NCM622||Li cells with excellent rate capability and outstanding long lifespan along with high capacity retention of ∼91.3 % after cycling 420 times, much superior to those using NMAC-based DEE (N-DEE-1:4). This work is instructive for high-quality DEEs innovation and emphasizes the close correlation between Li+ coordination environment and stable interphase chemistry within LMBs.
Collapse
Affiliation(s)
- Wenbo Li
- China National & Local United Engineering Laboratory for Power Battery Department of Chemistry, Northeast Normal University, Changchun 130024, China
| | - Shunchao Ma
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry Chinese Academy of Sciences, Changchun 130022, China
| | - Nan Zhang
- China National & Local United Engineering Laboratory for Power Battery Department of Chemistry, Northeast Normal University, Changchun 130024, China
| | - Yutong Yang
- China National & Local United Engineering Laboratory for Power Battery Department of Chemistry, Northeast Normal University, Changchun 130024, China
| | - Siqi Fan
- China National & Local United Engineering Laboratory for Power Battery Department of Chemistry, Northeast Normal University, Changchun 130024, China
| | - Lina Cong
- China National & Local United Engineering Laboratory for Power Battery Department of Chemistry, Northeast Normal University, Changchun 130024, China.
| | - Haiming Xie
- China National & Local United Engineering Laboratory for Power Battery Department of Chemistry, Northeast Normal University, Changchun 130024, China.
| |
Collapse
|
49
|
Qiu N, Xu C, Zhang Z, Wang R, Wei X, Xie Y, Wang S, Lu D, Wang K, Xu S, Shen C, Su R, Cen B, Liu Y, Shen Y, Xu X. Autologous tumoral esterase-driven therapeutic polymers sequentially orchestrated antigen-induction, STING activation and anti-angiogenesis for systemic cancer immune therapy. Biomaterials 2025; 320:123260. [PMID: 40138966 DOI: 10.1016/j.biomaterials.2025.123260] [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: 10/30/2024] [Revised: 02/23/2025] [Accepted: 03/15/2025] [Indexed: 03/29/2025]
Abstract
Effective cancer immune therapy requires the orchestration of antigen induction, presentation and T-cell activation, further enhanced by anti-angiogenesis treatment; therefore, multiple therapeutics are generally used for such combination therapy. Herein, we report esterase-hydrolysable cationic polymers, N-[3-((4-acetoxy benzyl) oxy)-3-oxopropyl]-N-methyl-quaternized PEI (ERP) and poly{N-[2-(acryloyl-oxy) ethyl]-N-[p-acetyloxyphenyl]-N,N-dimethylammonium chloride} (PQDMA), capable of simultaneously inducing tumor cell immunogenic cell death (ICD) to release antigens, activating the cGAS-STING pathways of tumor macrophages and dendritic cells, and releasing antiangiogenic agent p-hydroxybenzyl alcohol (HBA). Thus, intratumoral injection of ERP or PQDMA systemically boosted the anti-cancer immunities and inhibited tumor angiogenesis in mouse hepatocellular carcinoma and melanoma bilateral tumor models, leading to more effective tumor growth inhibition of both treated and abscopal untreated tumors than ICD alone induced by mitoxantrone and control cationic polymers. Further study using gene knockout mice and transcriptome sequencing analysis confirmed the involvement of cGAS-STING and type I IFN signaling pathways. This work demonstrates ERP and PQDMA as the first examples of inherent therapeutic polymers, accomplishing systemic tumor inhibition without combining other therapeutic agents.
Collapse
Affiliation(s)
- Nasha Qiu
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, China.
| | - Chang Xu
- Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Zhen Zhang
- Key Laboratory of Smart Biomaterials of Zhejiang Province and Key Laboratory of Biomass Chemical Engineering of the Ministry of Education of China, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Rui Wang
- Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Xuyong Wei
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, China
| | - Yangla Xie
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, China
| | - Shuai Wang
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, China
| | - Di Lu
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou, 310059, China
| | - Kai Wang
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou, 310059, China
| | - Shengjun Xu
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, China
| | - Chenchen Shen
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, China
| | - Renyi Su
- Zhejiang University School of Medicine, Hangzhou, 310058, Zhejiang, China
| | - Beini Cen
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, China
| | - Yanpeng Liu
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, 310006, China
| | - Youqing Shen
- Key Laboratory of Smart Biomaterials of Zhejiang Province and Key Laboratory of Biomass Chemical Engineering of the Ministry of Education of China, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310058, China.
| | - Xiao Xu
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou, 310059, China; Institute of Translational Medicine, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
50
|
Yu M, Qiu Y, Wang Y, Wang Y, Ma Z, Wang Y, Liu S. Engineering multifunctional high-entropy oxide nanozymes for robust marine antifouling. J Colloid Interface Sci 2025; 693:137604. [PMID: 40245833 DOI: 10.1016/j.jcis.2025.137604] [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: 01/15/2025] [Revised: 04/09/2025] [Accepted: 04/13/2025] [Indexed: 04/19/2025]
Abstract
High-performance interfacial antifouling coatings are crucial for sustainable marine resource utilization. This work reports a novel high-entropy oxide (HEO) nanozyme, CrMnFeNiCuOX nanoparticles, where the synergistic interplay of polymetallic cations and defect engineering yield remarkable multi-enzyme mimetic activity combined with a photothermal conversion efficiency of 40.06%. Under simulated solar irradiation, the HEO nanozyme exhibited complete (100%) bactericidal activity against both Escherichia coli and Staphylococcus aureus, and effectively suppresses biofilm formation in a simulated marine environment. Mechanistic investigations demonstrated that the HEO nanozyme exhibits a tailored electronic structure and adsorption properties, enabling disruption of bacterial membrane integrity, perturbation of intracellular redox homeostasis, and suppression of quorum sensing signaling. This multifaceted approach offers a promising strategy for developing durable and environmentally friendly antifouling coatings for diverse marine applications.
Collapse
Affiliation(s)
- Miao Yu
- Faculty of Life Science and Medicine, School of Medicine and Health, Key Laboratory of Microsystems and Microstructures Manufacturing, Harbin Institute of Technology, Harbin 150001, China
| | - Yunfeng Qiu
- Faculty of Life Science and Medicine, School of Medicine and Health, Key Laboratory of Microsystems and Microstructures Manufacturing, Harbin Institute of Technology, Harbin 150001, China.
| | - Yuhang Wang
- Faculty of Life Science and Medicine, School of Medicine and Health, Key Laboratory of Microsystems and Microstructures Manufacturing, Harbin Institute of Technology, Harbin 150001, China
| | - Yanxia Wang
- Faculty of Life Science and Medicine, School of Medicine and Health, Key Laboratory of Microsystems and Microstructures Manufacturing, Harbin Institute of Technology, Harbin 150001, China
| | - Zhuo Ma
- Faculty of Life Science and Medicine, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150001, China
| | - Youshan Wang
- National Key Laboratory of Science and Technology on Advanced Composites in Special Environments, Harbin Institute of Technology, Harbin 150001, China.
| | - Shaoqin Liu
- Faculty of Life Science and Medicine, School of Medicine and Health, Key Laboratory of Microsystems and Microstructures Manufacturing, Harbin Institute of Technology, Harbin 150001, China.
| |
Collapse
|